1
|
Beauchamp E, Gamma JM, Cromwell CR, Moussa EW, Pain R, Kostiuk MA, Acevedo-Morantes C, Iyer A, Yap M, Vincent KM, Postovit LM, Julien O, Hubbard BP, Mackey JR, Berthiaume LG. Multiomics analysis identifies oxidative phosphorylation as a cancer vulnerability arising from myristoylation inhibition. J Transl Med 2024; 22:431. [PMID: 38715059 PMCID: PMC11075276 DOI: 10.1186/s12967-024-05150-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 03/31/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND In humans, two ubiquitously expressed N-myristoyltransferases, NMT1 and NMT2, catalyze myristate transfer to proteins to facilitate membrane targeting and signaling. We investigated the expression of NMTs in numerous cancers and found that NMT2 levels are dysregulated by epigenetic suppression, particularly so in hematologic malignancies. This suggests that pharmacological inhibition of the remaining NMT1 could allow for the selective killing of these cells, sparing normal cells with both NMTs. METHODS AND RESULTS Transcriptomic analysis of 1200 NMT inhibitor (NMTI)-treated cancer cell lines revealed that NMTI sensitivity relates not only to NMT2 loss or NMT1 dependency, but also correlates with a myristoylation inhibition sensitivity signature comprising 54 genes (MISS-54) enriched in hematologic cancers as well as testis, brain, lung, ovary, and colon cancers. Because non-myristoylated proteins are degraded by a glycine-specific N-degron, differential proteomics revealed the major impact of abrogating NMT1 genetically using CRISPR/Cas9 in cancer cells was surprisingly to reduce mitochondrial respiratory complex I proteins rather than cell signaling proteins, some of which were also reduced, albeit to a lesser extent. Cancer cell treatments with the first-in-class NMTI PCLX-001 (zelenirstat), which is undergoing human phase 1/2a trials in advanced lymphoma and solid tumors, recapitulated these effects. The most downregulated myristoylated mitochondrial protein was NDUFAF4, a complex I assembly factor. Knockout of NDUFAF4 or in vitro cell treatment with zelenirstat resulted in loss of complex I, oxidative phosphorylation and respiration, which impacted metabolomes. CONCLUSIONS Targeting of both, oxidative phosphorylation and cell signaling partly explains the lethal effects of zelenirstat in select cancer types. While the prognostic value of the sensitivity score MISS-54 remains to be validated in patients, our findings continue to warrant the clinical development of zelenirstat as cancer treatment.
Collapse
Affiliation(s)
| | - Jay M Gamma
- Department of Medicine and Pathology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Christopher R Cromwell
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Eman W Moussa
- Department of Biochemistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Rony Pain
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Morris A Kostiuk
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Claudia Acevedo-Morantes
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Aishwarya Iyer
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Megan Yap
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Krista M Vincent
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Lynne M Postovit
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Olivier Julien
- Department of Biochemistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Basil P Hubbard
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | | | - Luc G Berthiaume
- Pacylex Pharmaceuticals Inc., Edmonton, AB, Canada.
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
2
|
Benegas P, Ziegler B, Dieminger V, Bengió R, Zapata P, Larripa I, Ferri C. Expression of genes potentially involved in loss of response in patients with chronic myeloid leukemia. Gene 2024; 896:148047. [PMID: 38042214 DOI: 10.1016/j.gene.2023.148047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 10/28/2023] [Accepted: 11/28/2023] [Indexed: 12/04/2023]
Abstract
Chronic Myeloid Leukemia (CML) is a hematological malignancy characterized by the presence of the BCR::ABL1 fusion gene, which leads to uncontrolled cell growth and survival. Tyrosine kinase inhibitors (TKIs) have revolutionized the treatment of CML, but a significant proportion of patients develop resistance or lose response to these drugs. Understanding the molecular mechanisms underlying treatment response and resistance is crucial for improving patient outcomes. This study aimed to analyze the expression patterns of genes involved in treatment response and resistance in CML patients receiving TKI therapy. The expression levels of MET, FOXO3, p15, p16, HCK, and FYN genes were examined in CML patients and compared to healthy donors. Gene expression levels were compared between optimal responders (OR) and resistant patients (R) vs. healthy donors (HD). The MET and FOXO3 OR group showed significant differences compared with the HD, (p < 0.0001) and (p = 0.0003), respectively. p15 expression showed significant differences between OR and HD groups (p = 0.0078), while no significant differences were found in p16 expression between the HD groups. FYN showed a statistically significant difference between R vs. HD (p = 0.0157). The results of HCK expression analysis revealed significant differences between OR and HD (p = 0.0041) and between R and HD (p = 0.0026). When we analyzed OR patients with undetectable BCR::ABL1 transcripts, a greater expression of HCK was observed in the R group. These findings suggest that monitoring the expression levels of MET and FOXO3 genes could be valuable in predicting treatment response and relapse in CML patients. Our study provides important insights into the potential use of gene expression analysis as a tool for predicting treatment response and guiding treatment decisions in CML patients. This knowledge may ultimately contribute to the development of personalized treatment strategies to improve patient outcomes in CML.
Collapse
Affiliation(s)
- Paula Benegas
- Universidad Nacional de Misiones, Facultad de Ciencias Exactas Químicas y Naturales, Instituto de Biotecnología Misiones "Dra. María Ebe Reca" (INBIOMIS), Laboratorio de Biotecnología Molecular (BIOTECMOL), Misiones, Argentina; Consejo Nacional de Invetigaciones Cientìficas y Tècnicas (CONICET), Buenos Aires, Argentina
| | - Betiana Ziegler
- Universidad Nacional de Misiones, Facultad de Ciencias Exactas Químicas y Naturales, Instituto de Biotecnología Misiones "Dra. María Ebe Reca" (INBIOMIS), Laboratorio de Biotecnología Molecular (BIOTECMOL), Misiones, Argentina; Laboratorio de Genética Hematológica, IMEX, CONICET-Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Victoria Dieminger
- Universidad Nacional de Misiones, Facultad de Ciencias Exactas Químicas y Naturales, Instituto de Biotecnología Misiones "Dra. María Ebe Reca" (INBIOMIS), Laboratorio de Biotecnología Molecular (BIOTECMOL), Misiones, Argentina
| | - Raquel Bengió
- Departamento de Hemato-oncología, Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Pedro Zapata
- Universidad Nacional de Misiones, Facultad de Ciencias Exactas Químicas y Naturales, Instituto de Biotecnología Misiones "Dra. María Ebe Reca" (INBIOMIS), Laboratorio de Biotecnología Molecular (BIOTECMOL), Misiones, Argentina; Consejo Nacional de Invetigaciones Cientìficas y Tècnicas (CONICET), Buenos Aires, Argentina
| | - Irene Larripa
- Laboratorio de Genética Hematológica, IMEX, CONICET-Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Cristian Ferri
- Universidad Nacional de Misiones, Facultad de Ciencias Exactas Químicas y Naturales, Instituto de Biotecnología Misiones "Dra. María Ebe Reca" (INBIOMIS), Laboratorio de Biotecnología Molecular (BIOTECMOL), Misiones, Argentina.
| |
Collapse
|
3
|
Devi P, Engdahl K, Punga T, Bergqvist A. Next-Generation Sequencing Analysis of CpG Methylation of a Tumor Suppressor Gene SHP-1 Promoter in Stable Cell Lines and HCV-Positive Patients. Viruses 2022; 14:v14112352. [PMID: 36366451 PMCID: PMC9695419 DOI: 10.3390/v14112352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 10/19/2022] [Accepted: 10/22/2022] [Indexed: 02/01/2023] Open
Abstract
Hepatitis C virus (HCV) is the major causative pathogen associated with hepatocellular carcinoma and liver cirrhosis. The main virion component, the Core (C) protein, is involved in multiple aspects of HCV pathology including oncogenesis and immune evasion. In this study, we established a next-generation bisulfite sequencing (NGS-BS) protocol to analyze the CpG methylation profile at the tumor suppressor gene SHP-1 P2 promoter as a model system. Our data show that HCV C protein expression in the immortalized T cells correlated with a specific CpG methylation profile at the SHP-1 P2. The NGS-BS on HCV-positive (HCV+) patient-derived PBMCs revealed a considerably different CpG methylation profile compared to the HCV C protein immortalized T cells. Notably, the CpG methylation profile was very similar in healthy and HCV+ PBMCs, suggesting that the SHP-1 P2 CpG methylation profile is not altered in the HCV+ individuals. Collectively, the NGS-BS is a highly sensitive method that can be used to quantitatively characterize the CpG methylation status at the level of individual CpG position and also allows the characterization of cis-acting effects on epigenetic regulation.
Collapse
Affiliation(s)
- Priya Devi
- Department of Medical Sciences, Uppsala University, SE 75185 Uppsala, Sweden
| | - Katarina Engdahl
- Department of Medical Sciences, Uppsala University, SE 75185 Uppsala, Sweden
| | - Tanel Punga
- Department of Medical Biochemistry and Microbiology, Uppsala University, SE 75123 Uppsala, Sweden
| | - Anders Bergqvist
- Department of Medical Sciences, Uppsala University, SE 75185 Uppsala, Sweden
- Clinical Microbiology and Hospital Infection Control, Uppsala University Hospital, SE 75185 Uppsala, Sweden
- Correspondence: ; Tel.: +46-186113937
| |
Collapse
|
4
|
Zhao Y, Huang T, Wang X, Chen Q, Shen H, Xiong B. Measurement for the Area of Red Blood Cells From Microscopic Images Based on Image Processing Technology and Its Applications in Aplastic Anemia, Megaloblastic Anemia, and Myelodysplastic Syndrome. Front Med (Lausanne) 2022; 8:796920. [PMID: 35145978 PMCID: PMC8822214 DOI: 10.3389/fmed.2021.796920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 12/23/2021] [Indexed: 11/13/2022] Open
Abstract
BackgroundAplastic anemia (AA), megaloblastic anemia (MA), and myelodysplastic syndrome (MDS) were common anemic diseases. Sometimes it was difficult to distinguish patients with these diseases.MethodsIn this article, we proposed one measurement method for the area of red blood cells (RBCs) from microscopic images based on image processing technology and analyzed the differences of the area in 25 patients with AA, 64 patients with MA, and 68 patients with MDS.ResultsThe area of RBCs was 44.19 ± 3.88, 42.09 ± 5.35, 52.87 ± 7.68, and 45.75 ± 8.07 μm2 in normal subjects, patients with AA, MA, and MDS, respectively. The coefficients of variation were 8.78%, 10.05%, 14.53%, and 14.00%, respectively, in these groups. The area of RBCs in patients with MA was significantly higher than normal subjects (p < 0.001). Compared with patients with AA and MDS, the area of RBCs in patients with MA was also significantly higher (p < 0.001). The results of correlation analysis between the area of RBCs and mean corpuscular volume (MCV), mean corpuscular hemoglobin (MCH), MCH concentration (MCHC), and red cell distribution width showed no significant correlations (p > 0.05). The area under the curve (AUC) results of the Receiver Operating Characteristic (ROC) curves of RBCs area were 0.421, 0.580, and 0.850, respectively, in patients with AA (p = 0.337), MDS (p = 0.237), and MA (p < 0.001).ConclusionIdentifying the area of RBCs in peripheral blood smears based on the image processing technology could achieve rapid and efficient diagnostic support for patients with MDS and MA, especially for patients with MA and in combination with MCV. However, a larger sample study is needed to find the cutoff area values.
Collapse
Affiliation(s)
- Yongfeng Zhao
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Department of Hematology, The First Affiliated Hospital of Yangtze University, Jingzhou, China
| | - Tingting Huang
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xian Wang
- Department of Pharmacy, The First Affiliated Hospital of Yangtze University, Jingzhou, China
| | - Qianjun Chen
- National Engineering Research Center for E-Learning, Central China Normal University, Wuhan, China
- The State Key Laboratory of Biocatalysis and Enzyme Engineering of China, College of Life Sciences, Hubei University, Wuhan, China
| | - Hui Shen
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Bei Xiong
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, China
- *Correspondence: Bei Xiong
| |
Collapse
|
5
|
Zhou F, Chen B. Prognostic significance of ferroptosis-related genes and their methylation in AML. Hematology 2021; 26:919-930. [PMID: 34789073 DOI: 10.1080/16078454.2021.1996055] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Ferroptosis involves in the development and therapeutic response of various types of tumors. This study aims to explore ferroptosis-related prognostic genes that could further accurately stratify AML patients. METHODS We investigated the prognosis significance of ferroptosis-related genes in AML by Univariate and multivariate Cox proportional hazards regression analyses. With the methylation data of TCGA samples, we looked for methylation sites associated with prognostic genes and compared the correlation between methylation and mRNA expression. R software and 'edgeR' packages were used to identify the DEGs between the high-and-low-risk groups divided by the FRPGs prognosis model and then run GO enrichment, KEGG pathway, and PPI network. RESULTS We found a prognostic risk model that included AKR1C2 and SOCS1 predicted outcomes in AML patients. Methylation analysis showed that AKR1C2 and SOCS1 are negatively regulated by their methylation, leading to their low expression in AML patients. Besides, both decreased SOCS1 expression and hypermethylation predicted favorable OS and PFS in AML patients. Finally, this prognostic risk model exhibited a close correlation with several clinical features, especially with age (P=0.005), cytogenetic type (P=0.031), risk_cytogenetic (P=0.001), and risk_molecular (P<0.001). Functional enrichment analysis showed that DEGs are most enriched in the regulation of cell death and the PI3K-Akt signaling pathway. CONCLUSION AKR1C2 and SOCS1 are promising biomarkers for predicting prognosis in patients with AML.
Collapse
Affiliation(s)
- Fang Zhou
- Medical School, Southeast University, Nanjing, Jiangsu Province, People's Republic of China
| | - Baoan Chen
- Department of Hematology and Oncology, Zhongda Hospital, Medical School, Southeast University, Nanjing, Jiangsu Province, People's Republic of China
| |
Collapse
|
6
|
Iltar U, Alhan FN, Vural E, Ataş Ü, Sözel H, Doğan Ö, Boduroğlu A, Yücel OK, Salim O, Ündar L. Recurrent arthritis as an unexpected side effect associated with azacitidine in a patient with myelodysplastic syndrome. J Oncol Pharm Pract 2021; 28:500-503. [PMID: 34636655 DOI: 10.1177/10781552211049728] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
INTRODUCTION Hypomethylating agents have confirmed efficacy for myelodysplastic syndrome and acute myeloid leukemia and are widely used. Although arthralgia is common side effect associated with hypomethylating agents, arthritis has not been reported previously. CASE REPORT We present the first recorded patient with arthritis after azacitidine treatment. The patient we presented here had severe cytopenias requiring transfusion with erythrocyte and platelet suspensions, and a complete hematological response was obtained for myelodysplastic syndrome after three cycles of azacitidine (AZA) treatment. However, interestingly, after each AZA treatment cycle, the patient had recurrent attacks of arthritis. MANAGEMENT AND OUTCOMES The episodes of arthritis were possibly acute flares of pre-existing crystal-induced arthritis, as exhibited with azacitidine treatments and were managed effectively with nonsteroidal anti-inflammatory drugs. DISCUSSION Because it is a rare condition, clinicians should not overlook AZA as a possible cause of arthritis exacerbations when arthritis of unknown etiology develops in patients treated with AZA.
Collapse
Affiliation(s)
- Utku Iltar
- Department of Internal Medicine, Division of Hematology, Faculty of Medicine, 37502Akdeniz University, Antalya, Turkey
| | - Fadime Nurcan Alhan
- Department of Internal Medicine, Division of Hematology, Faculty of Medicine, 37502Akdeniz University, Antalya, Turkey
| | - Ece Vural
- Department of Internal Medicine, Division of Hematology, Faculty of Medicine, 37502Akdeniz University, Antalya, Turkey
| | - Ünal Ataş
- Department of Internal Medicine, Division of Hematology, Faculty of Medicine, 37502Akdeniz University, Antalya, Turkey
| | - Hasan Sözel
- Department of Internal Medicine, Faculty of Medicine, 37502Akdeniz University, Antalya, Turkey
| | - Ömer Doğan
- Department of Radiology, Faculty of Medicine, 37502Akdeniz University, Antalya, Turkey
| | - Ahmet Boduroğlu
- Department of Pathology, Faculty of Medicine, 37502Akdeniz University, Antalya, Turkey
| | - Orhan Kemal Yücel
- Department of Internal Medicine, Division of Hematology, Faculty of Medicine, 37502Akdeniz University, Antalya, Turkey
| | - Ozan Salim
- Department of Internal Medicine, Division of Hematology, Faculty of Medicine, 37502Akdeniz University, Antalya, Turkey
| | - Levent Ündar
- