1
|
Amatori S, Persico G, Cantatore F, Rusin M, Formica M, Giorgi L, Macedi E, Casciaro F, Errico Provenzano A, Gambardella S, Noberini R, Bonaldi T, Fusi V, Giorgio M, Fanelli M. Small molecule-induced epigenomic reprogramming of APL blasts leading to antiviral-like response and c-MYC downregulation. Cancer Gene Ther 2022; 30:671-682. [PMID: 36536122 DOI: 10.1038/s41417-022-00576-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/23/2022] [Accepted: 11/30/2022] [Indexed: 12/23/2022]
Abstract
AbstractAcute promyelocytic leukemia (APL) is an aggressive subtype of acute myeloid leukemia (AML) in which the PML/RARα fusion protein exerts oncogenic activities by recruiting repressive complexes to the promoter of specific target genes. Other epigenetic perturbations, as alterations of histone H3 lysine 9 trimethylation (H3K9me3), have been frequently found in AMLs and are associated with leukemogenesis and leukemia progression. Here, we characterized the epigenomic effects of maltonis, a novel maltol-derived molecule, in APL cells. We demonstrate that maltonis treatments induce a profound remodulation of the histone code, reducing global H3K9me3 signal and modulating other histone post-translational modifications. Transcriptomic and epigenomic analyses revealed that maltonis exposure induces changes of genes expression associated with a genomic redistribution of histone H3 lysine 4 trimethylation (H3K4me3) and lysine 27 acetylation (H3K27ac). Upregulation of interferon alpha and gamma response and downregulation of c-MYC target genes, in function of c-MYC reduced expression (monitored in all the hematopoietic neoplasms tested), represent the most significant modulated pathways. These data demonstrate the ability of maltonis to epigenetically reprogram the gene expression profile of APL cells, inducing an intriguing antiviral-like response, concomitantly with the downregulation of c-MYC-related pathways, thus making it an attractive candidate for antileukemic therapy.
Collapse
|
2
|
Bioinformatics analysis of the network of histone H3 lysine 9 trimethylation in acute myeloid leukaemia. Oncol Rep 2020; 44:543-554. [PMID: 32468066 PMCID: PMC7336454 DOI: 10.3892/or.2020.7627] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 05/06/2020] [Indexed: 12/20/2022] Open
Abstract
Changes in histone H3 lysine 9 trimethylation (H3K9me3) may be related to the development of drug-resistant acute myeloid leukaemia (AML); insights into the network of H3K9me3 may improve patient prognosis. Patient data were derived from the Gene Expression Omnibus (GEO) database and data from AML cells treated with chidamide, a novel benzamide chemical class of histone deacetylase inhibitor (HDACi), in vitro were derived from ChIP-seq. Patients and AML cell data were analysed using GEO2R, GOseq, KOBAS, the STRING database and Cytoscape 3.5.1. We identified several genes related to the upregulation or downregulation of H3K9me3 in AML patients; some of these genes were related to apoptosis, autophagy, and the pathway of cell longevity. AML cells treated with chidamide in vitro showed the same gene changes. The protein interactions in the network did not have significantly more interactions than expected, suggesting the need for more research to identify these interactions. One compelling result from the protein interaction study was that sirtuin 1 (SIRT1) may have an indirect interaction with lysine-specific demethylase 4A (KDM4A). These results help explain alterations of H3K9me3 in AML that may direct further studies aimed at improving patient prognosis. These results may also provide a basis for chidamide as a treatment strategy for AML patients in the future.
Collapse
|
3
|
Monaghan L, Massett ME, Bunschoten RP, Hoose A, Pirvan PA, Liskamp RMJ, Jørgensen HG, Huang X. The Emerging Role of H3K9me3 as a Potential Therapeutic Target in Acute Myeloid Leukemia. Front Oncol 2019; 9:705. [PMID: 31428579 PMCID: PMC6687838 DOI: 10.3389/fonc.2019.00705] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 07/16/2019] [Indexed: 12/23/2022] Open
Abstract
Growing evidence has demonstrated that epigenetic dysregulation is a common pathological feature in human cancer cells. Global alterations in the epigenetic landscape are prevalent in malignant cells across different solid tumors including, prostate cancer, non-small-cell lung cancer, renal cell carcinoma, and in haemopoietic malignancy. In particular, DNA hypomethylation and histone hypoacetylation have been observed in acute myeloid leukemia (AML) patient blasts, with histone methylation being an emerging area of study. Histone 3 lysine 9 trimethylation (H3K9me3) is a post-translational modification known to be involved in the regulation of a broad range of biological processes, including the formation of transcriptionally silent heterochromatin. Following the observation of its aberrant methylation status in hematological malignancy and several other cancer phenotypes, recent studies have associated H3K9me3 levels with patient outcome and highlighted key molecular mechanisms linking H3K9me3 profile with AML etiology in a number of large-scale meta-analysis. Consequently, the development and application of small molecule inhibitors which target the histone methyltransferases or demethylase enzymes known to participate in the oncogenic regulation of H3K9me3 in AML represents an advancing area of ongoing study. Here, we provide a comprehensive review on how this particular epigenetic mark is regulated within cells and its emerging role as a potential therapeutic target in AML, along with an update on the current research into advancing the generation of more potent and selective inhibitors against known H3K9 methyltransferases and demethylases.
Collapse
Affiliation(s)
- Laura Monaghan
- Haemato-Oncology/Systems Medicine Group, Paul O'Gorman Leukemia Research Center, Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Matthew E. Massett
- Haemato-Oncology/Systems Medicine Group, Paul O'Gorman Leukemia Research Center, Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | | | - Alex Hoose
- School of Chemistry, University of Glasgow, Glasgow, United Kingdom
| | | | | | - Heather G. Jørgensen
- Haemato-Oncology/Systems Medicine Group, Paul O'Gorman Leukemia Research Center, Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Xu Huang
- Haemato-Oncology/Systems Medicine Group, Paul O'Gorman Leukemia Research Center, Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
4
|
Cardoso BA, Ramos TL, Belo H, Vilas-Boas F, Real C, Almeida AM. Vorinostat synergizes with antioxidant therapy to target myeloproliferative neoplasms. Exp Hematol 2019; 72:60-71.e11. [PMID: 30769020 DOI: 10.1016/j.exphem.2019.02.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 02/07/2019] [Accepted: 02/09/2019] [Indexed: 02/06/2023]
Abstract
BCR-ABL-negative myeloproliferative neoplasms (MPNs) are driven by JAK-STAT pathway activation, but epigenetic alterations also play an important pathophysiological role. These can be pharmacologically manipulated with histone deacetylase inhibitors (HDACIs), which have proven to be clinically effective in the treatment of MPNs but exhibit dose-limiting toxicity. The treatment of primary MPN cells with vorinostat modulates the expression of genes associated with apoptosis, cell cycle, inflammation, and signaling. The induction of this transcriptional program results in decreased cellular viability, paralleled by a decrease in levels of reactive oxygen species (ROS). In vitro manipulation of ROS levels revealed that the reduction of ROS levels promoted apoptosis. When vorinostat was combined with antioxidant agents, the apoptosis of MPN cells increased in a synergistic manner. The results described here suggest a novel and promising therapeutic strategy combining HDACIs with ROS-reducing agents to treat MPNs.
