1
|
Switzer RL, Hartman ZJ, Hewett GR, Carroll CF. Disease-Associated Mutation A554V Disrupts Normal Autoinhibition of DNMT1. DNA 2023; 3:119-133. [PMID: 37663147 PMCID: PMC10470860 DOI: 10.3390/dna3030010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
DNA methyltransferase 1 (DNMT1) is the enzyme primarily responsible for propagation of the methylation pattern in cells. Mutations in DNMT1 have been linked to the development of adult-onset neurodegenerative disorders; these disease-associated mutations occur in the regulatory replication foci-targeting sequence (RFTS) domain of the protein. The RFTS domain is an endogenous inhibitor of DNMT1 activity that binds to the active site and prevents DNA binding. Here, we examine the impact of the disease-associated mutation A554V on normal RFTS-mediated inhibition of DNMT1. Wild-type and mutant proteins were expressed and purified to homogeneity for biochemical characterization. The mutation increased DNA binding affinity ~8-fold. In addition, the mutant enzyme exhibited increased DNA methylation activity. Circular dichroism (CD) spectroscopy revealed that the mutation does not significantly impact the secondary structure or relative thermal stability of the isolated RFTS domain. However, the mutation resulted in changes in the CD spectrum in the context of the larger protein; a decrease in relative thermal stability was also observed. Collectively, this evidence suggests that A554V disrupts normal RFTS-mediated autoinhibition of DNMT1, resulting in a hyperactive mutant enzyme. While the disease-associated mutation does not significantly impact the isolated RFTS domain, the mutation results in a weakening of the interdomain stabilizing interactions generating a more open, active conformation of DNMT1. Hyperactive mutant DNMT1 could be responsible for the increased DNA methylation observed in affected individuals.
Collapse
Affiliation(s)
| | - Zach J. Hartman
- Department of Biology, Bucknell University, Lewisburg, PA 17837, USA
| | - Geoffrey R. Hewett
- Program in Cell Biology/Biochemistry, Bucknell University, Lewisburg, PA 17837, USA
| | - Clara F. Carroll
- Department of Chemistry, Bucknell University, Lewisburg, PA 17837, USA
| |
Collapse
|
2
|
Chen X, Zhu X, Dong J, Chen F, Gao Q, Zhang L, Cai D, Dong H, Ruan B, Wang Y, Jiang Q, Cao W. Reversal of Epigenetic Peroxisome Proliferator-Activated Receptor-γ Suppression by Diacerein Alleviates Oxidative Stress and Osteoarthritis in Mice. Antioxid Redox Signal 2022; 37:40-53. [PMID: 35196878 DOI: 10.1089/ars.2021.0219] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Aims: The pathogenesis of osteoarthritis (OA) is characterized by oxidative stress (OS) and sustained inflammation that are substantially associated with epigenetic DNA methylation alterations of osteogenic gene expression. Diacerein as an anthraquinone anti-OA drug exhibits multiple chondroprotective properties, but less clarified pharmacological actions. Since anthraquinone contain an epigenetic modulating property, in this study we investigate whether the anti-OA functions of diacerein involve DNA methylation modulation and antioxidant signaling. Results: The OA mice incurred by destabilization of medial meniscus exhibited marked suppression of peroxisome proliferator-activated receptor-gamma (PPARγ), a chondroprotective transcription factor with anti-inflammation and OS-balancing properties, aberrant upregulations of DNA methyltransferase (DNMT)1/3a, and PPARγ promoter hypermethylation in knee joint cartilage. Diacerein treatment mitigated the cartilage damage and significantly inhibited the DNMT1/3a upregulation, the PPARγ promoter hypermethylation, and the PPARγ loss, and it effectively corrected the adverse expression of antioxidant enzymes and inflammatory cytokines. In cultured chondrocytes, diacerein reduced the interleukin-1β-induced PPARγ suppression and the abnormal expression of its downstream antioxidant enzymes in a gain of DNMT and PPARγ inhibition-sensitive manner, and in PPARγ knockout mice, the anti-OA effects of diacerein were significantly reduced. Innovation: Our work reveals a novel anti-OA pharmacological property of diacerein and identifies the aberrant DNMT elevation and the resultant PPARγ suppression as an important epigenetic pathway that mediates diacerein's anti-OA activities. Conclusion: DNA methylation aberration and the resultant PPARγ suppression contribute significantly to epigenetic OA pathogenesis, and targeting PPARγ suppression via DNA demethylation is an important component of diacerein's anti-OA functions. Antioxid. Redox Signal. 37, 40-53.
Collapse
Affiliation(s)
- Xingren Chen
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedics, State Key Lab of Pharmaceutical Biotechnology, Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University School of Medicine, Nanjing, China
| | - Xiaobo Zhu
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedics, State Key Lab of Pharmaceutical Biotechnology, Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University School of Medicine, Nanjing, China
| | - Jian Dong
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedics, State Key Lab of Pharmaceutical Biotechnology, Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University School of Medicine, Nanjing, China
| | - Fang Chen
- Nanjing University School of Medicine, Department of Basic Medical Science, Jiangsu Key Laboratory of Molecular Medicine, Nanjing, China
| | - Qi Gao
- Nanjing University School of Medicine, Department of Basic Medical Science, Jiangsu Key Laboratory of Molecular Medicine, Nanjing, China
| | - Lijun Zhang
- Nanjing University School of Medicine, Department of Basic Medical Science, Jiangsu Key Laboratory of Molecular Medicine, Nanjing, China
| | - Dawei Cai
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedics, State Key Lab of Pharmaceutical Biotechnology, Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University School of Medicine, Nanjing, China
| | - Hui Dong
- Department of Orthopedics, Northern Jiangsu People's Hospital, The Affiliated Hospital of Nanjing University Medical School, Yangzhou, China
| | - Binjia Ruan
- Department of Orthopedics, Northern Jiangsu People's Hospital, The Affiliated Hospital of Nanjing University Medical School, Yangzhou, China
| | - Yongxiang Wang
- Department of Orthopedics, Northern Jiangsu People's Hospital, The Affiliated Hospital of Nanjing University Medical School, Yangzhou, China
| | - Qing Jiang
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedics, State Key Lab of Pharmaceutical Biotechnology, Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University School of Medicine, Nanjing, China
| | - Wangsen Cao
- Nanjing University School of Medicine, Department of Basic Medical Science, Jiangsu Key Laboratory of Molecular Medicine, Nanjing, China
| |
Collapse
|
3
|
Petrova D, Naumenko M, Khantakova D, Grin I, Zharkov D. Relative Efficiency of Recognition of 5-Methylcytosine and 5-Hydroxymethylcytosine by Methyl-Dependent DNA Endonuclease GlaI. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2020. [DOI: 10.1134/s1068162019060323] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
4
|
Dolen EK, McGinnis JH, Tavory RN, Weiss JA, Switzer RL. Disease-Associated Mutations G589A and V590F Relieve Replication Focus Targeting Sequence-Mediated Autoinhibition of DNA Methyltransferase 1. Biochemistry 2019; 58:5151-5159. [PMID: 31804802 DOI: 10.1021/acs.biochem.9b00749] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In eukaryotes, the most common epigenetic DNA modification is methylation of carbon 5 of cytosines, predominantly in CpG dinucleotides. Methylation patterns are established and maintained by a family of proteins known as DNA methyltransferases (DNMTs). DNA methylation is an important epigenetic mark associated with gene repression, and disruption of the normal DNA methylation pattern is known to play a role in several disease states. Methylation patterns are primarily maintained by DNMT1, which possesses specificity for methylation of hemimethylated DNA. DNMT1 is a multidomain protein with a C-terminal catalytic methyltransferase domain and a large N-terminal regulatory region. The replication focus targeting sequence (RFTS) domain, found in the regulatory region, is an endogenous inhibitor of DNMT1 activity. Recently, several mutations in the RFTS domain were shown to be causal for two adult onset neurodegenerative diseases; however, little is known about the impact of these mutations on the structure and function of DNMT1. Two of these mutations, G589A and V590F, are associated with development of autosomal dominant cerebellar ataxia, deafness, and narcolepsy (ADCA-DN). We have successfully expressed and purified G589A and V590F DNMT1 for in vitro studies. The mutations significantly decrease the thermal stability of DNMT1, yet the mutant proteins exhibit 2.5-3.5-fold increases in DNA binding affinity. In addition, the mutations weaken RFTS-mediated inhibition of DNA methylation activity. Taken together, these data suggest these disease-associated mutations decrease protein stability and, at least partially, relieve normal RFTS-mediated autoinhibition of DNMT1.
