1
|
Flores-Sierra JDJ, Muciño-Arellano MDR, Romo-Morales GDC, Sánchez-Palafox JE, Correa-Navarro VA, Colín-Castelán D, Pérez-Vázquez V, Rangel-Salazar R, Rivera-Bustamante R, de la Rocha C, Rodríguez-Ríos D, Trejo-Saavedra DL, Molina-Torres J, Ramírez-Chávez E, García-Rojas NS, Winkler R, Lund G, Zaina S. The DNA methyltransferase inhibitor decitabine blunts the response to a high-animal fat and protein diet in mice. J Lipid Res 2024; 65:100586. [PMID: 38942113 PMCID: PMC11325794 DOI: 10.1016/j.jlr.2024.100586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 06/30/2024] Open
Abstract
Increasing evidence hints that DNA hypermethylation may mediate the pathogenic response to cardiovascular risk factors. Here, we tested a corollary of that hypothesis, that is, that the DNA methyltransferase inhibitor decitabine (Dec) ameliorates the metabolic profile of mice fed a moderately high-animal fat and protein diet (HAFPD), a proxy of cardiovascular risk-associated Western-type diet. HAFPD-fed mice were exposed to Dec or vehicle for eight weeks (8W set, 4-32/group). To assess any memory of past exposure to Dec, we surveyed a second mice set treated as 8W but HAFPD-fed for further eight weeks without any Dec (16W set, 4-20/group). In 8W, Dec markedly reduced HAFPD-induced body weight gain in females, but marginally in males. Characterization of females revealed that Dec augmented skeletal muscle lipid content, while decreasing liver fat content and increasing plasma nonesterified fatty acids, adipose insulin resistance, and-although marginally-whole blood acylcarnitines, compared to HAFPD alone. Skeletal muscle mitochondrial DNA copy number was higher in 8W mice exposed to HAFPD and Dec, or in 16W mice fed HAFPD only, relative to 8W mice fed HAFPD only, but Dec induced a transcriptional profile indicative of ameliorated mitochondrial function. Memory of past Dec exposure was tissue-specific and sensitive to both duration of exposure to HAFPD and age. In conclusion, Dec redirected HAFPD-induced lipid accumulation toward the skeletal muscle, likely due to augmented mitochondrial functionality and increased lipid demand. As caveat, Dec induced adipose insulin resistance. Our findings may help identifying strategies for prevention and treatment of lipid dysmetabolism.
Collapse
Affiliation(s)
- José de Jesús Flores-Sierra
- Division of Health Sciences, Department of Medical Sciences, Leon Campus, University of Guanajuato, Leon, Mexico; Tecnológico Nacional de México/ITS de Purísima del Rincón, Purísima del Rincón, Guanajuato, Mexico
| | | | | | | | | | - Dannia Colín-Castelán
- Division of Health Sciences, Department of Medical Sciences, Leon Campus, University of Guanajuato, Leon, Mexico
| | - Victoriano Pérez-Vázquez
- Division of Health Sciences, Department of Medical Sciences, Leon Campus, University of Guanajuato, Leon, Mexico
| | - Rubén Rangel-Salazar
- Division of Health Sciences, Department of Medical Sciences, Leon Campus, University of Guanajuato, Leon, Mexico
| | | | - Carmen de la Rocha
- Department of Genetic Engineering, CINVESTAV Irapuato Unit, Irapuato, Mexico
| | | | | | - Jorge Molina-Torres
- Department of Biotechnology and Biochemistry, CINVESTAV Irapuato Unit, Irapuato, Mexico
| | | | | | | | - Gertrud Lund
- Department of Genetic Engineering, CINVESTAV Irapuato Unit, Irapuato, Mexico.
| | - Silvio Zaina
- Division of Health Sciences, Department of Medical Sciences, Leon Campus, University of Guanajuato, Leon, Mexico.
| |
Collapse
|
2
|
Beauchemin R, Merindol N, Fantino E, Lavoie P, Nouemssi SB, Meddeb-Mouelhi F, Desgagné-Penix I. Successful reversal of transgene silencing in Chlamydomonas reinhardtii. Biotechnol J 2024; 19:e2300232. [PMID: 37975165 DOI: 10.1002/biot.202300232] [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: 05/23/2023] [Revised: 09/13/2023] [Accepted: 11/14/2023] [Indexed: 11/19/2023]
Abstract
Chlamydomonas reinhardtii has been successfully engineered to produce compounds of interest following transgene integration and heterologous protein expression. The advantages of this model include the availability of validated tools for bioengineering, its photosynthetic ability, and its potential use as biofuel. Despite this, breakthroughs have been hindered by its ability to silence transgene expression through epigenetic changes. Histone deacetylases (HDAC) are main players in gene expression. We hypothesized that transgene silencing can be reverted with chemical treatments using HDAC inhibitors. To analyze this, we transformed C. reinhardtii, integrating into its genome the mVenus reporter gene under the HSP70-rbcs2 promoter. From 384 transformed clones, 88 (22.9%) displayed mVenus positive (mVenus+ ) cells upon flow-cytometry analysis. Five clones with different fluorescence intensities were selected. The number of integrated copies was measured by qPCR. Transgene expression levels were followed over the growth cycle and upon SAHA treatment, using a microplate reader, flow cytometry, RT-qPCR, and western blot analysis. First, we observed that expression varies with the cell cycle, reaching a maximum level just before the stationary phase in all clones. Second, we uncovered that supplementation with HDAC inhibitors of the hydroxamate family, such as vorinostat (suberoylanilide-hydroxamic-acid, SAHA) at the initiation of culture increases the frequency (% of mVenus+ cells) and the level of transgene expression per cell over the whole growth cycle, through histone deacetylase inhibition. Thus, we propose a new tool to successfully trigger the expression of heterologous proteins in the green algae C. reinhardtii, overcoming its main obstacle as an expression platform.
Collapse
Affiliation(s)
- Rémy Beauchemin
- Department of Chemistry, Biochemistry and Physics, Université du Québec à Trois-Rivières, Trois-Rivières, Québec, Canada
| | - Natacha Merindol
- Department of Chemistry, Biochemistry and Physics, Université du Québec à Trois-Rivières, Trois-Rivières, Québec, Canada
| | - Elisa Fantino
- Department of Chemistry, Biochemistry and Physics, Université du Québec à Trois-Rivières, Trois-Rivières, Québec, Canada
| | - Pamela Lavoie
- Department of Chemistry, Biochemistry and Physics, Université du Québec à Trois-Rivières, Trois-Rivières, Québec, Canada
| | - Serge Basile Nouemssi
- Department of Chemistry, Biochemistry and Physics, Université du Québec à Trois-Rivières, Trois-Rivières, Québec, Canada
| | - Fatma Meddeb-Mouelhi
- Department of Chemistry, Biochemistry and Physics, Université du Québec à Trois-Rivières, Trois-Rivières, Québec, Canada
| | - Isabel Desgagné-Penix
- Department of Chemistry, Biochemistry and Physics, Université du Québec à Trois-Rivières, Trois-Rivières, Québec, Canada
- Plant Biology Research Group, Trois-Rivières, Québec, Canada
| |
Collapse
|
3
|
Linowiecka K, Slominski AT, Reiter RJ, Böhm M, Steinbrink K, Paus R, Kleszczyński K. Melatonin: A Potential Regulator of DNA Methylation. Antioxidants (Basel) 2023; 12:1155. [PMID: 37371885 PMCID: PMC10295183 DOI: 10.3390/antiox12061155] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/17/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
The pineal gland-derived indoleamine hormone, melatonin, regulates multiple cellular processes, ranging from chronobiology, proliferation, apoptosis, and oxidative damage to pigmentation, immune regulation, and mitochondrial metabolism. While melatonin is best known as a master regulator of the circadian rhythm, previous studies also have revealed connections between circadian cycle disruption and genomic instability, including epigenetic changes in the pattern of DNA methylation. For example, melatonin secretion is associated with differential circadian gene methylation in night shift workers and the regulation of genomic methylation during embryonic development, and there is accumulating evidence that melatonin can modify DNA methylation. Since the latter one impacts cancer initiation, and also, non-malignant diseases development, and that targeting DNA methylation has become a novel intervention target in clinical therapy, this review discusses the potential role of melatonin as an under-investigated candidate epigenetic regulator, namely by modulating DNA methylation via changes in mRNA and the protein expression of DNA methyltransferases (DNMTs) and ten-eleven translocation (TET) proteins. Furthermore, since melatonin may impact changes in the DNA methylation pattern, the authors of the review suggest its possible use in combination therapy with epigenetic drugs as a new anticancer strategy.