Collapse
Affiliation(s)
- Bruno A Cardoso
- Unidade de Investigação em Patobiologia Molecular, Instituto Português de Oncologia de Lisboa Francisco Gentil, E.P.E, Lisboa, Portugal; Centro de Estudos de Doenças Crónicas, CEDOC, NOVA Medical School/Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisboa, Portugal
| | - Teresa L Ramos
- Unidade de Investigação em Patobiologia Molecular, Instituto Português de Oncologia de Lisboa Francisco Gentil, E.P.E, Lisboa, Portugal; Universidad de Salamanca-IBSAL-Hospital Universitario, Servicio de Hematología, Salamanca, Spain
| | - Hélio Belo
- Unidade de Investigação em Patobiologia Molecular, Instituto Português de Oncologia de Lisboa Francisco Gentil, E.P.E, Lisboa, Portugal; Centro de Estudos de Doenças Crónicas, CEDOC, NOVA Medical School/Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisboa, Portugal
| | - Filipe Vilas-Boas
- Centro de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - Carla Real
- Centro de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - António M Almeida
- Unidade de Investigação em Patobiologia Molecular, Instituto Português de Oncologia de Lisboa Francisco Gentil, E.P.E, Lisboa, Portugal; Centro de Estudos de Doenças Crónicas, CEDOC, NOVA Medical School/Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisboa, Portugal; Hospital da Luz, Lisboa, Portugal.
| |
Collapse
|
5
|
Zhang YY, Zhou JD, Yang DQ, He PF, Yao DM, Qian Z, Yang J, Xu WR, Lin J, Qian J. Intragenic hypomethylation of DNMT3A in patients with myelodysplastic syndrome. Clin Chem Lab Med 2018; 56:485-491. [PMID: 29031013 DOI: 10.1515/cclm-2016-0142] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 09/07/2017] [Indexed: 01/12/2023]
Abstract
BACKGROUND DNMT3A is a DNA methyltransferase that acts in de novo methylation. Aberrant expression of DNMT3A has been reported in several human diseases, including myelodysplastic syndrome (MDS). However, the pattern of DNMT3A methylation remains unknown in MDS. METHODS The present study was aimed to investigate the methylation status of DNMT3A intragenic differentially methylated region 2 (DMR2) using real-time quantitative methylation-specific PCR and analyze its clinical significance in MDS. RESULTS Aberrant hypomethylation of DNMT3A was found in 57% (51/90) MDS cases. There were no significant differences in age, sex, white blood cell counts, platelet counts, hemoglobin counts and World Health Organization, International Prognostic Scoring System and karyotype classifications between DNMT3A hypomethylated and DNMT3A hypermethylated groups. However, the patients with DNMT3A hypomethylation had shorter overall survival time than those without DNMT3A hypomethylation (11 months vs. 36 months, p=0.033). Multivariate analysis confirmed the independent adverse impact of DNMT3A hypomethylation in MDS. CONCLUSIONS Our data suggest that DNMT3A DMR2 hypomethylation may be a negative prognostic hallmark in MDS.
Collapse
Affiliation(s)
- Ying-Ying Zhang
- Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, P.R. China.,Department of Hematology and Oncology, Yizheng People's Hospital, Yangzhou, Jiangsu, P.R. China
| | - Jing-Dong Zhou
- Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, P.R. China.,The Key Laboratory of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, Jiangsu, P.R. China
| | - Dong-Qin Yang
- Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, P.R. China.,The Key Laboratory of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, Jiangsu, P.R. China
| | - Pin-Fang He
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, P.R. China.,The Key Laboratory of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, Jiangsu, P.R. China
| | - Dong-Ming Yao
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, P.R. China.,The Key Laboratory of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, Jiangsu, P.R. China
| | - Zhen Qian
- Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, P.R. China.,The Key Laboratory of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, Jiangsu, P.R. China
| | - Jing Yang
- Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, P.R. China.,The Key Laboratory of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, Jiangsu, P.R. China
| | - Wen-Rong Xu
- Key Laboratory of Laboratory Medicine of Jiangsu Province, Medical Key Talent Project of Zhenjiang, Zhenjiang, P.R. China
| | - Jiang Lin
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, P.R. China.,The Key Laboratory of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, Jiangsu, P.R. China.,Laboratory Center, Affiliated People's Hospital of Jiangsu University, 8 Dianli Rd., 212002 Zhenjiang, P.R. China, Fax: +86.511.85234387
| | - Jun Qian
- Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, P.R. China.,The Key Laboratory of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, Jiangsu, P.R. China.,Department of Hematology, Affiliated People's Hospital of Jiangsu University, 8 Dianli Rd., 212002 Zhenjiang, P.R. China, Fax: +86.511.85234387
| |
Collapse
|
6
|
Palomo L, Malinverni R, Cabezón M, Xicoy B, Arnan M, Coll R, Pomares H, García O, Fuster-Tormo F, Grau J, Feliu E, Solé F, Buschbeck M, Zamora L. DNA methylation profile in chronic myelomonocytic leukemia associates with distinct clinical, biological and genetic features. Epigenetics 2018; 13:8-18. [PMID: 29160764 DOI: 10.1080/15592294.2017.1405199] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Chromosomal abnormalities are detected in 20-30% of patients with chronic myelomonocytic leukemia (CMML) and correlate with prognosis. On the mutation level, disruptive alterations are particularly frequent in chromatin regulatory genes. However, little is known about the consequential alterations in the epigenetic marking of the genome. Here, we report the analysis of genomic DNA methylation patterns of 64 CMML patients and 10 healthy controls, using a DNA methylation microarray focused on promoter regions. Differential methylation analysis between patients and controls allowed us to identify abnormalities in DNA methylation, including hypermethylation of specific genes and large genome regions with aberrant DNA methylation. Unsupervised hierarchical cluster analysis identified two main clusters that associated with the clinical, biological, and genetic features of patients. Group 1 was enriched in patients with adverse clinical and biological characteristics and poorer overall and progression-free survival. In addition, significant differences in DNA methylation were observed between patients with low risk and intermediate/high risk karyotypes and between TET2 mutant and wild type patients. Taken together, our results demonstrate that altered DNA methylation patterns reflect the CMML disease state and allow to identify patient groups with distinct clinical features.
Collapse
Affiliation(s)
- Laura Palomo
- a MDS Group. Josep Carreras Leukaemia Research Institute (IJC), ICO-Hospital Germans Trias i Pujol , Universitat Autònoma de Barcelona , Carretera de Can Ruti, Camí de les Escoles, s/n. 08916, Badalona ( Barcelona ), Spain.,b Departament de Bioquímica i Biologia Molecular , Universitat Autònoma de Barcelona , Campus de la UAB, Plaça Cívica, s/n. 08913, Bellaterra ( Barcelona ), Spain
| | - Roberto Malinverni
- c Chromatin, Metabolism and Cell Fate Group. Josep Carreras Leukaemia Research Institute (IJC), Campus ICO-Hospital Germans Trias i Pujol , Program for Predictive and Personalized Medicine of Cancer at the Institute Germans Trias i Pujol (PMPPC-IGTP) , Carretera de Can Ruti, Camí de les Escoles, s/n. 08916, Badalona ( Barcelona ), Spain
| | - Marta Cabezón
- d Hematology Service, ICO-Hospital Germans Trias i Pujol, Josep Carreras Leukaemia Research Institute (IJC) , Universitat Autònoma de Barcelona , Carretera del Canyet, s/n. 08916, Badalona ( Barcelona ), Spain
| | - Blanca Xicoy
- d Hematology Service, ICO-Hospital Germans Trias i Pujol, Josep Carreras Leukaemia Research Institute (IJC) , Universitat Autònoma de Barcelona , Carretera del Canyet, s/n. 08916, Badalona ( Barcelona ), Spain
| | - Montserrat Arnan
- e Hematology Service , ICO-Hospital Duran i Reynals , Avinguda de la Gran Via de l'Hospitalet, 199-203, 08908 Hospitalet de Llobregat ( Barcelona ), Spain
| | - Rosa Coll
- f Hematology Service , ICO-Girona Hospital Josep Trueta, Girona, Spain , Avenida França, s/n. 17007 Girona , Spain
| | - Helena Pomares
- e Hematology Service , ICO-Hospital Duran i Reynals , Avinguda de la Gran Via de l'Hospitalet, 199-203, 08908 Hospitalet de Llobregat ( Barcelona ), Spain
| | - Olga García
- d Hematology Service, ICO-Hospital Germans Trias i Pujol, Josep Carreras Leukaemia Research Institute (IJC) , Universitat Autònoma de Barcelona , Carretera del Canyet, s/n. 08916, Badalona ( Barcelona ), Spain
| | - Francisco Fuster-Tormo
- a MDS Group. Josep Carreras Leukaemia Research Institute (IJC), ICO-Hospital Germans Trias i Pujol , Universitat Autònoma de Barcelona , Carretera de Can Ruti, Camí de les Escoles, s/n. 08916, Badalona ( Barcelona ), Spain
| | - Javier Grau
- d Hematology Service, ICO-Hospital Germans Trias i Pujol, Josep Carreras Leukaemia Research Institute (IJC) , Universitat Autònoma de Barcelona , Carretera del Canyet, s/n. 08916, Badalona ( Barcelona ), Spain
| | - Evarist Feliu
- d Hematology Service, ICO-Hospital Germans Trias i Pujol, Josep Carreras Leukaemia Research Institute (IJC) , Universitat Autònoma de Barcelona , Carretera del Canyet, s/n. 08916, Badalona ( Barcelona ), Spain
| | - Francesc Solé
- a MDS Group. Josep Carreras Leukaemia Research Institute (IJC), ICO-Hospital Germans Trias i Pujol , Universitat Autònoma de Barcelona , Carretera de Can Ruti, Camí de les Escoles, s/n. 08916, Badalona ( Barcelona ), Spain
| | - Marcus Buschbeck
- c Chromatin, Metabolism and Cell Fate Group. Josep Carreras Leukaemia Research Institute (IJC), Campus ICO-Hospital Germans Trias i Pujol , Program for Predictive and Personalized Medicine of Cancer at the Institute Germans Trias i Pujol (PMPPC-IGTP) , Carretera de Can Ruti, Camí de les Escoles, s/n. 08916, Badalona ( Barcelona ), Spain
| | - Lurdes Zamora
- d Hematology Service, ICO-Hospital Germans Trias i Pujol, Josep Carreras Leukaemia Research Institute (IJC) , Universitat Autònoma de Barcelona , Carretera del Canyet, s/n. 08916, Badalona ( Barcelona ), Spain
| |
Collapse
|
7
|
Sakaguchi H, Muramatsu H, Okuno Y, Makishima H, Xu Y, Furukawa-Hibi Y, Wang X, Narita A, Yoshida K, Shiraishi Y, Doisaki S, Yoshida N, Hama A, Takahashi Y, Yamada K, Miyano S, Ogawa S, Maciejewski JP, Kojima S. Aberrant DNA Methylation Is Associated with a Poor Outcome in Juvenile Myelomonocytic Leukemia. PLoS One 2015; 10:e0145394. [PMID: 26720758 PMCID: PMC4697810 DOI: 10.1371/journal.pone.0145394] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 12/03/2015] [Indexed: 12/18/2022] Open
Abstract
Juvenile myelomonocytic leukemia (JMML), an overlap of myelodysplastic / myeloproliferative neoplasm, is an intractable pediatric myeloid neoplasm. Epigenetic regulation of transcription, particularly by CpG methylation, plays an important role in tumor progression, mainly by repressing tumor-suppressor genes. To clarify the clinical importance of aberrant DNA methylation, we studied the hypermethylation status of 16 target genes in the genomes of 92 patients with JMML by bisulfite conversion and the pryosequencing technique. Among 16 candidate genes, BMP4, CALCA, CDKN2A, and RARB exhibited significant hypermethylation in 72% (67/92) of patients. Based on the number of hypermethylated genes, patients were stratified into three cohorts based on an aberrant methylation score (AMS) of 0, 1–2, or 3–4. In the AMS 0 cohort, the 5-year overall survival (OS) and transplantation-free survival (TFS) were good (69% and 76%, respectively). In the AMS 1–2 cohort, the 5-year OS was comparable to that in the AMS 0 cohort (68%), whereas TFS was poor (6%). In the AMS 3–4 cohort, 5-year OS and TFS were markedly low (8% and 0%, respectively). Epigenetic analysis provides helpful information for clinicians to select treatment strategies for patients with JMML. For patients with AMS 3–4 in whom hematopoietic stem cell transplantation does not improve the prognosis, alternative therapies, including DNA methyltransferase inhibitors and new molecular-targeting agents, should be established as treatment options.
Collapse
Affiliation(s)
- Hirotoshi Sakaguchi
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Hematology and Oncology, Children’s Medical Center, Japanese Red Cross Nagoya First Hospital, Nagoya, Japan
| | - Hideki Muramatsu
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yusuke Okuno
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hideki Makishima
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
- Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yinyan Xu
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoko Furukawa-Hibi
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Xinan Wang
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Atsushi Narita
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kenichi Yoshida
- Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yuichi Shiraishi
- Laboratory of DNA information Analysis, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Sayoko Doisaki
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Nao Yoshida
- Department of Hematology and Oncology, Children’s Medical Center, Japanese Red Cross Nagoya First Hospital, Nagoya, Japan
| | - Asahito Hama
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshiyuki Takahashi
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kiyofumi Yamada
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Satoru Miyano
- Laboratory of DNA information Analysis, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Seishi Ogawa
- Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Jaroslaw P. Maciejewski
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Seiji Kojima
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
- * E-mail:
| |
Collapse
|
8
|
Lai YS, Chen JY, Tsai HJ, Chen TY, Hung WC. The SUV39H1 inhibitor chaetocin induces differentiation and shows synergistic cytotoxicity with other epigenetic drugs in acute myeloid leukemia cells. Blood Cancer J 2015; 5:e313. [PMID: 25978433 PMCID: PMC4476016 DOI: 10.1038/bcj.2015.37] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 04/14/2015] [Indexed: 12/26/2022] Open
Abstract
Epigenetic modifying enzymes have a crucial role in the pathogenesis of acute myeloid leukemia (AML). Methylation of lysine 9 on histone H3 by the methyltransferase G9a and SUV39H1 is associated with inhibition of tumor suppressor genes. We studied the effect of G9a and SUV39H1 inhibitors on viability and differentiation of AML cells and tested the cytotoxicity induced by combination of G9a and SUV39H1 inhibitors and various epigenetic drugs. The SUV39H1 inhibitor (chaetocin) and the G9a inhibitor (UNC0638) caused cell death in AML cells at high concentrations. However, only chaetocin-induced CD11b expression and differentiation of AML cells at non-cytotoxic concentration. HL-60 and KG-1a cells were more sensitive to chaetocin than U937 cells. Long-term incubation of chaetocin led to downregulation of SUV39H1 and reduction of H3K9 tri-methylation in HL-60 and KG-1a cells. Combination of chaetocin with suberoylanilide hydroxamic acid (SAHA, a histone deacetylase inhibitor) or JQ (a BET (bromodomain extra terminal) bromodomain inhibitor) showed synergistic cytotoxicity. Conversely, no synergism was found by combining chaetocin and UNC0638. More importantly, chaetocin-induced differentiation and combined cytotoxicity were also found in the primary cells of AML patients. Collectively, the SUV39H1 inhibitor chaetocin alone or in combination with other epigenetic drugs may be effective for the treatment of AML.