Collapse
Affiliation(s)
- Emma K Dolen
- Department of Chemistry , Bucknell University , Lewisburg , Pennsylvania 17837 , United States
| | - James H McGinnis
- Program in Cell Biology/Biochemistry , Bucknell University , Lewisburg , Pennsylvania 17837 , United States
| | - Rachel N Tavory
- Program in Cell Biology/Biochemistry , Bucknell University , Lewisburg , Pennsylvania 17837 , United States
| | - Jill A Weiss
- Program in Cell Biology/Biochemistry , Bucknell University , Lewisburg , Pennsylvania 17837 , United States
| | - Rebecca L Switzer
- Department of Chemistry , Bucknell University , Lewisburg , Pennsylvania 17837 , United States
| |
Collapse
|
5
|
Switzer RL, Medrano J, Reedel DA, Weiss J. Substituted anthraquinones represent a potential scaffold for DNA methyltransferase 1-specific inhibitors. PLoS One 2019; 14:e0219830. [PMID: 31306451 PMCID: PMC6629088 DOI: 10.1371/journal.pone.0219830] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 07/03/2019] [Indexed: 12/13/2022] Open
Abstract
In humans, the most common epigenetic DNA modification is methylation of the 5-carbon of cytosines, predominantly in CpG dinucleotides. DNA methylation is an important epigenetic mark associated with gene repression. Disruption of the normal DNA methylation pattern is known to play a role in the initiation and progression of many cancers. DNA methyltransferase 1 (DNMT1), the most abundant DNA methyltransferase in humans, is primarily responsible for maintenance of the DNA methylation pattern and is considered an important cancer drug target. Recently, laccaic acid A (LCA), a highly substituted anthraquinone natural product, was identified as a direct, DNA-competitive inhibitor of DNMT1. Here, we have successfully screened a small library of simplified anthraquinone compounds for DNMT1 inhibition. Using an endonuclease-coupled DNA methylation assay, we identified two anthraquinone compounds, each containing an aromatic substituent, that act as direct DNMT1 inhibitors. These simplified anthraquinone compounds retain the DNA-competitive mechanism of action of LCA and exhibit some selectivity for DNMT1 over DNMT3a. The newly identified compounds are at least 40-fold less potent than LCA, but have significantly less complex structures. Collectively, this data indicates that substituted anthraquinone compounds could serve as a novel scaffold for developing DNMT1-specific inhibitors.
Collapse
Affiliation(s)
- Rebecca L. Switzer
- Department of Chemistry, Bucknell University, Lewisburg, Pennsylvania, United States of America
- * E-mail:
| | - Jessica Medrano
- Program in Cell Biology/Biochemistry, Bucknell University, Lewisburg, Pennsylvania, United States of America
| | - David A. Reedel
- Program in Cell Biology/Biochemistry, Bucknell University, Lewisburg, Pennsylvania, United States of America
| | - Jill Weiss
- Program in Cell Biology/Biochemistry, Bucknell University, Lewisburg, Pennsylvania, United States of America
| |
Collapse
|
6
|
Loo SK, Ch'ng ES, Lawrie CH, Muruzabal MA, Gaafar A, Pomposo MP, Husin A, Md Salleh MS, Banham AH, Pedersen LM, Møller MB, Green TM, Wong KK. DNMT1 is predictive of survival and associated with Ki-67 expression in R-CHOP-treated diffuse large B-cell lymphomas. Pathology 2017; 49:731-739. [PMID: 29074044 DOI: 10.1016/j.pathol.2017.08.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Revised: 08/16/2017] [Accepted: 08/20/2017] [Indexed: 11/26/2022]
Abstract
DNMT1 is a target of approved anti-cancer drugs including decitabine. However, the prognostic value of DNMT1 protein expression in R-CHOP-treated diffuse large B-cell lymphomas (DLBCLs) remains unexplored. Here we showed that DNMT1 was expressed in the majority of DLBCL cases (n = 209/230, 90.9%) with higher expression in germinal centre B-cell-like (GCB)-DLBCL subtype. Low and negative DNMT1 expression (20% cut-off, n = 33/230, 14.3%) was predictive of worse overall survival (OS; p < 0.001) and progression-free survival (PFS; p < 0.001). Nonetheless, of the 209 DNMT1 positive patients, 33% and 42% did not achieve 5-year OS and PFS, respectively, indicating that DNMT1 positive patients showed considerably heterogeneous outcomes. Moreover, DNMT1 was frequently expressed in mitotic cells and significantly correlated with Ki-67 or BCL6 expression (r = 0.60 or 0.44, respectively; p < 0.001). We demonstrate that DNMT1 is predictive of DLBCL patients' survival, and suggest that DNMT1 could be a DLBCL therapeutic target due to its significant association with Ki-67.
Collapse
Affiliation(s)
- Suet Kee Loo
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Ewe Seng Ch'ng
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam, Kepala Batas, Pulau Pinang, Malaysia
| | - Charles H Lawrie
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom; Oncology Department, Biodonostia Research Institute, San Sebastian, Spain
| | | | - Ayman Gaafar
- Department of Pathology, Hospital Universitario Cruces, Barakaldo, Spain
| | | | - Azlan Husin
- Department of Medicine, School of Medical Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Md Salzihan Md Salleh
- Department of Pathology, School of Medical Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Alison H Banham
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Lars M Pedersen
- Department of Haematology, Herlev University Hospital, Copenhagen, Denmark
| | - Michael B Møller
- Department of Pathology, Odense University Hospital, Odense, Denmark
| | - Tina M Green
- Department of Pathology, Odense University Hospital, Odense, Denmark
| | - Kah Keng Wong
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kelantan, Malaysia.
| |
Collapse
|
7
|
Thakur S, Brenner C. KRAS-driven miR-29b expression is required for tumor suppressor gene silencing. Oncotarget 2017; 8:74755-74766. [PMID: 29088821 PMCID: PMC5650376 DOI: 10.18632/oncotarget.20364] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Accepted: 07/26/2017] [Indexed: 12/16/2022] Open
Abstract
KRAS activation drives DNA methylation and silencing of specific tumor suppressor genes (TSGs). We previously showed that the ERK pathway induces transcriptional repression of TET1, which results in conversion of TSG promoters from a hydroxymethylated, active state to a hypermethylated and silenced state. Here we identified miR-29b as a KRAS-induced molecule that represses TET1 expression. In KRAS-transformed cells, ectopic miR-29b inhibition restores expression of TET1, thereby reactivating TSGs by reducing methylation and restoring hydroxymethylation. Mining gene expression data of lung cancer cell lines identified additional TSGs suppressed by KRAS signaling whose expression was restored by inhibition of miR-29b and re-expression of TET1. Because KRAS changes TSG promoters from hydroxymethylated to hypermethylated with miR-29b-dependent silencing of TET1, we demonstrate a model in which DNMT1 is present on target promoters prior to KRAS transformation. In addition, we propose miR-29b as a potential circulating biomarker and target for rational treatment of specific malignancies.
Collapse
Affiliation(s)
- Shilpa Thakur
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Charles Brenner
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
8
|
Cayo MA, Mallanna SK, Di Furio F, Jing R, Tolliver LB, Bures M, Urick A, Noto FK, Pashos EE, Greseth MD, Czarnecki M, Traktman P, Yang W, Morrisey EE, Grompe M, Rader DJ, Duncan SA. A Drug Screen using Human iPSC-Derived Hepatocyte-like Cells Reveals Cardiac Glycosides as a Potential Treatment for Hypercholesterolemia. Cell Stem Cell 2017; 20:478-489.e5. [PMID: 28388428 DOI: 10.1016/j.stem.2017.01.011] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 12/22/2016] [Accepted: 01/27/2017] [Indexed: 12/17/2022]
Abstract
Efforts to identify pharmaceuticals to treat heritable metabolic liver diseases have been hampered by the lack of models. However, cells with hepatocyte characteristics can be produced from induced pluripotent stem cells (iPSCs). Here, we have used hepatocyte-like cells generated from homozygous familial hypercholesterolemia (hoFH) iPSCs to identify drugs that can potentially be repurposed to lower serum LDL-C. We found that cardiac glycosides reduce the production of apolipoprotein B (apoB) from human hepatocytes in culture and the serum of avatar mice harboring humanized livers. The drugs act by increasing the turnover of apoB protein. Analyses of patient medical records revealed that the treatment of patients with cardiac glycosides reduced serum LDL-C levels. These studies highlight the effectiveness of using iPSCs to screen for potential treatments for inborn errors of hepatic metabolism and suggest that cardiac glycosides could provide an approach for reducing hepatocyte production of apoB and treating hypercholesterolemia.
Collapse
Affiliation(s)
- Max A Cayo
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Sunil K Mallanna
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA
| | - Francesca Di Furio
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA
| | - Ran Jing
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA
| | - Lauren B Tolliver
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA
| | - Matthew Bures
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Amanda Urick
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA; Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA
| | - Fallon K Noto
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Evanthia E Pashos
- Departments of Medicine and Genetics and the Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Matthew D Greseth
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA
| | - Maciej Czarnecki
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA; Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Paula Traktman
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA; Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA; Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas Street, MSC 955, Charleston, SC 29425, USA
| | - Wenli Yang
- Department of Medicine and Penn Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Edward E Morrisey
- Department of Medicine and Penn Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Markus Grompe
- Papé Family Pediatric Research Institute, Oregon Health & Science University, 3181 South West Sam Jackson Park Road/L321, Portland, OR 97239, USA
| | - Daniel J Rader
- Departments of Medicine and Genetics and the Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Stephen A Duncan
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA; Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA; Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas Street, MSC 955, Charleston, SC 29425, USA.
| |
Collapse
|
9
|
Xu P, Hu G, Luo C, Liang Z. DNA methyltransferase inhibitors: an updated patent review (2012-2015). Expert Opin Ther Pat 2016; 26:1017-30. [PMID: 27376512 DOI: 10.1080/13543776.2016.1209488] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
INTRODUCTION DNA methyltransferases (DNMTs), important enzymes involved in epigenetic regulation of gene expression, represent promising targets in cancer therapy. DNMT inhibitors (DNMTi), which can modulate the aberrant DNA methylation pattern in a reversible way via inhibiting DNMT activity, have attracted significant attention in recent years. AREAS COVERED This review outlines the newly patented inhibitors targeting DNMTs, mainly incorporating small molecular inhibitors and oligonucleotide derivatives. The chemical structures, biological activity, and the encouraging clinical research in progress are delineated in detail. EXPERT OPINION Two drugs, azacitidine and decitabine, have evidently shown efficacy in hematologic malignancies, yet do not work well on solid tumors, have low specificity, substantial toxicity, and poor bioavailability. With the rapid advancement in systems biology, drug combinations, such as DNMTi, in conjugation with histone deacetylase inhibitors (HDACi) or immunotherapy, probably serve as an efficient way of implementing epigenetic therapy. Meanwhile, the resolved autoinhibitory structures of DNMTs afford a novel strategy for targeting the protein-protein interface involved in the autoinhi-bitory interactions. The molecular mechanism underlying the conformational transitions would also shed new light on the design of allosteric inhibitors. Both strategies would produce inhibitors with more selectivity compared to nucleotide derivatives.
Collapse
Affiliation(s)
- Pan Xu
- a Center for Systems Biology , Soochow University , Jiangsu , China.,b Shanghai Institute of Materia Medica, State Key Laboratory of Drug Research , Chinese Academy of Sciences , Shanghai , China
| | - Guang Hu
- a Center for Systems Biology , Soochow University , Jiangsu , China
| | - Cheng Luo
- b Shanghai Institute of Materia Medica, State Key Laboratory of Drug Research , Chinese Academy of Sciences , Shanghai , China
| | - Zhongjie Liang
- a Center for Systems Biology , Soochow University , Jiangsu , China
| |
Collapse
|
10
|
Chapleau RR, McElroy CA, Ruark CD, Fleming EJ, Ghering AB, Schlager JJ, Poeppelman LD, Gearhart JM. High-Throughput Screening for Positive Allosteric Modulators Identified Potential Therapeutics against Acetylcholinesterase Inhibition. JOURNAL OF BIOMOLECULAR SCREENING 2015; 20:1142-9. [PMID: 26078409 DOI: 10.1177/1087057115591006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 05/20/2015] [Indexed: 11/16/2022]
Abstract
The current standard of care for treatment of organophosphate (OP) poisoning includes pretreatment with the weak reversible acetylcholinesterase (AChE) inhibitor pyridostigmine bromide. Because this drug is an AChE inhibitor, similar side effects exist as with OP poisoning. In an attempt to provide a therapeutic capable of mitigating AChE inhibition without such side effects, high-throughput screening was performed to identify a compound capable of increasing the catalytic activity of AChE. Herein, two such novel positive allosteric modulators (PAMs) of AChE are presented. These PAMs increase AChE activity threefold, but they fail to upshift the apparent IC50 of a variety of OPs. Further development and optimization of these compounds may lead to pre- and/or postexposure therapeutics with broad-spectrum efficacy against pesticide and nerve agent poisoning. In addition, they could be used to complement the current therapeutic standard of care to increase the activity of uninhibited AChE, potentially increasing the efficacy of current therapeutics in addition to altering the therapeutic window.
Collapse
Affiliation(s)
- Richard R Chapleau
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Wright Patterson AFB, OH, USA Molecular Bioeffects Branch, Bioeffects Division, Human Effectiveness Directorate, 711th Human Performance Wing, Air Force Research Laboratory (711 HPW/RHDJ), Wright Patterson AFB, OH, USA
| | - Craig A McElroy
- College of Pharmacy, Ohio State University, Columbus, OH, USA
| | - Christopher D Ruark
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Wright Patterson AFB, OH, USA Molecular Bioeffects Branch, Bioeffects Division, Human Effectiveness Directorate, 711th Human Performance Wing, Air Force Research Laboratory (711 HPW/RHDJ), Wright Patterson AFB, OH, USA
| | - Emily J Fleming
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Wright Patterson AFB, OH, USA Molecular Bioeffects Branch, Bioeffects Division, Human Effectiveness Directorate, 711th Human Performance Wing, Air Force Research Laboratory (711 HPW/RHDJ), Wright Patterson AFB, OH, USA
| | - Amy B Ghering
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Wright Patterson AFB, OH, USA Molecular Bioeffects Branch, Bioeffects Division, Human Effectiveness Directorate, 711th Human Performance Wing, Air Force Research Laboratory (711 HPW/RHDJ), Wright Patterson AFB, OH, USA
| | - John J Schlager
- Molecular Bioeffects Branch, Bioeffects Division, Human Effectiveness Directorate, 711th Human Performance Wing, Air Force Research Laboratory (711 HPW/RHDJ), Wright Patterson AFB, OH, USA
| | - Lee D Poeppelman
- Molecular Bioeffects Branch, Bioeffects Division, Human Effectiveness Directorate, 711th Human Performance Wing, Air Force Research Laboratory (711 HPW/RHDJ), Wright Patterson AFB, OH, USA
| | - Jeffery M Gearhart
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Wright Patterson AFB, OH, USA Molecular Bioeffects Branch, Bioeffects Division, Human Effectiveness Directorate, 711th Human Performance Wing, Air Force Research Laboratory (711 HPW/RHDJ), Wright Patterson AFB, OH, USA
| |
Collapse
|
11
|
Medina-Franco JL, Méndez-Lucio O, Dueñas-González A, Yoo J. Discovery and development of DNA methyltransferase inhibitors using in silico approaches. Drug Discov Today 2014; 20:569-77. [PMID: 25526932 DOI: 10.1016/j.drudis.2014.12.007] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Revised: 11/19/2014] [Accepted: 12/10/2014] [Indexed: 01/08/2023]
Abstract
Multiple strategies have evolved during the past few years to advance epigenetic compounds targeting DNA methyltransferases (DNMTs). Significant progress has been made in HTS, lead optimization and determination of 3D structures of DNMTs. In light of the emerging concept of epi-informatics, computational approaches are employed to accelerate the development of DNMT inhibitors helping to screen chemical databases, mine the DNMT-relevant chemical space, uncover SAR and design focused libraries. Computational methods also synergize with natural-product-based drug discovery and drug repurposing. Herein, we survey the latest developments of in silico approaches to advance epigenetic drug and probe discovery targeting DNMTs.
Collapse
Affiliation(s)
- José L Medina-Franco
- Facultad de Química, Departamento de Farmacia, Universidad Nacional Autónoma de México, Avenida Universidad 3000, Mexico City 04510, Mexico.
| | - Oscar Méndez-Lucio
- Unilever Centre for Molecular Science Informatics, Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
| | - Alfonso Dueñas-González
- Unidad de Investigación Biomédica en Cáncer, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México and Instituto Nacional de Cancerología, Av. San Fernando 22, Mexico City 14080, Mexico
| | - Jakyung Yoo
- Life Science Research Institute, Daewoong Pharmaceutical Co. Ltd., 72 Dugye-Ro, Pogok-Eup, Gyeonggi-do 449-814, Republic of Korea
| |
Collapse
|
12
|
Erdmann A, Halby L, Fahy J, Arimondo PB. Targeting DNA Methylation with Small Molecules: What’s Next? J Med Chem 2014; 58:2569-83. [DOI: 10.1021/jm500843d] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Alexandre Erdmann
- Epigenetic Targeting of Cancer,
USR3388 ETaC, CNRS-Pierre Fabre, 3 Avenue H. Curien, 31035 Toulouse Cedex 01, France
| | - Ludovic Halby
- Epigenetic Targeting of Cancer,
USR3388 ETaC, CNRS-Pierre Fabre, 3 Avenue H. Curien, 31035 Toulouse Cedex 01, France
| | - Jacques Fahy
- Epigenetic Targeting of Cancer,
USR3388 ETaC, CNRS-Pierre Fabre, 3 Avenue H. Curien, 31035 Toulouse Cedex 01, France
| | - Paola B Arimondo
- Epigenetic Targeting of Cancer,
USR3388 ETaC, CNRS-Pierre Fabre, 3 Avenue H. Curien, 31035 Toulouse Cedex 01, France
| |
Collapse
|
13
|
Suppression of TET1-dependent DNA demethylation is essential for KRAS-mediated transformation. Cell Rep 2014; 9:1827-1840. [PMID: 25466250 DOI: 10.1016/j.celrep.2014.10.063] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Revised: 09/23/2014] [Accepted: 10/25/2014] [Indexed: 12/31/2022] Open
Abstract
Hypermethylation-mediated tumor suppressor gene (TSG) silencing is a central epigenetic alteration in RAS-dependent tumorigenesis. Ten-eleven translocation (TET) enzymes can depress DNA methylation by hydroxylation of 5-methylcytosine (5mC) bases to 5-hydroxymethylcytosine (5hmC). Here, we report that suppression of TET1 is required for KRAS-induced DNA hypermethylation and cellular transformation. In distinct nonmalignant cell lines, oncogenic KRAS promotes transformation by inhibiting TET1 expression via the ERK-signaling pathway. This reduces chromatin occupancy of TET1 at TSG promoters, lowers levels of 5hmC, and increases levels of 5mC and 5mC-dependent transcriptional silencing. Restoration of TET1 expression by ERK pathway inhibition or ectopic TET1 reintroduction in KRAS-transformed cells reactivates TSGs and inhibits colony formation. KRAS knockdown increases TET1 expression and diminishes colony-forming ability, whereas KRAS/TET1 double knockdown bypasses the KRAS dependence of KRAS-addicted cancer cells. Thus, suppression of TET1-dependent DNA demethylation is critical for KRAS-mediated transformation.
Collapse
|
14
|
Challenges in developing novel DNA methyltransferases inhibitors for cancer therapy. Future Med Chem 2014; 6:1237-40. [DOI: 10.4155/fmc.14.82] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
|
15
|
Nikiforov TT. Generic assay format for endo- and exonucleases based on fluorogenic substrates labeled with single fluorophores. Anal Biochem 2014; 461:67-73. [PMID: 24907506 DOI: 10.1016/j.ab.2014.05.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Revised: 05/16/2014] [Accepted: 05/24/2014] [Indexed: 12/25/2022]
Abstract
We previously described the development of fluorogenic assays for nucleic acid-modifying enzymes based on synthetic oligonucleotides labeled with a single fluorophore. In the current work, we studied the performance of such singly labeled substrates as a function of the nucleotide sequence in the vicinity of the fluorophore and the nature of the fluorophore itself. In agreement with published studies, we found that a 3' end of the primer terminating in a dC residue opposite a 5' dG provides the greatest degree of fluorophore quenching. Adding a second dC residue at the 3' penultimate position opposite another dG increased the quenching further. Among the various dyes tested, the difluoro substituted fluorescein derivative Oregon Green emerged as a superior fluorophore for this assay format. We have now combined these findings into a new generic format for endonuclease assays. This format allows a substrate for any endonuclease to be obtained rapidly by simply replacing the enzyme's recognition sequence within the generic labeled molecule. Compared with our previous format, the new assays show greatly expanded signal dynamic ranges. The format is applicable to other nucleic acid-modifying enzymes such as exonucleases (e.g., T7 gene 6 exonuclease) and DNA repair enzymes (e.g., uracil-DNA glycosylase).
Collapse
|