Collapse
Affiliation(s)
- Kinga Linowiecka
- Department of Human Biology, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, Lwowska 1, 87-100 Toruń, Poland
- Dr. Phillip Frost Department of Dermatology & Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL 33125, USA
| | - Andrzej T. Slominski
- Department of Dermatology, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Pathology and Laboratory Medicine Service, VA Medical Center, Birmingham, AL 35294, USA
| | - Russel J. Reiter
- Department of Cell Systems and Anatomy, UT Health, Long School of Medicine, San Antonio, TX 78229, USA
| | - Markus Böhm
- Department of Dermatology, University of Münster, Von-Esmarch-Str. 58, 48149 Münster, Germany
| | - Kerstin Steinbrink
- Department of Dermatology, University of Münster, Von-Esmarch-Str. 58, 48149 Münster, Germany
| | - Ralf Paus
- Dr. Phillip Frost Department of Dermatology & Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL 33125, USA
| | - Konrad Kleszczyński
- Department of Dermatology, University of Münster, Von-Esmarch-Str. 58, 48149 Münster, Germany
| |
Collapse
|
4
|
Szczepanek J, Skorupa M, Jarkiewicz-Tretyn J, Cybulski C, Tretyn A. Harnessing Epigenetics for Breast Cancer Therapy: The Role of DNA Methylation, Histone Modifications, and MicroRNA. Int J Mol Sci 2023; 24:ijms24087235. [PMID: 37108398 PMCID: PMC10138995 DOI: 10.3390/ijms24087235] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 03/24/2023] [Accepted: 04/10/2023] [Indexed: 04/29/2023] Open
Abstract
Breast cancer exhibits various epigenetic abnormalities that regulate gene expression and contribute to tumor characteristics. Epigenetic alterations play a significant role in cancer development and progression, and epigenetic-targeting drugs such as DNA methyltransferase inhibitors, histone-modifying enzymes, and mRNA regulators (such as miRNA mimics and antagomiRs) can reverse these alterations. Therefore, these epigenetic-targeting drugs are promising candidates for cancer treatment. However, there is currently no effective epi-drug monotherapy for breast cancer. Combining epigenetic drugs with conventional therapies has yielded positive outcomes and may be a promising strategy for breast cancer therapy. DNA methyltransferase inhibitors, such as azacitidine, and histone deacetylase inhibitors, such as vorinostat, have been used in combination with chemotherapy to treat breast cancer. miRNA regulators, such as miRNA mimics and antagomiRs, can alter the expression of specific genes involved in cancer development. miRNA mimics, such as miR-34, have been used to inhibit tumor growth, while antagomiRs, such as anti-miR-10b, have been used to inhibit metastasis. The development of epi-drugs that target specific epigenetic changes may lead to more effective monotherapy options in the future.
Collapse
Affiliation(s)
- Joanna Szczepanek
- Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University, 87-100 Torun, Poland
| | - Monika Skorupa
- Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University, 87-100 Torun, Poland
- Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, 87-100 Torun, Poland
| | | | - Cezary Cybulski
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University, 70-204 Szczecin, Poland
| | - Andrzej Tretyn
- Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University, 87-100 Torun, Poland
- Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, 87-100 Torun, Poland
| |
Collapse
|
5
|
Kaluscha S, Domcke S, Wirbelauer C, Stadler MB, Durdu S, Burger L, Schübeler D. Evidence that direct inhibition of transcription factor binding is the prevailing mode of gene and repeat repression by DNA methylation. Nat Genet 2022; 54:1895-1906. [PMID: 36471082 PMCID: PMC9729108 DOI: 10.1038/s41588-022-01241-6] [Citation(s) in RCA: 64] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 10/24/2022] [Indexed: 12/12/2022]
Abstract
Cytosine methylation efficiently silences CpG-rich regulatory regions of genes and repeats in mammalian genomes. To what extent this entails direct inhibition of transcription factor (TF) binding versus indirect inhibition via recruitment of methyl-CpG-binding domain (MBD) proteins is unclear. Here we show that combinatorial genetic deletions of all four proteins with functional MBDs in mouse embryonic stem cells, derived neurons or a human cell line do not reactivate genes or repeats with methylated promoters. These do, however, become activated by methylation-restricted TFs if DNA methylation is removed. We identify several causal TFs in neurons, including ONECUT1, which is methylation sensitive only at a motif variant. Rampantly upregulated retrotransposons in methylation-free neurons feature a CRE motif, which activates them in the absence of DNA methylation via methylation-sensitive binding of CREB1. Our study reveals methylation-sensitive TFs in vivo and argues that direct inhibition, rather than indirect repression by the tested MBD proteins, is the prevailing mechanism of methylation-mediated repression at regulatory regions and repeats.
Collapse
Affiliation(s)
- Sebastian Kaluscha
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
- University of Basel, Faculty of Sciences, Basel, Switzerland
| | - Silvia Domcke
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | | | - Michael B Stadler
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
- Swiss Institute of Bioinformatics, Basel, Switzerland
- University of Basel, Faculty of Sciences, Basel, Switzerland
| | - Sevi Durdu
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Lukas Burger
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
- Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Dirk Schübeler
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland.
- University of Basel, Faculty of Sciences, Basel, Switzerland.
| |
Collapse
|
6
|
Ulahannan N, Cutler R, Doña-Termine R, Simões-Pires CA, Wijetunga NA, Croken MM, Johnston AD, Kong Y, Maqbool SB, Suzuki M, Greally JM. Genomic insights into host and parasite interactions during intracellular infection by Toxoplasma gondii. PLoS One 2022; 17:e0275226. [PMID: 36178892 PMCID: PMC9524707 DOI: 10.1371/journal.pone.0275226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 08/02/2022] [Indexed: 11/18/2022] Open
Abstract
To gain insights into the molecular interactions of an intracellular pathogen and its host cell, we studied the gene expression and chromatin states of human fibroblasts infected with the Apicomplexan parasite Toxoplasma gondii. We show a striking activation of host cell genes that regulate a number of cellular processes, some of which are protective of the host cell, others likely to be advantageous to the pathogen. The simultaneous capture of host and parasite genomic information allowed us to gain insights into the regulation of the T. gondii genome. We show how chromatin accessibility and transcriptional profiling together permit novel annotation of the parasite's genome, including more accurate mapping of known genes and the identification of new genes and cis-regulatory elements. Motif analysis reveals not only the known T. gondii AP2 transcription factor-binding site but also a previously-undiscovered candidate TATA box-containing motif at one-quarter of promoters. By inferring the transcription factor and upstream cell signaling responses involved in the host cell, we can use genomic information to gain insights into T. gondii's perturbation of host cell physiology. Our resulting model builds on previously-described human host cell signalling responses to T. gondii infection, linked to induction of specific transcription factors, some of which appear to be solely protective of the host cell, others of which appear to be co-opted by the pathogen to enhance its own survival.
Collapse
Affiliation(s)
- Netha Ulahannan
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, United States of America
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Ronald Cutler
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Reanna Doña-Termine
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Claudia A. Simões-Pires
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - N. Ari Wijetunga
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Matthew McKnight Croken
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Andrew D. Johnston
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Yu Kong
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Shahina B. Maqbool
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Masako Suzuki
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - John M. Greally
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, United States of America
| |
Collapse
|
7
|
Boideau F, Richard G, Coriton O, Huteau V, Belser C, Deniot G, Eber F, Falentin C, Ferreira de Carvalho J, Gilet M, Lodé-Taburel M, Maillet L, Morice J, Trotoux G, Aury JM, Chèvre AM, Rousseau-Gueutin M. Epigenomic and structural events preclude recombination in Brassica napus. THE NEW PHYTOLOGIST 2022; 234:545-559. [PMID: 35092024 DOI: 10.1111/nph.18004] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 01/15/2022] [Indexed: 06/14/2023]
Abstract
Meiotic recombination is a major evolutionary process generating genetic diversity at each generation in sexual organisms. However, this process is highly regulated, with the majority of crossovers lying in the distal chromosomal regions that harbor low DNA methylation levels. Even in these regions, some islands without recombination remain, for which we investigated the underlying causes. Genetic maps were established in two Brassica napus hybrids to detect the presence of such large nonrecombinant islands. The role played by DNA methylation and structural variations in this local absence of recombination was determined by performing bisulfite sequencing and whole genome comparisons. Inferred structural variations were validated using either optical mapping or oligo fluorescence in situ hybridization. Hypermethylated or inverted regions between Brassica genomes were associated with the absence of recombination. Pairwise comparisons of nine B. napus genome assemblies revealed that such inversions occur frequently and may contain key agronomic genes such as resistance to biotic stresses. We conclude that such islands without recombination can have different origins, such as DNA methylation or structural variations in B. napus. It is thus essential to take into account these features in breeding programs as they may hamper the efficient combination of favorable alleles in elite varieties.
Collapse
Affiliation(s)
- Franz Boideau
- IGEPP, INRAE, Institut Agro, Univ Rennes, Le Rheu, 35653, France
| | - Gautier Richard
- IGEPP, INRAE, Institut Agro, Univ Rennes, Le Rheu, 35653, France
| | - Olivier Coriton
- IGEPP, INRAE, Institut Agro, Univ Rennes, Le Rheu, 35653, France
| | - Virginie Huteau
- IGEPP, INRAE, Institut Agro, Univ Rennes, Le Rheu, 35653, France
| | - Caroline Belser
- Génomique Métabolique, Genoscope, Institut François Jacob, CEA, CNRS, Université Evry, Université Paris-Saclay, 2 Rue Gaston Crémieux, Evry, 91057, France
| | - Gwenaelle Deniot
- IGEPP, INRAE, Institut Agro, Univ Rennes, Le Rheu, 35653, France
| | - Frédérique Eber
- IGEPP, INRAE, Institut Agro, Univ Rennes, Le Rheu, 35653, France
| | - Cyril Falentin
- IGEPP, INRAE, Institut Agro, Univ Rennes, Le Rheu, 35653, France
| | | | - Marie Gilet
- IGEPP, INRAE, Institut Agro, Univ Rennes, Le Rheu, 35653, France
| | | | - Loeiz Maillet
- IGEPP, INRAE, Institut Agro, Univ Rennes, Le Rheu, 35653, France
| | - Jérôme Morice
- IGEPP, INRAE, Institut Agro, Univ Rennes, Le Rheu, 35653, France
| | - Gwenn Trotoux
- IGEPP, INRAE, Institut Agro, Univ Rennes, Le Rheu, 35653, France
| | - Jean-Marc Aury
- Génomique Métabolique, Genoscope, Institut François Jacob, CEA, CNRS, Université Evry, Université Paris-Saclay, 2 Rue Gaston Crémieux, Evry, 91057, France
| | | | | |
Collapse
|
8
|
Planques A, Kerner P, Ferry L, Grunau C, Gazave E, Vervoort M. DNA methylation atlas and machinery in the developing and regenerating annelid Platynereis dumerilii. BMC Biol 2021; 19:148. [PMID: 34340707 PMCID: PMC8330077 DOI: 10.1186/s12915-021-01074-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 06/16/2021] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Methylation of cytosines in DNA (5mC methylation) is a major epigenetic modification that modulates gene expression and constitutes the basis for mechanisms regulating multiple aspects of embryonic development and cell reprogramming in vertebrates. In mammals, 5mC methylation of promoter regions is linked to transcriptional repression. Transcription regulation by 5mC methylation notably involves the nucleosome remodeling and deacetylase complex (NuRD complex) which bridges DNA methylation and histone modifications. However, less is known about regulatory mechanisms involving 5mC methylation and their function in non-vertebrate animals. In this paper, we study 5mC methylation in the marine annelid worm Platynereis dumerilii, an emerging evolutionary and developmental biology model capable of regenerating the posterior part of its body post-amputation. RESULTS Using in silico and experimental approaches, we show that P. dumerilii displays a high level of DNA methylation comparable to that of mammalian somatic cells. 5mC methylation in P. dumerilii is dynamic along the life cycle of the animal and markedly decreases at the transition between larval to post-larval stages. We identify a full repertoire of mainly single-copy genes encoding the machinery associated with 5mC methylation or members of the NuRD complex in P. dumerilii and show that this repertoire is close to the one inferred for the last common ancestor of bilaterians. These genes are dynamically expressed during P. dumerilii development and regeneration. Treatment with the DNA hypomethylating agent Decitabine impairs P. dumerilii larval development and regeneration and has long-term effects on post-regenerative growth. CONCLUSIONS Our data reveal high levels of 5mC methylation in the annelid P. dumerilii, highlighting that this feature is not specific to vertebrates in the bilaterian clade. Analysis of DNA methylation levels and machinery gene expression during development and regeneration, as well as the use of a chemical inhibitor of DNA methylation, suggest an involvement of 5mC methylation in P. dumerilii development and regeneration. We also present data indicating that P. dumerilii constitutes a promising model to study biological roles and mechanisms of DNA methylation in non-vertebrate bilaterians and to provide new knowledge about evolution of the functions of this key epigenetic modification in bilaterian animals.
Collapse
Affiliation(s)
- Anabelle Planques
- Université de Paris, CNRS, Institut Jacques Monod, F-75006, Paris, France
| | - Pierre Kerner
- Université de Paris, CNRS, Institut Jacques Monod, F-75006, Paris, France
| | - Laure Ferry
- Université de Paris, CNRS, Epigenetics and Cell Fate, F-75006, Paris, France
| | - Christoph Grunau
- IHPE, Univ Montpellier, CNRS, IFREMER, Univ Perpignan Via Domitia, F-66860, Perpignan, France
| | - Eve Gazave
- Université de Paris, CNRS, Institut Jacques Monod, F-75006, Paris, France.
| | - Michel Vervoort
- Université de Paris, CNRS, Institut Jacques Monod, F-75006, Paris, France.
| |
Collapse
|
9
|
Ali SR, Ahmad W, Naeem N, Salim A, Khan I. Small molecule 2'-deoxycytidine differentiates human umbilical cord-derived MSCs into cardiac progenitors in vitro and their in vivo xeno-transplantation improves cardiac function. Mol Cell Biochem 2020; 470:99-113. [PMID: 32415417 DOI: 10.1007/s11010-020-03750-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 05/08/2020] [Indexed: 12/11/2022]
Abstract
Small molecules are widely used to induce stem cell differentiation. 2'-deoxycytidine (2-DC) belongs to the cytidine family. It stimulates the expression of cardiac-specific genes and proteins, and directs mesenchymal stem cells towards cardiomyogenic differentiation. We aim to investigate the role of 2-DC-treated human umbilical cord mesenchymal stem cells (UC-MSCs) into myogenic lineage and explore their application in regeneration of infarcted myocardium. UC-MSCs were treated with 5, 10, 20, and 40 µM 2-DC following optimization by cytotoxicity analysis. Rat model of myocardial infarction (MI) was induced by ligating left anterior descending coronary artery. Normal, and 2-DC treated UC-MSCs were transplanted in the left ventricular wall immediately after ligation. Echocardiographic measurements were performed to assess cardiac function. Tissue architecture of the myocardium was examined by histological analysis to determine fate of the transplanted cells. MSCs were successfully isolated from human umbilical cord tissue. 2-DC treatment did not produce any significant cytotoxic effect in UC-MSCs at all concentrations. qPCR analysis of treated UC-MSCs showed induction of myogenic differentiation, which is more pronounced at 20 μM concentration. Fluorescently labeled 2-DC-treated UC-MSCs showed significant (**P < 0.01) homing in the infarcted myocardium as compared to normal UC-MSCs. Hearts transplanted with 2-DC-treated UC-MSCs significantly (***P < 0.001) improved the cardiac systolic and diastolic functions and pumping ability as compared to normal UC-MSCs and MI groups. Fibrotic area and left ventricular wall thickness were significantly improved (***P < 0.001) in 2-DC-treated group as compared to normal UC-MSCs. Immunohistochemical staining showed co-localization of fluorescently labeled cells and patches of differentiated myocytes which were stained for cardiac proteins in the infarct zone implying that the treated UC-MSCs regenerated cardiomyocytes. We report for the first time that 2-DC induces cardiac differentiation in UC-MSCs. Transplanted cells differentiated into functional cardiomyocytes and significantly improved cardiac performance. These pre-differentiated cardiac progenitors showed better survival, homing, and distribution in the infarcted zone. 2-DC treated cells not only improved cardiac function, but also restored tissue homeostasis, suggesting a better therapeutic option for the regeneration of cardiac tissue in the clinical setup.
Collapse
Affiliation(s)
- Syeda Roohina Ali
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Waqas Ahmad
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Nadia Naeem
- Dow University of Health Sciences, Ojha Campus, Gulzar-e-Hijri, Suparco Road, KDA Scheme-33, Karachi, Pakistan
| | - Asmat Salim
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Irfan Khan
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan.
| |
Collapse
|
10
|
Shin Y, Kim M, Won J, Kim J, Oh SB, Lee JH, Park K. Epigenetic Modification of CFTR in Head and Neck Cancer. J Clin Med 2020; 9:jcm9030734. [PMID: 32182826 PMCID: PMC7141320 DOI: 10.3390/jcm9030734] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 02/29/2020] [Accepted: 03/06/2020] [Indexed: 12/24/2022] Open
Abstract
Cystic fibrosis transmembrane conductance regulator (CFTR), a cyclic AMP (cAMP)-regulated chloride channel, is critical for secretion and absorption across diverse epithelia. Mutations or absence of CFTR result in pathogeneses, including cancer. While CFTR has been proposed as a tumor suppressing gene in tumors of the intestine, lung, and breast cancers, its effects in head and neck cancer (HNC) have yet to be investigated. This study aimed to define expression patterns and epigenetic modifications of CFTR in HNC. CFTR was expressed in normal but not in HNC cells and tissues. Treatment with 5-aza-2'-deoxycytidine (5-Aza-CdR) was associated with rescued expression of CFTR, whose function was confirmed by patch clamp technique. Further experiments demonstrated that CFTR CpG islands were hypermethylated in cancer cells and tissues and hypomethylated in normal cells and tissue. Our results suggest that CFTR epigenetic modifications are critical in both down-regulation and up-regulation of CFTR expression in HNC and normal cells respectively. We then investigated the impact of CFTR on expressions and functions of cancer-related genes. CFTR silencing was closely associated with changes to other cancer-related genes, suppressing apoptosis while enhancing proliferation, cell motility, and invasion in HNC. Our findings demonstrate that hypermethylation of CFTR CpG islands and CFTR deficiency is closely related to HNC.
Collapse
Affiliation(s)
- Yonghwan Shin
- Department of Physiology, School of Dentistry, Seoul National University and Dental Research Institute, Seoul 110-749, Korea; (Y.S.); (M.K.); (J.K.)
| | - Minkyoung Kim
- Department of Physiology, School of Dentistry, Seoul National University and Dental Research Institute, Seoul 110-749, Korea; (Y.S.); (M.K.); (J.K.)
| | - Jonghwa Won
- Department of Neurobiology and Physiology, School of Dentistry, Seoul National University and Dental Research Institute, Seoul 110-749, Korea; (J.W.); (S.B.O.)
| | - Junchul Kim
- Department of Physiology, School of Dentistry, Seoul National University and Dental Research Institute, Seoul 110-749, Korea; (Y.S.); (M.K.); (J.K.)
| | - Seog Bae Oh
- Department of Neurobiology and Physiology, School of Dentistry, Seoul National University and Dental Research Institute, Seoul 110-749, Korea; (J.W.); (S.B.O.)
| | - Jong-Ho Lee
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Seoul National University, Seoul 110-749, Korea;
| | - Kyungpyo Park
- Department of Physiology, School of Dentistry, Seoul National University and Dental Research Institute, Seoul 110-749, Korea; (Y.S.); (M.K.); (J.K.)
- Correspondence: ; Tel.: +82-02-740-8658
| |
Collapse
|
11
|
Gao Y, Li L, Zhang W, Ru X, Hou L, Wang Y. Clinical efficacy of cytarabine + aclarubicin + recombinant human granulocyte colony-stimulating factor regimen combined with decitabine for adult acute myeloid leukemia. ALL LIFE 2020. [DOI: 10.1080/26895293.2020.1776773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Affiliation(s)
- Ying Gao
- Department of Hematology, Shaanxi Provincial People's Hospital, Xi'an, People’s Republic of China
| | - Lan Li
- Department of Hematology, Shaanxi Provincial People's Hospital, Xi'an, People’s Republic of China
| | - Weihua Zhang
- Department of Hematology, Shaanxi Provincial People's Hospital, Xi'an, People’s Republic of China
| | - Xingli Ru
- Department of Hematology, Shaanxi Provincial People's Hospital, Xi'an, People’s Republic of China
| | - Limin Hou
- Department of Hematology, Shaanxi Provincial People's Hospital, Xi'an, People’s Republic of China
| | - Yi Wang
- Department of Hematology, Shaanxi Provincial People's Hospital, Xi'an, People’s Republic of China
| |
Collapse
|
12
|
Cook N, Parker DJ, Tauber E, Pannebakker BA, Shuker DM. Validating the Demethylating Effects of 5-aza-2'-deoxycytidine in Insects Requires a Whole-Genome Approach. Am Nat 2019; 194:432-438. [PMID: 31553206 DOI: 10.1086/704248] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
We previously demonstrated that treatment with the demethylating agent 5-aza-2'-deoxycytidine (5-aza-dC) alters the offspring sex ratios produced by females of the parasitoid wasp Nasonia vitripennis. Females allocate offspring sex ratio in line with local mate competition theory, producing more or less female-biased sex ratios as the number of other females laying eggs on a patch varies, thereby reducing competition among their sons for mates. Interestingly, treatment with 5-aza-dC did not ablate the facultative sex allocation response. Instead, sex ratios became less female biased, a shift in the direction of the optimum sex ratio for paternally inherited alleles according to genomic conflict theory. This was the first (albeit indirect) experimental evidence for genomic conflict over sex allocation. In their comment, Ellers and colleagues assayed the effects of 5-aza-dC on DNA methylation in 10 Nasonia genes, finding no evidence of demethylation in these 10 genes, from which they conclude that 5-aza-dC has no demethylating capability in N. vitripennis. Quantifying the efficacy of 5-aza-dC in terms of demethylation is indeed crucial to in-depth interpretation of studies using 5-aza-dC to link phenotypes to epigenetic regulation. Here we outline the mode of action of 5-aza-dC and demonstrate that determining the efficacy of 5-aza-dC in insect systems requires a whole-genome approach.
Collapse
|
13
|
Teixeira SR, Abreu CM, Parkes L, Davies J, Yao S, Sawhney MA, Margarit L, Gonzalez D, Pinto IM, Francis LW, Conlan RS. Direct monitoring of breast and endometrial cancer cell epigenetic response to DNA methyltransferase and histone deacetylase inhibitors. Biosens Bioelectron 2019; 141:111386. [PMID: 31220725 DOI: 10.1016/j.bios.2019.111386] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 05/23/2019] [Accepted: 05/30/2019] [Indexed: 12/19/2022]
Abstract
DNA methylation and histone deacetylation are key epigenetic processes involved in normal cellular function and tumorigenesis. Therapeutic strategies based on DNA methyltransferase (DNMT) and histone deacetylase (HDAC) inhibitors are currently in use and under development for the treatment of cancers. Genome-wide DNA methylation profiling has been proposed for use in disease diagnosis, and histone modification profiling for disease stratification will follow suit. However, whether epigenome sequencing technologies will be feasible for rapid clinic diagnosis and patient treatment monitoring remains to be seen, and alternative detection technologies will almost certainly be needed. Here we used electrochemical impedance spectroscopy (EIS) employing a graphene-based screen-printed electrode system to directly measure global DNA methylation and histone H3 acetylation to compare non-cancer and breast cancer cell lines. We demonstrated that whilst global methylation was not useful as a differential marker in the cellular systems tested, histone H3 acetylation was effective at higher chromatin levels. Using breast and endometrial cancer cell models, EIS was then used to monitor cellular responses to the DNMT and HDAC inhibitors 5-Aza-2'-deoxycytidine and suberoylanilide hydroxamic acid in vitro, and proved very effective at detecting global cellular responses to either treatment, indicating that this approach could be useful in following treatment response to epigenetic drugs. Moreover, this work reports the first combined analysis of two epigenetic markers using a unified graphene-based biosensor platform, demonstrating the potential for multiplex analysis of both methylation and acetylation on the same sample.
Collapse
Affiliation(s)
- S R Teixeira
- College of Engineering, Swansea University, Bay Campus, Swansea, SA1 8QQ, UK; Centre for NanoHealth, Swansea University, Singleton Park, Swansea, SA2 8PP, UK
| | - C M Abreu
- International Iberian Nanotechnology Laboratory (INL), Portugal
| | - L Parkes
- Swansea University Medical School, Singleton Park, Swansea, SA2 8PP, UK; Centre for NanoHealth, Swansea University, Singleton Park, Swansea, SA2 8PP, UK
| | - J Davies
- Swansea University Medical School, Singleton Park, Swansea, SA2 8PP, UK; Centre for NanoHealth, Swansea University, Singleton Park, Swansea, SA2 8PP, UK
| | - S Yao
- Swansea University Medical School, Singleton Park, Swansea, SA2 8PP, UK; Centre for NanoHealth, Swansea University, Singleton Park, Swansea, SA2 8PP, UK
| | - M A Sawhney
- Swansea University Medical School, Singleton Park, Swansea, SA2 8PP, UK; Centre for NanoHealth, Swansea University, Singleton Park, Swansea, SA2 8PP, UK
| | - L Margarit
- Swansea University Medical School, Singleton Park, Swansea, SA2 8PP, UK; Abertawe Bro Morganwg University Health Board, Princess of Wales Hospital Bridgend, UK
| | - D Gonzalez
- Swansea University Medical School, Singleton Park, Swansea, SA2 8PP, UK; Centre for NanoHealth, Swansea University, Singleton Park, Swansea, SA2 8PP, UK
| | - I Mendes Pinto
- International Iberian Nanotechnology Laboratory (INL), Portugal
| | - L W Francis
- Swansea University Medical School, Singleton Park, Swansea, SA2 8PP, UK; Centre for NanoHealth, Swansea University, Singleton Park, Swansea, SA2 8PP, UK
| | - R S Conlan
- Swansea University Medical School, Singleton Park, Swansea, SA2 8PP, UK; Centre for NanoHealth, Swansea University, Singleton Park, Swansea, SA2 8PP, UK.
| |
Collapse
|
14
|
Sato H, Wu B, Delahaye F, Singer RH, Greally JM. Retargeting of macroH2A following mitosis to cytogenetic-scale heterochromatic domains. J Cell Biol 2019; 218:1810-1823. [PMID: 31110057 PMCID: PMC6548134 DOI: 10.1083/jcb.201811109] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 03/20/2019] [Accepted: 04/19/2019] [Indexed: 12/12/2022] Open
Abstract
How macroH2A, a histone variant involved in silencing gene expression, is inherited from parent to daughter cells is unclear. Using a combination of imaging, biochemical, and genomic approaches, Sato et al. describe how newly synthesized macroH2A is incorporated predominantly in the G1 phase of human mitosis, targeting heterochromatic regions. The heritability of chromatin states through cell division is a potential contributor to the epigenetic maintenance of cellular memory of prior states. The macroH2A histone variant has properties of a regulator of epigenetic cell memory, including roles controlling gene silencing and cell differentiation. Its mechanisms of regional genomic targeting and maintenance through cell division are unknown. Here, we combined in vivo imaging with biochemical and genomic approaches to show that human macroH2A is incorporated into chromatin in the G1 phase of the cell cycle following DNA replication. The newly incorporated macroH2A retargets the same large heterochromatic domains where macroH2A was already enriched in the previous cell cycle. It remains heterotypic, targeting individual nucleosomes that do not already contain a macroH2A molecule. The pattern observed resembles that of a new deposition of centromeric histone variants during the cell cycle, indicating mechanistic similarities for macrodomain-scale regulation of epigenetic properties of the cell.
Collapse
Affiliation(s)
- Hanae Sato
- Center for Epigenomics and Department of Genetics, Albert Einstein College of Medicine, Bronx, NY.,Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY
| | - Bin Wu
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY.,Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY
| | - Fabien Delahaye
- Center for Epigenomics and Department of Genetics, Albert Einstein College of Medicine, Bronx, NY.,Department of Obstetrics and Gynecology and Women's Health, Albert Einstein College of Medicine, Bronx, NY
| | - Robert H Singer
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY .,Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY.,Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY.,Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA
| | - John M Greally
- Center for Epigenomics and Department of Genetics, Albert Einstein College of Medicine, Bronx, NY
| |
Collapse
|
15
|
Giri AK, Aittokallio T. DNMT Inhibitors Increase Methylation in the Cancer Genome. Front Pharmacol 2019; 10:385. [PMID: 31068808 PMCID: PMC6491738 DOI: 10.3389/fphar.2019.00385] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 03/27/2019] [Indexed: 01/16/2023] Open
Abstract
DNA methyltransferase inhibitors (DNMTi) decitabine and azacytidine are approved therapies for myelodysplastic syndrome and acute myeloid leukemia, and their combinations with other anticancer agents are being tested as therapeutic options for multiple solid cancers such as colon, ovarian, and lung cancer. However, the current therapeutic challenges of DNMTis include development of resistance, severe side effects and no or partial treatment responses, as observed in more than half of the patients. Therefore, there is a critical need to better understand the mechanisms of action of these drugs. In order to discover molecular targets of DNMTi therapy, we identified 638 novel CpGs with an increased methylation in response to decitabine treatment in HCT116 cell lines and validated the findings in multiple cancer types (e.g., bladder, ovarian, breast, and lymphoma) cell lines, bone marrow mononuclear cells from primary leukemia patients, as well as peripheral blood mononuclear cells and ascites from platinum resistance epithelial ovarian cancer patients. Azacytidine treatment also increased methylation of these CpGs in colon, ovarian, breast, and lymphoma cancer cell lines. Methylation at 166 identified CpGs strongly correlated (|r|≥ 0.80) with corresponding gene expression in HCT116 cell line. Differences in methylation at some of the identified CpGs and expression changes of the corresponding genes was observed in TCGA colon cancer tissue as compared to adjacent healthy tissue. Our analysis revealed that hypermethylated CpGs are involved in cancer cell proliferation and apoptosis by P53 and olfactory receptor pathways, hence influencing DNMTi responses. In conclusion, we showed hypermethylation of CpGs as a novel mechanism of action for DNMTi agents and identified 638 hypermethylated molecular targets (CpGs) common to decitabine and azacytidine therapy. These novel results suggest that hypermethylation of CpGs should be considered when predicting the DNMTi responses and side effects in cancer patients.
Collapse
Affiliation(s)
- Anil K Giri
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Tero Aittokallio
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland.,Helsinki Institute for Information Technology, Department of Computer Science, Aalto University, Espoo, Finland.,Department of Mathematics and Statistics, University of Turku, Turku, Finland
| |
Collapse
|
16
|
Zhou J, Yi Z, Fu Q. Dynamic decreased expression and hypermethylation of secreted frizzled-related protein 1 and 4 over the course of pulmonary fibrosis in mice. Life Sci 2019; 218:241-252. [PMID: 30586565 DOI: 10.1016/j.lfs.2018.12.041] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 12/18/2018] [Accepted: 12/22/2018] [Indexed: 01/08/2023]
Abstract
Aberrantly activated Wnt signaling pathway and dysregulation of extracellular antagonists of Wnt signaling have been revealed in pulmonary fibrosis. In this study we evaluated the expression of secreted frizzled-related proteins (SFRPs) and their aberrant promoter methylation to investigate the involvement of epigenetic regulation in pulmonary fibrosis. The pulmonary fibrosis induced by intratracheal injection of bleomycin (BLM) into mice was adopted. The transcription and relative protein expression of SFRPs were detected at Day 7 (D7), D14, and D21. DNA methylation analysis was performed by methylation-specific polymerase chain reaction (MSP). A DNA methyltransferase (DNMT) inhibitor (5-aza-2'-deoxycytidine; 5-aza) was used for demethylation and the relative β-catenin expression levels were measured to assess overactivity of the canonical Wnt signaling pathway. The transcription and protein expression of SFRP1 significantly decreased at D14 and D21, whereas the transcription and protein expression of SFRP4 significantly decreased at D7 and stayed downregulated until D21. The significantly hypermethylated promoters of SFRP1 and SFRP4 resulted in impaired transcription and decreased expression during pulmonary fibrosis in mice. Besides, reactivation of SFRP1 and SFRP4 by 5-aza reduced β-catenin mRNA and protein expression in vivo and in vitro. Animal experiments confirmed that 5-aza could significantly alleviate bleomycin-induced pulmonary fibrosis in mice. Thus, changes of promoter hypermethylation might downregulate SFRP1 and SFRP4 at different stages of pulmonary fibrosis, and the finding supports the usefulness of DNMT inhibitors, which might effectively reverse activation of β-catenin and reduce pulmonary fibrosis in mice. These data provide a possible new direction in the research on pulmonary fibrosis treatments.
Collapse
Affiliation(s)
- Junfei Zhou
- Department of Rheumatology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, PR China
| | - Zheng Yi
- Department of Rheumatology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, PR China.
| | - Qiang Fu
- Department of Rheumatology, The First Affiliated Hospital of University of South China, HengYang 421001, PR China
| |
Collapse
|
17
|
Combination treatment of acute myeloid leukemia cells with DNMT and HDAC inhibitors: predominant synergistic gene downregulation associated with gene body demethylation. Leukemia 2018; 33:945-956. [PMID: 30470836 DOI: 10.1038/s41375-018-0293-8] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 06/21/2018] [Accepted: 09/06/2018] [Indexed: 12/20/2022]
Abstract
DNA methyltransferase inhibitors (DNMTi) approved for older AML patients are clinically tested in combination with histone deacetylase inhibitors (HDACi). The mechanism of action of these drugs is still under debate. In colon cancer cells, 5-aza-2'-deoxycytidine (DAC) can downregulate oncogenes and metabolic genes by reversing gene body DNA methylation, thus implicating gene body methylation as a novel drug target. We asked whether DAC-induced gene body demethylation in AML cells is also associated with gene repression, and whether the latter is enhanced by HDACi.Transcriptome analyses revealed that a combined treatment with DAC and the HDACi panobinostat or valproic acid affected significantly more transcripts than the sum of the genes regulated by either treatment alone, demonstrating a quantitative synergistic effect on genome-wide expression in U937 cells. This effect was particularly striking for downregulated genes. Integrative methylome and transcriptome analyses showed that a massive downregulation of genes, including oncogenes (e.g., MYC) and epigenetic modifiers (e.g., KDM2B, SUV39H1) often overexpressed in cancer, was associated predominantly with gene body DNA demethylation and changes in acH3K9/27. These findings have implications for the mechanism of action of combined epigenetic treatments, and for a better understanding of responses in trials where this approach is clinically tested.
Collapse
|
18
|
Marofi F, Vahedi G, Solali S, Alivand M, Salarinasab S, Zadi Heydarabad M, Farshdousti Hagh M. Gene expression of TWIST1 and ZBTB16 is regulated by methylation modifications during the osteoblastic differentiation of mesenchymal stem cells. J Cell Physiol 2018; 234:6230-6243. [PMID: 30246336 DOI: 10.1002/jcp.27352] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Accepted: 08/17/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND Osteoblastic differentiation of mesenchymal stem cells (MSCs) is the principal stage during the restoration and regeneration of bone tissue. Epigenetic modifications such as DNA methylation play a key role in the differentiation process of stem cells. In this study, the methylation status of the promoter region of ZBTB16 and Twist1 genes and their role in controlling osteoblastic differentiation in MSCs was investigated during the osteoblastic differentiation of MSCs. METHODS The MSCs were cultured under standard conditions and differentiated into the osteoblasts. We had three treatment groups including 5-azacytidine (methylation inhibitor), metformin (Twist-inhibitor), and procaine (Wnt/β-catenin inhibitor) and a non-treated group (control). Methylation level of DNA in the promoter regions was monitored by methylation specific-quantitative polymerase chain reaction (PCR). Also, the mRNA levels of key genes in osteoblastic differentiation were measured using real-time PCR. RESULTS ZBTB16 gene expression was upregulated, and promoter methylation was decreased. For Twist1 messenger RNA (mRNA) level decreased and promoter methylation increased during osteoblastic differentiation of MSCs. 5-Azacytidine caused a significant reduction in methylation and increased the mRNA expression of ZBTB16 and Twist1. Metformin repressed the Twist1 expression, and therefore osteoblastic differentiation was increased. On the opposite side, procaine could block the WNT/β-catenin signaling pathway, as a consequence the gene expression of key genes involved in osteoblastic differentiation was declined. CONCLUSION We found that methylation of DNA in the promoter region of ZBTB16 and Twist1 genes might be one of the main mechanisms that controlling the gene expression during osteoblastic differentiation of MSCs. Also, we could find an association between regulation of Twist1 and ZBTB16 genes and osteoblastic differentiation in MSCs by showing the relation between their expression and some key genes involved in osteoblastic differentiation. In addition, we found a connection between the Twist1 expression level and osteoblastic differentiation by using a Twist-inhibitor (metformin).
Collapse
Affiliation(s)
- Faroogh Marofi
- Department of Hematology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ghasem Vahedi
- Department of Immunology, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Saeed Solali
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammadreza Alivand
- Department of Medical genetic, Faculty of Medicine, Tabriz University of medical sciences, Tabriz, Iran
| | - Sadegh Salarinasab
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical science, Tabriz, Iran
| | - Milad Zadi Heydarabad
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | |
Collapse
|
19
|
Ribas L, Vanezis K, Imués MA, Piferrer F. Treatment with a DNA methyltransferase inhibitor feminizes zebrafish and induces long-term expression changes in the gonads. Epigenetics Chromatin 2017; 10:59. [PMID: 29216900 PMCID: PMC5721477 DOI: 10.1186/s13072-017-0168-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 11/30/2017] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The role of epigenetic modifications such as DNA methylation during vertebrate sexual development is far from being clear. Using the zebrafish model, we tested the effects of one of the most common DNA methyltransferase (dnmt) inhibitor, 5-aza-2'-deoxycytidine (5-aza-dC), which is approved for the treatment of acute myeloid leukaemia and is under active investigation for the treatment of solid tumours. Several dose-response experiments were carried out during two periods, including not only the very first days of development (0-6 days post-fertilization, dpf), as done in previous studies, but also, and as a novelty, the period of gonadal development (10-30 dpf). RESULTS Early treatment with 5-aza-dC altered embryonic development, delayed hatching and increased teratology and mortality, as expected. The most striking result, however, was an increase in the number of females, suggesting that alterations induced by 5-aza-dC treatment can affect sexual development as well. Results were confirmed when treatment coincided with gonadal development. In addition, we also found that the adult gonadal transcriptome of 5-aza-dC-exposed females included significant changes in the expression of key reproduction-related genes (e.g. cyp11a1, esr2b and figla), and that several pro-female-related pathways such as the Fanconi anaemia or the Wnt signalling pathways were downregulated. Furthermore, an overall inhibition of genes implicated in epigenetic regulatory mechanisms (e.g. dnmt1, dicer, cbx4) was also observed. CONCLUSIONS Taken together, our results indicate that treatment with a DNA methylation inhibitor can also alter the sexual development in zebrafish, with permanent alterations of the adult gonadal transcriptome, at least in females. Our results show the importance of DNA methylation for proper control of sexual development, open new avenues for the potential control of sex ratios in fish (aquaculture, population control) and call attention to possibly hidden long-term effects of dnmt therapy when used, for example, in the treatment of prepuberal children affected by some types of cancer.
Collapse
Affiliation(s)
- Laia Ribas
- Institut de Ciències del Mar, Consejo Superior de Investigaciones Científicas (CSIC), Passeig Marítim, 37-45, 08003, Barcelona, Spain
| | - Konstantinos Vanezis
- Imperial Centre for Translational and Experimental Medicine, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| | - Marco Antonio Imués
- Departamento de Recursos Hidrobiológicos, Universidad de Nariño, Torobajo, Pasto, Colombia
| | - Francesc Piferrer
- Institut de Ciències del Mar, Consejo Superior de Investigaciones Científicas (CSIC), Passeig Marítim, 37-45, 08003, Barcelona, Spain.
| |
Collapse
|
20
|
Przybilla J, Hopp L, Lübbert M, Loeffler M, Galle J. Targeting DNA hypermethylation: Computational modeling of DNA demethylation treatment of acute myeloid leukemia. Epigenetics 2017; 12:886-896. [PMID: 28758855 DOI: 10.1080/15592294.2017.1361090] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
In acute myeloid leukemia (AML) DNA hypermethylation of gene promoters is frequently observed and often correlates with a block of differentiation. Treatment of AML patients with DNA methyltransferase inhibitors results in global hypomethylation of genes and, thereby, can lead to a reactivation of the differentiation capability. Unfortunately, after termination of treatment both hypermethylation and differentiation block return in most cases. Here, we apply, for the first time, a computational model of epigenetic regulation of transcription to: i) provide a mechanistic understanding of the DNA (de-) methylation process in AML and; ii) improve DNA demethylation treatment strategies. By in silico simulation, we analyze promoter hypermethylation scenarios referring to DNMT dysfunction, decreased H3K4me3 and increased H3K27me3 modification activity, and accelerated cell proliferation. We quantify differences between these scenarios with respect to gene repression and activation. Moreover, we compare the scenarios regarding their response to DNMT inhibitor treatment alone and in combination with inhibitors of H3K27me3 histone methyltransferases and of H3K4me3 histone demethylases. We find that the different hypermethylation scenarios respond specifically to therapy, suggesting that failure of remission originates in patient-specific deregulation. We observe that inappropriate demethylation therapy can result even in enforced deregulation. As an example, our results suggest that application of high DNMT inhibitor concentration can induce unwanted global gene activation if hypermethylation originates in increased H3K27me3 modification. Our results underline the importance of a personalized therapy requiring knowledge about the patient-specific mechanism of epigenetic deregulation.
Collapse
Affiliation(s)
- Jens Przybilla
- a Interdisciplinary Center for Bioinformatics, University of Leipzig , Leipzig , Germany.,d Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig , Leipzig , Germany
| | - Lydia Hopp
- a Interdisciplinary Center for Bioinformatics, University of Leipzig , Leipzig , Germany
| | - Michael Lübbert
- b Division of Hematology, Oncology and Stem Cell Transplantation, Faculty of Medicine, University of Freiburg , Freiburg , Germany.,c German Cancer Consortium (DKTK) , Freiburg , Germany
| | - Markus Loeffler
- d Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig , Leipzig , Germany
| | - Joerg Galle
- a Interdisciplinary Center for Bioinformatics, University of Leipzig , Leipzig , Germany
| |
Collapse
|
21
|
An Initial Investigation of an Alternative Model to Study rat Primordial Germ Cell Epigenetic Reprogramming. Biol Proced Online 2017; 19:9. [PMID: 28785173 PMCID: PMC5541664 DOI: 10.1186/s12575-017-0058-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 07/14/2017] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Primordial germ cells (PGC) are the precursors of the gametes. During pre-natal development, PGC undergo an epigenetic reprogramming when bulk DNA demethylation occurs and is followed by sex-specific de novo methylation. The de novo methylation and the maintenance of the methylation patterns depend on DNA methyltransferases (DNMTs). PGC reprogramming has been widely studied in mice but not in rats. We have previously shown that the rat might be an interesting model to study germ cell development. In face of the difficulties of getting enough PGC for molecular studies, the aim of this study was to propose an alternative method to study rat PGC DNA methylation. Rat embryos were collected at 14, 15 and 19 days post-coitus (dpc) for the analysis of 5mC, 5hmC, DNMT1, DNMT3a and DNMT3b expression or at 16dpc for treatment 5-Aza-CdR, a DNMT inhibitor, in vitro. METHODS Once collected, the gonads were placed in 24-well plates previously containing 45μm pore membrane and medium with or without 5-Aza-CdR. The culture was kept for five days and medium was changed daily. The gonads were either fixed or submitted to RNA extraction. RESULTS 5mC and DNMTs labelling suggests that PGC are undergoing epigenetic reprogramming around 14/15dpc. The in vitro treatment of rat embryonic gonads with 1 μM of 5-Aza-CdR lead to a loss of 5mC labelling and to the activation of Pax6 expression in PGC, but not in somatic cells, suggesting that 5-Aza-CdR promoted a PGC-specific global DNA hypomethylation. CONCLUSIONS This study suggests that the protocol used here can be a potential method to study the wide DNA demethylation that takes place during PGC reprogramming.
Collapse
|
22
|
Ponnaluri VKC, Ehrlich KC, Zhang G, Lacey M, Johnston D, Pradhan S, Ehrlich M. Association of 5-hydroxymethylation and 5-methylation of DNA cytosine with tissue-specific gene expression. Epigenetics 2017; 12:123-138. [PMID: 27911668 PMCID: PMC5330441 DOI: 10.1080/15592294.2016.1265713] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 11/10/2016] [Accepted: 11/21/2016] [Indexed: 12/15/2022] Open
Abstract
Differentially methylated or hydroxymethylated regions (DMRs) in mammalian DNA are often associated with tissue-specific gene expression but the functional relationships are still being unraveled. To elucidate these relationships, we studied 16 human genes containing myogenic DMRs by analyzing profiles of their epigenetics and transcription and quantitatively assaying 5-hydroxymethylcytosine (5hmC) and 5-methylcytosine (5mC) at specific sites in these genes in skeletal muscle (SkM), myoblasts, heart, brain, and diverse other samples. Although most human promoters have little or no methylation regardless of expression, more than half of the genes that we chose to study-owing to their myogenic DMRs-overlapped tissue-specific alternative or cryptic promoters displaying corresponding tissue-specific differences in histone modifications. The 5mC levels in myoblast DMRs were significantly associated with 5hmC levels in SkM at the same site. Hypermethylated myogenic DMRs within CDH15, a muscle- and cerebellum-specific cell adhesion gene, and PITX3, a homeobox gene, were used for transfection in reporter gene constructs. These intragenic DMRs had bidirectional tissue-specific promoter activity that was silenced by in vivo-like methylation. The CDH15 DMR, which was previously associated with an imprinted maternal germline DMR in mice, had especially strong promoter activity in myogenic host cells. These findings are consistent with the controversial hypothesis that intragenic DNA methylation can facilitate transcription and is not just a passive consequence of it. Our results support varied roles for tissue-specific 5mC- or 5hmC-enrichment in suppressing inappropriate gene expression from cryptic or alternative promoters and in increasing the plasticity of gene expression required for development and rapid responses to tissue stress or damage.
Collapse
Affiliation(s)
| | - Kenneth C. Ehrlich
- Center for Bioinformatics and Genomics, Tulane University Health Sciences Center, New Orleans, LA, USA
| | | | - Michelle Lacey
- Department of Mathematics, Tulane Health Sciences Center and Tulane University, New Orleans, LA, USA
| | - Douglas Johnston
- Department of Microbiology, Immunology and Parasitology, LSU Health Sciences Center, New Orleans, LA, USA
| | | | - Melanie Ehrlich
- Center for Bioinformatics and Genomics, Tulane University Health Sciences Center, New Orleans, LA, USA
- Hayward Genetics Center and Tulane Cancer Center, Tulane University Health Sciences Center, New Orleans, LA, USA
| |
Collapse
|
23
|
Shi W, Li K, Song L, Liu M, Wang Y, Liu W, Xia X, Qin Z, Zhen B, Wang Y, He F, Qin J, Ding C. Transcription Factor Response Elements on Tip: A Sensitive Approach for Large-Scale Endogenous Transcription Factor Quantitative Identification. Anal Chem 2016; 88:11990-11994. [DOI: 10.1021/acs.analchem.6b03150] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Wenhao Shi
- School of Life Sciences, Tsinghua University, Beijing 100084, China
- State Key Laboratory of Proteomics, Beijing Proteome Research Center,
Beijing Institute of Radiation Medicine, National Center for Protein Sciences (The PHOENIX Center, Beijing), Beijing 102206, China
| | - Kai Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center,
Beijing Institute of Radiation Medicine, National Center for Protein Sciences (The PHOENIX Center, Beijing), Beijing 102206, China
- Department of Pathogeny Biology, School
of Basic Medical Sciences, North China University of Science and Technology, Tangshan 063009, Hebei, China
| | - Lei Song
- School of Life Sciences, Tsinghua University, Beijing 100084, China
- State Key Laboratory of Proteomics, Beijing Proteome Research Center,
Beijing Institute of Radiation Medicine, National Center for Protein Sciences (The PHOENIX Center, Beijing), Beijing 102206, China
| | - Mingwei Liu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center,
Beijing Institute of Radiation Medicine, National Center for Protein Sciences (The PHOENIX Center, Beijing), Beijing 102206, China
| | - Yunzhi Wang
- State Key Laboratory of Genetic Engineering
and Collaborative Innovation Center for Genetics and Development,
School of Life Sciences, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Wanlin Liu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center,
Beijing Institute of Radiation Medicine, National Center for Protein Sciences (The PHOENIX Center, Beijing), Beijing 102206, China
| | - Xia Xia
- State Key Laboratory of Proteomics, Beijing Proteome Research Center,
Beijing Institute of Radiation Medicine, National Center for Protein Sciences (The PHOENIX Center, Beijing), Beijing 102206, China
| | - Zhaoyu Qin
- State Key Laboratory of Genetic Engineering
and Collaborative Innovation Center for Genetics and Development,
School of Life Sciences, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Bei Zhen
- State Key Laboratory of Proteomics, Beijing Proteome Research Center,
Beijing Institute of Radiation Medicine, National Center for Protein Sciences (The PHOENIX Center, Beijing), Beijing 102206, China
| | - Yi Wang
- Alkek Center for Molecular Discovery, Verna and Marrs
McLean Department of Biochemistry and Molecular Biology, Department
of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, United States
| | - Fuchu He
- School of Life Sciences, Tsinghua University, Beijing 100084, China
- State Key Laboratory of Proteomics, Beijing Proteome Research Center,
Beijing Institute of Radiation Medicine, National Center for Protein Sciences (The PHOENIX Center, Beijing), Beijing 102206, China
- State Key Laboratory of Genetic Engineering
and Collaborative Innovation Center for Genetics and Development,
School of Life Sciences, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Jun Qin
- State Key Laboratory of Proteomics, Beijing Proteome Research Center,
Beijing Institute of Radiation Medicine, National Center for Protein Sciences (The PHOENIX Center, Beijing), Beijing 102206, China
- State Key Laboratory of Genetic Engineering
and Collaborative Innovation Center for Genetics and Development,
School of Life Sciences, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
- Alkek Center for Molecular Discovery, Verna and Marrs
McLean Department of Biochemistry and Molecular Biology, Department
of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, United States
| | - Chen Ding
- State Key Laboratory of Proteomics, Beijing Proteome Research Center,
Beijing Institute of Radiation Medicine, National Center for Protein Sciences (The PHOENIX Center, Beijing), Beijing 102206, China
- State Key Laboratory of Genetic Engineering
and Collaborative Innovation Center for Genetics and Development,
School of Life Sciences, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| |
Collapse
|
24
|
Mor-Shaked H, Eiges R. Modeling Fragile X Syndrome Using Human Pluripotent Stem Cells. Genes (Basel) 2016; 7:genes7100077. [PMID: 27690107 PMCID: PMC5083916 DOI: 10.3390/genes7100077] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 09/02/2016] [Accepted: 09/12/2016] [Indexed: 02/06/2023] Open
Abstract
Fragile X syndrome (FXS) is the most common heritable form of cognitive impairment. It results from a loss-of-function mutation by a CGG repeat expansion at the 5′ untranslated region of the X-linked fragile X mental retardation 1 (FMR1) gene. Expansion of the CGG repeats beyond 200 copies results in protein deficiency by leading to aberrant methylation of the FMR1 promoter and the switch from active to repressive histone modifications. Additionally, the CGGs become increasingly unstable, resulting in high degree of variation in expansion size between and within tissues of affected individuals. It is still unclear how the FMR1 protein (FMRP) deficiency leads to disease pathology in neurons. Nor do we know the mechanisms by which the CGG expansion results in aberrant DNA methylation, or becomes unstable in somatic cells of patients, at least in part due to the lack of appropriate animal or cellular models. This review summarizes the current contribution of pluripotent stem cells, mutant human embryonic stem cells, and patient-derived induced pluripotent stem cells to disease modeling of FXS for basic and applied research, including the development of new therapeutic approaches.
Collapse
Affiliation(s)
- Hagar Mor-Shaked
- Stem Cell Research Laboratory, Medical Genetics Institute, Shaare Zedek Medical Center Affiliated with the Hebrew University School of Medicine, Jerusalem 91031, Israel.
| | - Rachel Eiges
- Stem Cell Research Laboratory, Medical Genetics Institute, Shaare Zedek Medical Center Affiliated with the Hebrew University School of Medicine, Jerusalem 91031, Israel.
| |
Collapse
|
25
|
Anastasakis D, Skeparnias I, Shaukat AN, Grafanaki K, Kanellou A, Taraviras S, Papachristou DJ, Papakyriakou A, Stathopoulos C. Mammalian PNLDC1 is a novel poly(A) specific exonuclease with discrete expression during early development. Nucleic Acids Res 2016; 44:8908-8920. [PMID: 27515512 PMCID: PMC5062988 DOI: 10.1093/nar/gkw709] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 08/02/2016] [Indexed: 12/28/2022] Open
Abstract
PNLDC1 is a homologue of poly(A) specific ribonuclease (PARN), a known deadenylase with additional role in processing of non-coding RNAs. Both enzymes were reported recently to participate in piRNA biogenesis in silkworm and C. elegans, respectively. To get insights on the role of mammalian PNLDC1, we characterized the human and mouse enzymes. PNLDC1 shows limited conservation compared to PARN and represents an evolutionary related but distinct group of enzymes. It is expressed specifically in mouse embryonic stem cells, human and mouse testes and during early mouse embryo development, while it fades during differentiation. Its expression in differentiated cells, is suppressed through methylation of its promoter by the de novo methyltransferase DNMT3B. Both enzymes are localized mainly in the ER and exhibit in vitro specificity restricted solely to 3′ RNA or DNA polyadenylates. Knockdown of Pnldc1 in mESCs and subsequent NGS analysis showed that although the expression of the remaining deadenylases remains unaffected, it affects genes involved mainly in reprogramming, cell cycle and translational regulation. Mammalian PNLDC1 is a novel deadenylase expressed specifically in cell types which share regulatory mechanisms required for multipotency maintenance. Moreover, it could be involved both in posttranscriptional regulation through deadenylation and genome surveillance during early development.
Collapse
Affiliation(s)
- Dimitrios Anastasakis
- Department of Biochemistry, School of Medicine, University of Patras, 26504 Rio Achaia, Greece
| | - Ilias Skeparnias
- Department of Biochemistry, School of Medicine, University of Patras, 26504 Rio Achaia, Greece
| | | | - Katerina Grafanaki
- Department of Biochemistry, School of Medicine, University of Patras, 26504 Rio Achaia, Greece
| | - Alexandra Kanellou
- Department of Physiology, School of Medicine, University of Patras, 26504 Rio Achaia, Greece
| | - Stavros Taraviras
- Department of Physiology, School of Medicine, University of Patras, 26504 Rio Achaia, Greece
| | - Dionysios J Papachristou
- Department of Anatomy, Histology and Embryology, School of Medicine, University of Patras, 26504 Rio Achaia, Greece
| | - Athanasios Papakyriakou
- Laboratory of Chemical Biology, National Centre for Scientific Research 'Demokritos', 15341 Athens, Greece
| | | |
Collapse
|
26
|
Marczylo EL, Jacobs MN, Gant TW. Environmentally induced epigenetic toxicity: potential public health concerns. Crit Rev Toxicol 2016; 46:676-700. [PMID: 27278298 PMCID: PMC5030620 DOI: 10.1080/10408444.2016.1175417] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Throughout our lives, epigenetic processes shape our development and enable us to adapt to a constantly changing environment. Identifying and understanding environmentally induced epigenetic change(s) that may lead to adverse outcomes is vital for protecting public health. This review, therefore, examines the present understanding of epigenetic mechanisms involved in the mammalian life cycle, evaluates the current evidence for environmentally induced epigenetic toxicity in human cohorts and rodent models and highlights the research considerations and implications of this emerging knowledge for public health and regulatory toxicology. Many hundreds of studies have investigated such toxicity, yet relatively few have demonstrated a mechanistic association among specific environmental exposures, epigenetic changes and adverse health outcomes in human epidemiological cohorts and/or rodent models. While this small body of evidence is largely composed of exploratory in vivo high-dose range studies, it does set a precedent for the existence of environmentally induced epigenetic toxicity. Consequently, there is worldwide recognition of this phenomenon, and discussion on how to both guide further scientific research towards a greater mechanistic understanding of environmentally induced epigenetic toxicity in humans, and translate relevant research outcomes into appropriate regulatory policies for effective public health protection.
Collapse
Affiliation(s)
- Emma L Marczylo
- a Toxicology Department, CRCE, PHE, Chilton , Oxfordshire , UK
| | - Miriam N Jacobs
- a Toxicology Department, CRCE, PHE, Chilton , Oxfordshire , UK
| | - Timothy W Gant
- a Toxicology Department, CRCE, PHE, Chilton , Oxfordshire , UK
| |
Collapse
|
27
|
Akinjo OO, Gant TW, Marczylo EL. Perturbation of epigenetic processes by doxorubicin in the mouse testis. Toxicol Res (Camb) 2016; 5:1229-1243. [PMID: 30090428 DOI: 10.1039/c6tx00078a] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 05/30/2016] [Indexed: 12/19/2022] Open
Abstract
Epigenetic processes play a major role in normal mammalian development, particularly during gametogenesis and early embryogenesis. Thus, perturbation of epigenetic processes in the testis by xenobiotics could have a major impact on testicular function and fertility, and potentially affect the development and health of subsequent generations. There has been substantial research into the epigenetic toxicity of environmental exposures over the last decade. However, few studies have focussed on pharmaceutical drugs, which due to the nature of their use are typically found at much higher concentrations within exposed individuals than environmental chemicals. Here, we investigated genome-wide changes in testicular mRNA transcription, microRNA expression and DNA methylation to assess the contribution of epigenetic mechanisms to the testicular toxicity induced by doxorubicin (DOX) as a representative, widely used and well-characterised anti-cancer drug. We demonstrated that DOX is able to induce transcriptional, microRNA and DNA methylation changes, which perturb pathways involved in stress/cell death and survival and testicular function and lead to germ cell loss and reproductive organ damage. This identified potential novel mechanisms of DOX-induced testicular toxicity for further focussed investigations. Such work is required to fully assess the role of epigenetics in toxicity, determine whether single and/or multigenerational epigenetic toxicity is a real public health concern, and begin to develop and incorporate relevant epigenetic endpoints into regulatory toxicology.
Collapse
Affiliation(s)
- Oluwajoba O Akinjo
- Toxicology Department , CRCE , PHE , Chilton , Oxfordshire OX11 0RQ , UK .
| | - Timothy W Gant
- Toxicology Department , CRCE , PHE , Chilton , Oxfordshire OX11 0RQ , UK .
| | - Emma L Marczylo
- Toxicology Department , CRCE , PHE , Chilton , Oxfordshire OX11 0RQ , UK .
| |
Collapse
|
28
|
Wang X, Chen E, Yang X, Wang Y, Quan Z, Wu X, Luo C. 5-azacytidine inhibits the proliferation of bladder cancer cells via reversal of the aberrant hypermethylation of the hepaCAM gene. Oncol Rep 2015; 35:1375-84. [PMID: 26677113 DOI: 10.3892/or.2015.4492] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Accepted: 07/24/2015] [Indexed: 11/06/2022] Open
Abstract
Hepatocyte cell adhesion molecule (hepaCAM), a tumor-suppressor gene, is rarely expressed in bladder carcinoma. However, little is known concerning the mechanisms of low hepaCAM expression in bladder cancer. Abnormal hypermethylation in the promoter plays a crucial role in cancer by silencing tumor-suppressor genes, which is catalyzed by DNA methyltransferases (DNMTs). In the present study, a total of 31 bladder cancer and 22 adjacent tissues were assessed by immunohistochemistry to detect DNMT3A/3B and hepaCAM expression. Methylation of hepaCAM was determined by methylation‑specific polymerase chain reaction (MSP). The mRNA and protein levels of DNMT3A/3B and hepaCAM were determined by RT-PCR and western blot analysis after treatment with 5-azacytidine (AZAC). Following AZAC treatment, the proliferation of bladder cancer cells was detected by CCK-8 and colony formation assays. Cell cycle distribution was examined by flow cytometry. To further evaluate the tumor‑suppressive roles of AZAC and the involved mechanisms, the anti-tumorigenicity of AZAC was tested in vivo. The expression of DNMT3A/3B protein was markedly increased in the bladder carcinoma tissues (P<0.05), and had a negative linear correlation with hepaCAM expression in the same patients according to Pearson's analysis (r=-0.7176/-0.7127, P<0.05). The MSP results indicated that the hepaCAM gene was hypermethylated in three bladder cancer cell lines. Furthermore, we found that downregulation of DNMT3A/3B expression, after treatment with AZAC, reversed the hypermethylation and expression of hepaCAM in bladder cancer cells. In addition, AZAC inhibited the proliferation of bladder cancer cells and arrested cells at the G0/G1 phase. The in vivo results showed that expression of DNMT3A/3B and hepaCAM as well as tumor growth of nude mice were markedly altered which corresponded with the in vitro results. Due to the ability to reactivate expression of hepaCAM and inhibit growth of bladder cancer cells, AZAC may represent an effective treatment for bladder cancer.
Collapse
Affiliation(s)
- Xiaorong Wang
- Key Laboratory of Diagnostic Medicine designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, P.R. China
| | - E Chen
- Key Laboratory of Diagnostic Medicine designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, P.R. China
| | - Xue Yang
- Key Laboratory of Diagnostic Medicine designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, P.R. China
| | - Yin Wang
- Key Laboratory of Diagnostic Medicine designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, P.R. China
| | - Zhen Quan
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Xiaohou Wu
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Chunli Luo
- Key Laboratory of Diagnostic Medicine designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, P.R. China
| |
Collapse
|
29
|
Yang B, Yang ZG, Gao B, Shao GG, Li GH. 5-Aza-CdR can reverse gefitinib resistance caused by DAPK gene promoter methylation in lung adenocarcinoma cells. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:12961-12966. [PMID: 26722491 PMCID: PMC4680436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 09/25/2015] [Indexed: 06/05/2023]
Abstract
To explore the relationship between death associated protein kinase (DAPK) gene promoter methylation and gefitinib resistance in Lung adenocarcinoma cell lines. EGFR-mutation lung adenocarcinoma cell lines PC9 and the gefitinib-resistant with T790M Mutation cell lines PC9/GR were chosen as cell models, and PC9/GR were treated with 5-aza-CdR (1 μmol/L). The experiments were divided into three groups: PC9 group, PC9/GR group and PC9/GR with 5-Aza-CdR pretreatment group. Treat three groups cell with different concentrations gefitinib, the cell proliferation was determined by MTT assay. The apoptotic rates were detected by flow cytometry. The methylation of DAPK gene promoter region was examined by methylation-specific PCR (MSP). The expressions of DAPK protein were detected by Western blot. MTT results showed that the half maximal inhibitory concentration (IC50) of PC9 and PC9/GR cell lines increase from 0.12 μmol/L to 8.52 μmol/L. But after treated with 5-aza-CdR, the IC50 of PC9/GR cell lines decrease to 4.35 μmol/L, and the resistance index (RI) decrease from 71 to 36 (P<0.05). Flow cytometry results showed that the apoptosis rate were 24.80% ± 0.28%, 12.70% ± 0.31%, 19.8% ± 0.15% respectively. MSP results showed that DAPK gene promoter region was un-methylated in PC9 cells and methylated in PC9/GR cells, when treated with 5-aza-CdR, DAPK gene promoter region was partly methylated in PC9/GR cells (P<0.05). Western blot results showed that the levels of DAPK protein were reduced significantly in PC9/GR cell lines compared with PC9, and after treated with 5-aza-CdR, the expression levels of DAPK protein in PC9/GR were increased (P<0.05). In conclusion, DAPK gene promoter methylation may contribute to the downregulation of DAPK gene and protein, and consequently affect the sensitivity of gefitinib in lung adenocarcinoma lines, induced gefitinib resistance. But 5-Aza-CdR can reverse gefitinib resistance by demethylation of DAPK gene promoter.
Collapse
Affiliation(s)
- Bo Yang
- Department of Thoracic Surgery, The First Bethune Hospital of Jilin University Changchun 13000, Jilin, China
| | - Zhi-Guang Yang
- Department of Thoracic Surgery, The First Bethune Hospital of Jilin University Changchun 13000, Jilin, China
| | - Bao Gao
- Department of Thoracic Surgery, The First Bethune Hospital of Jilin University Changchun 13000, Jilin, China
| | - Guo-Guang Shao
- Department of Thoracic Surgery, The First Bethune Hospital of Jilin University Changchun 13000, Jilin, China
| | - Guang-Hu Li
- Department of Thoracic Surgery, The First Bethune Hospital of Jilin University Changchun 13000, Jilin, China
| |
Collapse
|
30
|
Busch C, Burkard M, Leischner C, Lauer UM, Frank J, Venturelli S. Epigenetic activities of flavonoids in the prevention and treatment of cancer. Clin Epigenetics 2015; 7:64. [PMID: 26161152 PMCID: PMC4497414 DOI: 10.1186/s13148-015-0095-z] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 06/17/2015] [Indexed: 02/07/2023] Open
Abstract
Aberrant epigenetic modifications are described in an increasing number of pathological conditions, including neurodegenerative diseases, cardiovascular diseases, diabetes mellitus type 2, obesity and cancer. The general reversibility of epigenetic changes makes them an attractive and promising target e.g. in the treatment of cancer. Thus, a growing number of epigenetically active compounds are currently tested in clinical trials for their therapeutic potential. Interestingly, many phytochemicals present in plant foods, particularly flavonoids, are suggested to be able to alter epigenetic cellular mechanisms. Flavonoids are natural phenol compounds that form a large group of secondary plant metabolites with interesting biological activities. They can be categorized into six major subclasses, which display diverse properties affecting the two best characterized epigenetic mechanisms: modulation of the DNA methylation status and histone acetylation. High dietary flavonoid intake has strongly been suggested to reduce the risk of numerous cancer entities in a large body of epidemiological studies. Established health-promoting effects of diets rich in fruit and vegetables are faced by efforts to use purified flavonoids as supplements or pharmaceuticals, whereupon data on the latter applications remain controversial. The purpose of this review is to give an overview of current research on flavonoids to further elucidate their potential in cancer prevention and therapy, thereby focusing on their distinct epigenetic activities.
Collapse
Affiliation(s)
- Christian Busch
- Division of Dermatologic Oncology, Department of Dermatology and Allergology, Medical University Hospital, Tuebingen, Germany
| | - Markus Burkard
- Division of Dermatologic Oncology, Department of Dermatology and Allergology, Medical University Hospital, Tuebingen, Germany ; Department of Internal Medicine I, Medical University Hospital, Otfried-Mueller-Str. 27, 72076 Tuebingen, Germany
| | - Christian Leischner
- Department of Internal Medicine I, Medical University Hospital, Otfried-Mueller-Str. 27, 72076 Tuebingen, Germany
| | - Ulrich M Lauer
- Department of Internal Medicine I, Medical University Hospital, Otfried-Mueller-Str. 27, 72076 Tuebingen, Germany
| | - Jan Frank
- Institute of Biological Chemistry and Nutrition, University of Hohenheim, Stuttgart, Germany
| | - Sascha Venturelli
- Department of Internal Medicine I, Medical University Hospital, Otfried-Mueller-Str. 27, 72076 Tuebingen, Germany
| |
Collapse
|