Collapse
Affiliation(s)
- Y-S Lai
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - J-Y Chen
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - H-J Tsai
- 1] National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan [2] Division of Hematology/Oncology, Department of Internal Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
| | - T-Y Chen
- Division of Hematology/Oncology, Department of Internal Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
| | - W-C Hung
- 1] National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan [2] Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
9
|
Bodoor K, Haddad Y, Alkhateeb A, Al-Abbadi A, Dowairi M, Magableh A, Bsoul N, Ghabkari A. DNA hypermethylation of cell cycle (p15 and p16) and apoptotic (p14, p53, DAPK and TMS1) genes in peripheral blood of leukemia patients. Asian Pac J Cancer Prev 2014; 15:75-84. [PMID: 24528084 DOI: 10.7314/apjcp.2014.15.1.75] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Aberrant DNA methylation of tumor suppressor genes has been reported in all major types of leukemia with potential involvement in the inactivation of regulatory cell cycle and apoptosis genes. However, most of the previous reports did not show the extent of concurrent methylation of multiple genes in the four leukemia types. Here, we analyzed six key genes (p14, p15, p16, p53, DAPK and TMS1) for DNA methylation using methylation specific PCR to analyze peripheral blood of 78 leukemia patients (24 CML, 25 CLL, 12 AML, and 17 ALL) and 24 healthy volunteers. In CML, methylation was detected for p15 (11%), p16 (9%), p53 (23%) and DAPK (23%), in CLL, p14 (25%), p15 (19%), p16 (12%), p53 (17%) and DAPK (36%), in AML, p14 (8%), p15 (45%), p53 (9%) and DAPK (17%) and in ALL, p15 (14%), p16 (8%), and p53 (8%). This study highlighted an essential role of DAPK methylation in chronic leukemia in contrast to p15 methylation in the acute cases, whereas TMS1 hypermethylation was absent in all cases. Furthermore, hypermethylation of multiple genes per patient was observed, with obvious selectiveness in the 9p21 chromosomal region genes (p14, p15 and p16). Interestingly, methylation of p15 increased the risk of methylation in p53, and vice versa, by five folds (p=0.03) indicating possible synergistic epigenetic disruption of different phases of the cell cycle or between the cell cycle and apoptosis. The investigation of multiple relationships between methylated genes might shed light on tumor specific inactivation of the cell cycle and apoptotic pathways.
Collapse
Affiliation(s)
- Khaldon Bodoor
- Department of Biology, Jordan University of Science and Technology, Irbid, Jordan E-mail :
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Pretreatment of leukemic cells with low-dose decitabine markedly enhances the cytotoxicity of gemtuzumab ozogamicin. Leukemia 2012; 27:233-5. [PMID: 22814292 PMCID: PMC3542629 DOI: 10.1038/leu.2012.178] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
11
|
Fan R, Zhang LY, Wang H, Yang B, Han T, Zhao XL, Wang W, Wang XQ, Lin GW. Methylation of the CpG Island Near SOX7 Gene Promoter Is Correlated with the Poor Prognosis of Patients with Myelodysplastic Syndrome. TOHOKU J EXP MED 2012; 227:119-128. [DOI: 10.1620/tjem.227.119] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Affiliation(s)
- Rong Fan
- Department of Hematology, Huashan Hospital of Fudan University
| | | | - Hong Wang
- Department of Hematology, The First Affiliated Hospital of Soochow University
| | - Bo Yang
- Fudan-Cinpathogen Clinical & Molecular Research Center
| | - Tao Han
- Fudan-Cinpathogen Clinical & Molecular Research Center
| | - Xiao-Li Zhao
- Department of Hematology, Huashan Hospital of Fudan University
| | - Wei Wang
- Department of Hematology, Huashan Hospital of Fudan University
| | - Xiao-Qin Wang
- Department of Hematology, Huashan Hospital of Fudan University
| | - Guo-Wei Lin
- Department of Hematology, Huashan Hospital of Fudan University
| |
Collapse
|
12
|
Hussein K, Theophile K, Büsche G, Schlegelberger B, Göhring G, Kreipe H, Bock O. Significant inverse correlation of microRNA-150/MYB and microRNA-222/p27 in myelodysplastic syndrome. Leuk Res 2010; 34:328-34. [DOI: 10.1016/j.leukres.2009.06.014] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2009] [Revised: 06/11/2009] [Accepted: 06/11/2009] [Indexed: 10/20/2022]
|
13
|
Potapova A, Hasemeier B, Römermann D, Metzig K, Göhring G, Schlegelberger B, Länger F, Kreipe H, Lehmann U. Epigenetic inactivation of tumour suppressor gene KLF11 in myelodysplastic syndromes*. Eur J Haematol 2009; 84:298-303. [PMID: 20002157 DOI: 10.1111/j.1600-0609.2009.01389.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The identification of aberrantly hypermethylated genes may lead to the development of new diagnostic markers and the identification of novel targets of epigenetic therapy in myelodysplastic syndromes (MDS). We therefore investigated the methylation status of transcription factor genes KLF5, KLF11, and MAFB, shown to be aberrantly methylated in myelogeneous leukaemia cells, in a series of 115 MDS patient as well as in 25 control subjects. Using quantitative high-resolution pyrosequencing methodology, KLF11, MAFB, and KLF5 were shown for the first time to be hypermethylated in 17 (15%), 8 (7%), and 2 (1.7%) cases, respectively, but not in any of the patients with an isolated 5q-deletion. Patient samples harbouring KLF11 methylation displayed reduced KLF11 mRNA expression and KLF11 hypermethylation correlated with a high International Prognostic Scoring System score (P < 0.05). In conclusion, epigenetic inactivation and subsequent transcriptional repression of the KLF11 gene is quite frequent in MDS. Patients with an isolated 5q-deletion seem to harbour a distinct epigenetic profile.
Collapse
Affiliation(s)
- Anna Potapova
- Institute of Pathology, Medizinische Hochschule Hannover, Hannover, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Wang H, Wang XQ, Xu XP, Lin GW. ID4 methylation predicts high risk of leukemic transformation in patients with myelodysplastic syndrome. Leuk Res 2009; 34:598-604. [PMID: 19853913 DOI: 10.1016/j.leukres.2009.09.031] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2009] [Revised: 09/24/2009] [Accepted: 09/24/2009] [Indexed: 12/24/2022]
Abstract
Epigenetic gene silencing due to promoter methylation is observed in human cancers like acute myeloid leukemia (AML). Little is known about aberrant methylation in myelodysplastic syndrome (MDS), a heterogeneous clonal stem cell disorder with a approximately 30% risk of transformation into secondary AML. Recent evidence demonstrated that ID4, a negative regulator of transcription, may act as a tumor-suppressor gene. To clarify the role of ID4 in MDS, we employed methylation-specific PCR (MSP) to examine the methylation status of ID4 in 144 adult de novo MDS patients. We found that ID4 methylation was present in 35.4% (n=51) of these MDS patients and methylaiton was correlated significantly with World Health Organization (WHO) subtypes and International Prognostic Scoring System (IPSS) risk groups. Patients with advanced stages of WHO subtypes (45.8% vs. 21.3%, P=0.002) and higher risk IPSS subgroups (45.7% vs. 26.0%, P=0.014) exhibited a significantly higher frequency of ID4 methylation. The median survival of patients with ID4 methylation was shorter than patients without ID4 methylation (12.2 months vs. 26.9 months, P=0.005). Multivariate analysis indicated that ID4 methylation status was the independent factor that impacted leukemia-free survival (LFS). Disease in patients with ID4 methylation progressed more rapidly than those without ID4 methylation (P=0.047, HR=2.11). Our results suggest that ID4 may be a therapeutic target in MDS.
Collapse
Affiliation(s)
- Hong Wang
- Department of Haematology, Huashan Hospital of Fudan University, 12 Wulumuqi Road Central, 200040 Shanghai, China
| | | | | | | |
Collapse
|
15
|
Martin-Subero JI, Ammerpohl O, Bibikova M, Wickham-Garcia E, Agirre X, Alvarez S, Brüggemann M, Bug S, Calasanz MJ, Deckert M, Dreyling M, Du MQ, Dürig J, Dyer MJS, Fan JB, Gesk S, Hansmann ML, Harder L, Hartmann S, Klapper W, Küppers R, Montesinos-Rongen M, Nagel I, Pott C, Richter J, Román-Gómez J, Seifert M, Stein H, Suela J, Trümper L, Vater I, Prosper F, Haferlach C, Cigudosa JC, Siebert R. A comprehensive microarray-based DNA methylation study of 367 hematological neoplasms. PLoS One 2009; 4:e6986. [PMID: 19750229 PMCID: PMC2737286 DOI: 10.1371/journal.pone.0006986] [Citation(s) in RCA: 98] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2009] [Accepted: 08/12/2009] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Alterations in the DNA methylation pattern are a hallmark of leukemias and lymphomas. However, most epigenetic studies in hematologic neoplasms (HNs) have focused either on the analysis of few candidate genes or many genes and few HN entities, and comprehensive studies are required. METHODOLOGY/PRINCIPAL FINDINGS Here, we report for the first time a microarray-based DNA methylation study of 767 genes in 367 HNs diagnosed with 16 of the most representative B-cell (n = 203), T-cell (n = 30), and myeloid (n = 134) neoplasias, as well as 37 samples from different cell types of the hematopoietic system. Using appropriate controls of B-, T-, or myeloid cellular origin, we identified a total of 220 genes hypermethylated in at least one HN entity. In general, promoter hypermethylation was more frequent in lymphoid malignancies than in myeloid malignancies, being germinal center mature B-cell lymphomas as well as B and T precursor lymphoid neoplasias those entities with highest frequency of gene-associated DNA hypermethylation. We also observed a significant correlation between the number of hypermethylated and hypomethylated genes in several mature B-cell neoplasias, but not in precursor B- and T-cell leukemias. Most of the genes becoming hypermethylated contained promoters with high CpG content, and a significant fraction of them are targets of the polycomb repressor complex. Interestingly, T-cell prolymphocytic leukemias show low levels of DNA hypermethylation and a comparatively large number of hypomethylated genes, many of them showing an increased gene expression. CONCLUSIONS/SIGNIFICANCE We have characterized the DNA methylation profile of a wide range of different HNs entities. As well as identifying genes showing aberrant DNA methylation in certain HN subtypes, we also detected six genes--DBC1, DIO3, FZD9, HS3ST2, MOS, and MYOD1--that were significantly hypermethylated in B-cell, T-cell, and myeloid malignancies. These might therefore play an important role in the development of different HNs.
Collapse
Affiliation(s)
- Jose I. Martin-Subero
- Institute of Human Genetics, Christian-Albrechts University, Kiel, Germany
- Cancer Epigenetics and Biology Program, Bellvitge Institute for Biomedical Research-Catalan Institute of Oncology, Barcelona, Spain
| | - Ole Ammerpohl
- Institute of Human Genetics, Christian-Albrechts University, Kiel, Germany
| | - Marina Bibikova
- Illumina, Inc., San Diego, California, United States of America
| | | | - Xabier Agirre
- Division of Cancer and Area of Cell Therapy and Hematology Service, Universidad de Navarra, Pamplona, Spain
| | - Sara Alvarez
- Molecular Cytogenetics Group, Centro Nacional Investigaciones Oncologicas, Madrid, Spain
| | - Monika Brüggemann
- Second Medical Department, Christian-Albrechts University, Kiel, Germany
| | - Stefanie Bug
- Institute of Human Genetics, Christian-Albrechts University, Kiel, Germany
| | | | - Martina Deckert
- Department of Neuropathology, University Hospital of Cologne, Cologne, Germany
| | - Martin Dreyling
- Department of Medicine III, University Hospital Grosshadern, Munich, Germany
| | - Ming Q. Du
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Jan Dürig
- Department of Hematology, University of Duisburg-Essen, Essen, Germany
| | | | - Jian-Bing Fan
- Illumina, Inc., San Diego, California, United States of America
| | - Stefan Gesk
- Institute of Human Genetics, Christian-Albrechts University, Kiel, Germany
| | - Martin-Leo Hansmann
- Institute of Pathology, University Hospital of Frankfurt, Frankfurt, Germany
| | - Lana Harder
- Institute of Human Genetics, Christian-Albrechts University, Kiel, Germany
| | - Sylvia Hartmann
- Institute of Pathology, University Hospital of Frankfurt, Frankfurt, Germany
| | - Wolfram Klapper
- Institute of Pathology, Christian-Albrechts University, Kiel, Germany
| | - Ralf Küppers
- Institute of Cell Biology, University of Duisburg-Essen, Essen, Germany
| | | | - Inga Nagel
- Institute of Human Genetics, Christian-Albrechts University, Kiel, Germany
| | - Christiane Pott
- Second Medical Department, Christian-Albrechts University, Kiel, Germany
| | - Julia Richter
- Institute of Human Genetics, Christian-Albrechts University, Kiel, Germany
| | - José Román-Gómez
- Reina Sofia Hospital, Instituto Maimonides de Investigación Biomédica de Córdoba, Cordoba, Spain
| | - Marc Seifert
- Institute of Cell Biology, University of Duisburg-Essen, Essen, Germany
| | - Harald Stein
- Institute of Pathology, Campus Benjamin Franklin, Berlin, Germany
| | - Javier Suela
- Molecular Cytogenetics Group, Centro Nacional Investigaciones Oncologicas, Madrid, Spain
| | - Lorenz Trümper
- Department of Hematology and Oncology, Georg-August University of Göttingen, Göttingen, Germany
| | - Inga Vater
- Institute of Human Genetics, Christian-Albrechts University, Kiel, Germany
| | - Felipe Prosper
- Division of Cancer and Area of Cell Therapy and Hematology Service, Universidad de Navarra, Pamplona, Spain
| | | | - Juan Cruz Cigudosa
- Molecular Cytogenetics Group, Centro Nacional Investigaciones Oncologicas, Madrid, Spain
| | - Reiner Siebert
- Institute of Human Genetics, Christian-Albrechts University, Kiel, Germany
| |
Collapse
|
16
|
Plimack ER, Kantarjian HM, Issa JP. Decitabine and its role in the treatment of hematopoietic malignancies. Leuk Lymphoma 2009; 48:1472-81. [PMID: 17701577 DOI: 10.1080/10428190701471981] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
DNA methylation is responsible for abnormal silencing of many genes, including tumor suppressor genes, in cancer. Decitabine, an S-phase specific inhibitor of DNA methyltransferase, has been shown to decrease levels of abnormal methylation in neoplasia. Though initially investigated at high doses as a cytotoxic agent, recent studies show that when administered at low doses, the hypomethylating activity of decitabine is increased with a demonstrated increase in activity in hematopoietic malignancies. Multiple clinical trials, both in the United States and in Europe, have demonstrated the efficacy of decitabine in acute myeloid leukemia, chronic myeloid leukemia, and myelodysplastic syndrome (MDS). Recently approved by the United States Food and Drug Administration for the treatment of (MDS), decitabine represents an effective and well-tolerated therapeutic option in this disease, for which treatment options were previously scarce. While the activity in MDS is promising, primary and secondary resistance remain a problem. Investigations of combinations of decitabine with other agents, including histone deacetylase inhibitors, are currently ongoing in the hope of substantially prolonging survival in patients with hematologic malignancies.
Collapse
Affiliation(s)
- Elizabeth R Plimack
- Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | | | | |
Collapse
|
17
|
Murphy BC, O'Reilly RL, Singh SM. DNA methylation and mRNA expression of SYN III, a candidate gene for schizophrenia. BMC MEDICAL GENETICS 2008; 9:115. [PMID: 19102774 PMCID: PMC2630979 DOI: 10.1186/1471-2350-9-115] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2008] [Accepted: 12/22/2008] [Indexed: 01/26/2023]
Abstract
BACKGROUND The synapsin III (SYN III) gene on chromosome 22q is a candidate gene for schizophrenia susceptibility due to its chromosome location, neurological function, expression patterns and functional polymorphisms. METHODS This research has established the mRNA expression of SYN III in 22 adult human brain regions as well as the methylation specificity in the closest CpG island of this gene. The methylation specificity studied in 31 brain regions (from a single individual) was also assessed in 51 human blood samples (representing 20 people affected with schizophrenia and 31 normal controls) including a pair of monozygotic twin discordant for schizophrenia and 2 non-human primates. RESULTS The results show that the cytosine methylation in this genomic region is 1) restricted to cytosines in CpG dinucleotides 2) similar in brain regions and blood and 3) appears conserved in primate evolution. Two cytosines (cytosine 8 and 20) localized as the CpG dinucleotide are partially methylated in all brain regions studied. The methylation of these sites in schizophrenia and control blood samples was variable. While cytosine 8 was partially methylated in all samples, the distribution of partial to complete methylation at the cytosine 20 was 22:9 in controls as compared to 18:2 in schizophrenia (p = 0.82). Also, there is no difference in methylation between the affected and unaffected member of a monozygotic twin pair. CONCLUSION The variation in SYN III methylation studied is 1) not related to schizophrenia in the population sample or a monozygotic twin pair discordant for schizophrenia and 2) not related to the mRNA level of SYN IIIa in different human brain regions.
Collapse
Affiliation(s)
- Brenda C Murphy
- Molecular Genetics Unit, Department of Biology and Division of Medical Genetics, The University of Western Ontario, London, Ontario N6A 5B7, Canada.
| | | | | |
Collapse
|
18
|
Grövdal M, Khan R, Aggerholm A, Antunovic P, Astermark J, Bernell P, Engström LM, Kjeldsen L, Linder O, Nilsson L, Olsson A, Wallvik J, Tangen JM, Oberg G, Jacobsen SE, Hokland P, Porwit A, Hellström-Lindberg E. Negative effect of DNA hypermethylation on the outcome of intensive chemotherapy in older patients with high-risk myelodysplastic syndromes and acute myeloid leukemia following myelodysplastic syndrome. Clin Cancer Res 2008; 13:7107-12. [PMID: 18056190 DOI: 10.1158/1078-0432.ccr-07-1193] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Promoter hypermethylation of, for example, tumor-suppressor genes, is considered to be an important step in cancerogenesis and a negative risk factor for survival in patients with myelodysplastic syndromes (MDS); however, its role for response to therapy has not been determined. This study was designed to assess the effect of methylation status on the outcome of conventional induction chemotherapy. EXPERIMENTAL DESIGN Sixty patients with high-risk MDS or acute myeloid leukemia following MDS were treated with standard doses of daunorubicin and 1-beta-d-arabinofuranosylcytosine. Standard prognostic variables and methylation status of the P15(ink4b) (P15), E-cadherin (CDH), and hypermethylated in cancer 1 (HIC) genes were analyzed before treatment. RESULTS Forty percent of the patients achieved complete remission (CR). CR rate was lower in patients with high WBC counts (P = 0.03) and high CD34 expression on bone marrow cells (P = 0.02). Whereas P15 status alone was not significantly associated with CR rate (P = 0.25), no patient with hypermethylation of all three genes achieved CR (P = 0.03). Moreover, patients with CDH methylation showed a significantly lower CR rate (P = 0.008), and CDH methylation retained its prognostic value also in the multivariate analysis. Hypermethylation was associated with increased CD34 expression, but not with other known predictive factors for response, such as cytogenetic profile. CONCLUSIONS We show for the first time a significant effect of methylation status on the outcome of conventional chemotherapy in high-risk MDS and acute myelogenous leukemia following MDS. Provided confirmed in an independent study, our results should be used as a basis for therapeutic decision-making in this patient group.
Collapse
Affiliation(s)
- Michael Grövdal
- Division of Haematology, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Huddinge, Stockholm, Denmark
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Plimack ER, Stewart DJ, Issa JPJ. Combining epigenetic and cytotoxic therapy in the treatment of solid tumors. J Clin Oncol 2007; 25:4519-21. [PMID: 17925545 DOI: 10.1200/jco.2007.12.6029] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
|
20
|
Abstract
Genetic abnormalities of proto-oncogenes and tumor suppressor genes have been demonstrated to be changes that are frequently involved in esophageal cancer pathogenesis. However, hypermethylation of CpG islands, an epigenetic event, is coming more and more into focus in carcinogenesis of the esophagus. Recent studies have proved that promoter hypermethylation of tumor suppressor genes is frequently observed in esophageal carcinomas and seems to play an important role in the pathogenesis of this tumor type. In this review, we will discuss current research on genes that are hypermethylated in human esophageal cancer and precancerous lesions of the esophagus. We will also discuss the potential use of hypermethylated genes as targets for detection, prognosis and treatment of esophageal cancer.
Collapse
Affiliation(s)
- Da-Long Wu
- Department of Pharmacology, School of Medicine, College of Jiaxing, Jiaxing 314001, Zhejiang Province, China.
| | | | | | | |
Collapse
|
21
|
Brakensiek K, Wingen LU, Länger F, Kreipe H, Lehmann U. Quantitative high-resolution CpG island mapping with Pyrosequencing reveals disease-specific methylation patterns of the CDKN2B gene in myelodysplastic syndrome and myeloid leukemia. Clin Chem 2006; 53:17-23. [PMID: 17095538 DOI: 10.1373/clinchem.2007.072629] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Gene silencing through aberrant CpG island methylation is the most extensively analyzed epigenetic event in human tumorigenesis and has huge diagnostic and prognostic potential. Methylation patterns are often very heterogeneous, however, presenting a serious challenge for the development of methylation assays for diagnostic purposes. METHODS We used Pyrosequencing technology to determine the methylation status of 68 CpG sites in the CpG island of the CDKN2B gene [cyclin-dependent kinase inhibitor 2B (p15, inhibits CDK4)], frequently hypermethylated in myeloid malignancies, in a series of bone marrow samples from patients with myelodysplasia and myeloid leukemia (n = 82) and from 32 controls. A total of 7762 individual methylation sites were quantitatively evaluated. Precision and reproducibility of the quantification was evaluated with several overlapping primers. RESULTS The use of optimized sequencing primers and the new Pyro Q-CpG software enabled precise and reproducible quantification with a single sequencing primer of up to 15 CpG sites distributed over approximately 100 bp. Extensive statistical analyses of the whole CpG island revealed for the first time disease-specific methylation patterns of the CDKN2B gene in myeloid malignancies and small regions of differential methylation with high discriminatory power that enabled differentiation of even low-grade myelodysplastic syndrome samples from the controls, a result that was confirmed in an independent group of 9 control and 36 patient samples. CONCLUSION The precise quantitative methylation mapping of whole CpG islands is now possible with Pyrosequencing software in combination with optimized sequencing primers. This method reveals disease-specific methylation patterns and enables the development of specific diagnostic assays.
Collapse
Affiliation(s)
- Kai Brakensiek
- Institute of Pathology, Medizinische Hochschule Hannover, Hannover, Germany
| | | | | | | | | |
Collapse
|
22
|
Guo Y, Engelhardt M, Wider D, Abdelkarim M, Lübbert M. Effects of 5-aza-2'-deoxycytidine on proliferation, differentiation and p15/INK4b regulation of human hematopoietic progenitor cells. Leukemia 2006; 20:115-21. [PMID: 16307025 DOI: 10.1038/sj.leu.2404019] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The demethylating agents 5-azacytidine and 5-aza-2'-deoxycytidine (DAC) have been shown to induce differentiation and inhibit growth of leukemic myeloid cells at low concentrations. However, the effect of DAC in changing the differentiation and proliferation behavior of normal human myeloid progenitors has rarely been investigated. Therefore, we established an in vitro model of normal hematopoietic differentiation, using CD34+ cells from mobilized peripheral blood, to study proliferation and colony formation, expression of several myeloid maturation markers and of the inhibitor of cyclin-dependent kinases p15/INK4b. Upon DAC treatment, cell growth was significantly decreased in a dose-dependent manner, without an increase in cytotoxicity. DAC treatment also resulted in a substantial increase of lysozyme-positive cells, which could be enhanced by G-CSF, a modest increase of myeloperoxidase+ and CD15+ cells, as well as an increase of colony-forming cells (CFU-GM) compared to control cells. p15/INK4b protein expression was strongly upregulated upon myeloid maturation, and additional DAC treatment did not change p15 expression or the methylation status of the p15 promoter at the noncytotoxic concentrations used. Taken together, these data indicate a role of DAC in changing myeloid progenitor cell expansion and differentiation. This model appears suitable also for global analyses of multiple differentially methylated genes.
Collapse
Affiliation(s)
- Y Guo
- Department of Hematology/Oncology, University of Freiburg Medical Center, Freiburg, Germany
| | | | | | | | | |
Collapse
|
23
|
Kovalenko TF, Vaniusheva OV, Shilov IA, Sosin DV, Sukhoverkhova AS, Kozlova TV, Bokarev IN, Sorokina AV, Ozolinia LA, Patrushev LI. [Promoters of genes MTHFR from patients with hyperhomocysteinemia and PTEN from patients with malignant and benign endometrial and ovarian tumors]. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2006; 32:414-23. [PMID: 16909866 DOI: 10.1134/s106816200604008x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Mutational changes in the promoter regions of MTHFR genes from patients with hyperhomocysteinemia and PTEN genes from patients with endometrial and ovarian tumors were studied. An increased level of homocysteine was found in a part of the patients with a heterozygous C677T mutation in the MTHFR gene, although a moderate hyperhomocysteinemia is usually associated with homozygous mutation. We hypothesized that, in this case, the allele lacking the C677T mutation may be inactivated by the promoter mutation. The sequencing of both DNA strands of the minimal promoter region of the MTHFR gene in ten patients did not reveal any mutation, which implied another mechanism of the development of hyperhomocysteinemia in these patients. A PCR analysis of the minimal promoter region of the tumor suppressor PTEN in the presence of 2-pyrrolidone in 101 patients from Moscow clinics revealed changes in it in patients with endometrial (56%) or ovarian (29%) cancer, as well as in patients with endometrial hyperplasia and benign ovarian tumors (34.6 and 29%, respectively). It was presumed that the found PTEN gene promoters may arise from epigenetic alterations (erroneous methylation) or may (more rarely) be induced by mutations. As a result of the studies, new molecular markers associated with endometrial and ovarian tumors were revealed and a simple and effective method of detection of these markers was developed.
Collapse
|
24
|
Brakensiek K, Länger F, Kreipe H, Lehmann U. Absence of p21CIP1, p27KIP1 and p57KIP2 methylation in MDS and AML. Leuk Res 2005; 29:1357-60. [PMID: 15936816 DOI: 10.1016/j.leukres.2005.04.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2005] [Accepted: 04/14/2005] [Indexed: 11/23/2022]
Abstract
Transcriptional silencing of tumour suppressor genes (TSG) due to hypermethylation is a common event in human tumours. The three members of the KIP/CIP family of cyclin dependent kinase inhibitors (CDKIs), p21(CIP 1), p27(KIP 1), and p 57(KIP 2), play key roles in cell cycle regulation, but little is known about their methylation in myeloid neoplasia. Therefore, we analysed 9 haematopoietic cell lines, 67 myelodysplastic syndrome (MDS) and 26 acute myeloid leukaemia (AML) cases as well as 11 controls. p 57(KIP 2) hypermethylation was found in 4/9 cell lines, but methylation of p21(CIP 1) and p27(KIP 1) was infrequent. All patient samples analysed were methylation-negative for these three genes.
Collapse
Affiliation(s)
- Kai Brakensiek
- Institute of Pathology, Medizinische Hochschule Hannover, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany
| | | | | | | |
Collapse
|
25
|
Betz JL, Behairy AS, Rabionet P, Tirtorahardjo B, Moore MW, Cotter PD. Acquired inv(9): what is its significance? ACTA ACUST UNITED AC 2005; 160:76-8. [PMID: 15949575 DOI: 10.1016/j.cancergencyto.2004.12.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2004] [Revised: 11/26/2004] [Accepted: 12/02/2004] [Indexed: 10/25/2022]
Abstract
Pericentric inversion of the heterochromatic region of chromosome 9 [inv(9)] is a common heteromorphism in the general population. It is presumed familial as there are no reports of de novo inv(9) chromosomes in constitutional karyotypes. We report 2 cases of acquired inv(9) chromosomes; 1 patient with acute myeloid leukemia, 46,XY,inv(9)(p11q13)[11]/46,XY[9], and a second with severe anemia, 46,XX,inv(9)(p11q13)[14]/46,XX[6]. The acquired nature of the inv(9) was confirmed by constitutional karyotyping and/or molecular analysis. The inv(9) in these patients may be a de novo inversion that cytogenetically mimics the constitutional inv(9) heteromorphism. Alternatively, it may be the result of neocentromere activation in 9q due to epigenetic events associated with the disease in these patients that results in a metacentric chromosome similarly mimicking the constitutional inv(9). One previous report of an acquired inv(9) was in a patient with essential thrombocythemia. The differences in clinical presentation may represent different underlying mechanisms generating the inv(9). The significance of an acquired inv(9) is unknown and will require reporting of additional cases.
Collapse
Affiliation(s)
- Jaime L Betz
- Division of Genetics, US Labs Inc., Irvine, CA, USA
| | | | | | | | | | | |
Collapse
|
26
|
Brakensiek K, Länger F, Schlegelberger B, Kreipe H, Lehmann U. Hypermethylation of the suppressor of cytokine signalling-1 (SOCS-1) in myelodysplastic syndrome. Br J Haematol 2005; 130:209-17. [PMID: 16029449 DOI: 10.1111/j.1365-2141.2005.05590.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Transcriptional silencing because of hypermethylation is now recognised to be a hallmark of human tumours. In contrast to acute myeloid leukaemia (AML), comparably little is known about aberrant methylation in myelodysplastic syndrome (MDS), a heterogeneous clonal stem cell disorder with a risk of transformation into secondary AML of up to 30%. Recent evidence demonstrates that suppressor of cytokine signalling SOCS-1, a negative regulator of cytokine pathways, may act as a tumour suppressor gene, and inactivation because of hypermethylation was shown in various malignancies. Employing a newly developed quantitative real-time polymerase chain reaction-based methylation assay we analysed, for the first time, SOCS-1 methylation in MDS and found disease-specific hypermethylation in 27 of 86 MDS patients (31%). Demethylation experiments provided direct evidence that aberrant methylation of SOCS-1 induces transcriptional silencing in myeloid cells. In addition, by analysing the expression of signal transducers and activators of transcription (STAT)-induced genes we provide for the first time evidence that the activity of the Janus kinase/STAT pathway is increased in primary patient samples showing SOCS-1 hypermethylation.
Collapse
Affiliation(s)
- Kai Brakensiek
- Institute of Pathology, Medizinische Hochschule Hannover, Hannover, Germany
| | | | | | | | | |
Collapse
|
27
|
Hackanson B, Guo Y, Lübbert M. The silence of the genes: epigenetic disturbances in haematopoietic malignancies. Expert Opin Ther Targets 2005; 9:45-61. [PMID: 15757481 DOI: 10.1517/14728222.9.1.45] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Cancer-associated disturbances of regulated DNA methylation include both global hypomethylation and gene-specific (often even cancer-specific) hypermethylation. Both coexist and have become the subject of intense investigation. In haematological neoplasias, distinct sets of genes, including the p15/INK4b cell cycle inhibitor (mostly in myeloid malignancies) as well as p16/INK4a (only very infrequently in myeloid neoplasia), have been well characterised as to incidence of hypermethylation, concurrent gene inactivation and their re-expression following treatment with DNA methylation inhibitors. Several genes frequently methylated in haematological neoplasias have been studied with respect to their prognostic value. With the advance of low-dose schedules of demethylating agents (explored particularly in the elderly patient population) the rationale for reverting the 'hyper-methylator phenotype' has also prompted in vivo studies of gene reactivation following this type of treatment. However, ubiquitous surrogate markers for the efficacy of this type of treatment need to be developed. These may include reactivated haemoglobin F (HbF), as demethylating agents can result in clinically meaningful induction of HbF in patients with haemoglobinopathies. Because 'cancer testis antigens', which provide powerful signals for T cell cytotoxic activity on solid tumour cells, are usually silenced in leukaemia but can be reactivated in vitro and in vivo, they provide a rationale for an immuno-modulatory effect of demethylating therapy.
Collapse
Affiliation(s)
- Björn Hackanson
- University of Freiburg Medical Center, Department of Hematology/Oncology, 79106 Freiburg, Germany
| | | | | |
Collapse
|
28
|
Novotny L, Szekeres T. Recent developments in cancer chemotherapy oriented towards new targets. Expert Opin Ther Targets 2005; 9:343-57. [PMID: 15934920 DOI: 10.1517/14728222.9.2.343] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Malignant diseases are one of the major causes of death in the western world. Patients are treated by surgery, radiation and chemotherapy. Chemotherapeutic treatment is used to decrease the tumour burden and to eliminate malignant cells. However, in most cases, resistance against chemotherapy develops. Therefore, there is a permanent need for new additional treatment strategies and chemotherapeutic combination regimens. In the present review article, the authors try to highlight the most promising approaches and summarise a selection of potential targets and compounds which might become alternative treatment options against malignant diseases. Due to the high number of scientific articles and the rapid developments in the area of cancer research, the authors can only mention a few selected targets and treatment options; however, the review focuses on new and notably important targets and compounds.
Collapse
Affiliation(s)
- Ladislav Novotny
- Kuwait University, Faculty of Pharmacy, PO Box 24923, Safat 1311, Kuwait.
| | | |
Collapse
|
29
|
Länger F, Dingemann J, Kreipe H, Lehmann U. Up-regulation of DNA methyltransferases DNMT1, 3A, and 3B in myelodysplastic syndrome. Leuk Res 2005; 29:325-9. [PMID: 15661269 DOI: 10.1016/j.leukres.2004.08.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2004] [Accepted: 08/16/2004] [Indexed: 02/08/2023]
Abstract
Analyzing bone marrow trephines from (myelodysplastic syndrome) MDS patients we show for the first time strong over-expression of the DNA methyltransferases 1, and 3A (and 3B to a much lesser extent) in the myelodysplastic syndrome FAB subtypes refractory anaemia (RA) and refractory anaemia with excess of blasts (RAEB). The increase in mRNA expression was much less pronounced in refractory anaemia with ringed sideroblasts (RARS). Classification according to the new WHO guidelines revealed distinct differences between RCMD and RARS. This elevated mRNA expression most probably contributes to the frequently found aberrant hypermethylation in MDS and might explain the promising clinical response of MDS patients treated with DNMT inhibitors.
Collapse
Affiliation(s)
- Florian Länger
- Institute of Pathology, Medizinische Hochschule Hannover, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | | | | | | |
Collapse
|
30
|
Murphy BC, O'Reilly RL, Singh SM. Site-specific cytosine methylation in S-COMT promoter in 31 brain regions with implications for studies involving schizophrenia. Am J Med Genet B Neuropsychiatr Genet 2005; 133B:37-42. [PMID: 15635661 DOI: 10.1002/ajmg.b.30134] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The catechol-o-methyltransferase (COMT) gene on chromosome 22q11 has been considered a strong candidate gene for schizophrenia (SZ) susceptibility. A functional Val/Met polymorphism in exon 4, with potential to affect COMT activity has been implicated in SZ, but the results remain inconclusive. We hypothesized that the association of COMT gene with SZ is not strictly a genetic alteration but could involve DNA methylation, as an epigenetic alteration. Thus, we chose to examine the cytosine DNA methylation profile of the human COMT promoter regions, which partially overlaps with the MB-COMT coding region and covers a total of 56 cytosines. Our analysis of 31 brain regions and 51 individual blood samples suggests that the cytosine methylation in his region is restricted to the CpG dinucleotides only. Also, the methylation pattern is nearly identical in the brain and blood with few exceptions. One cytosine (#27) is partially methylated in 5 brain regions and another cytosine (#23) is partially methylated in 81 of 82 samples studied. The exception being the blood DNA from a single SZ patient with prominent extreme negative symptoms, which was completely methylated. Interestingly, there was no difference in methylation at these sites in the blood DNA from three pairs of monozygotic twins discordant for SZ. The results support the use of blood DNA in methylation studies and rule out S-COMT promoter methylation as a common cause of SZ. The unique observation of a completely methylated cytosine 23 in one patient with SZ may have the potential to affect COMT mRNA transcription and gene activity, but remains to be evaluated.
Collapse
Affiliation(s)
- Brenda C Murphy
- Molecular Genetics Unit, Department of Biology and Division of Medical Genetics, The University of Western Ontario, London, Ontario, Canada
| | | | | |
Collapse
|
31
|
Rüter B, Wijermans PW, Lübbert M. DNA methylation as a therapeutic target in hematologic disorders: recent results in older patients with myelodysplasia and acute myeloid leukemia. Int J Hematol 2004; 80:128-35. [PMID: 15481440 DOI: 10.1532/ijh97.04094] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
DNA methylation provides a major epigenetic code (besides histone modification) of the lineage- and development-specific genes (such as regulators of differentiation in the hematopoietic lineages) that control expression of normal cells. However, DNA methylation is also involved in malignancies because aberrant methylating gene activity occurs during leukemic transformation. Thus, genes such as tumor suppressor genes, growth-regulatory genes, and adhesion molecules are often silenced in various hematopoietic malignancies by epigenetic inactivation via DNA hypermethylation. This inactivation is frequently seen not only in transformed cell lines but also in primary leukemia cells. Because this defect is amenable to reversion by pharmacologic means, agents that inhibit DNA methylation have been developed to specifically target this hypermethylation defect in leukemia and preleukemia cases. The most clinically advanced agents, the azanucleosides 5-azacytidine and 5-aza-2'-deoxycytidine (decitabine), were discovered more than 25 years ago, when their methylation-inhibitory activities, even at low concentrations, became apparent. Although both of these agents, like cytarabine, had been clinically used until then at high doses, the redevelopment of these agents for low-dose schedules has revealed very interesting clinical activities for treating myelodysplasia (MDS) and acute myeloid leukemia (AML). Because these diseases occur mostly in patients over 60 years of age, low-dose schedules with these compounds provide a very promising approach in such patient groups by virtue of their low nonhematologic toxicity profiles. In the present review, we describe the development of treatments that target DNA hypermethylation in MDS and AML, and clinical results are presented. In addition, pharmacologic DNA demethylation may be viewed as a platform for biological modification of malignant cells to become sensitized (or resensitized) to secondary signals, such as differentiating signals (retinoids, vitamin D3) and hormonal signals (eg, estrogen receptor in breast cancer cells, androgen receptor in prostate cancer cells). Finally, an in vitro synergism between the reactivating potency of demethylating agents and inhibitors of histone deacetylation has been tested in several pilot studies of AML and MDS treatment. Finally, gene reactivation by either group of compounds results in therapeutically meaningful reactivation of fetal hemoglobin in patients with severe hemoglobinopathies, extending the therapeutic range of derepressive epigenetic agents to nonmalignant hematopoietic disorders.
Collapse
Affiliation(s)
- Björn Rüter
- Department of Hematology/Oncology, Albert-Ludwigs- University (ALU) Freiburg, Germany
| | | | | |
Collapse
|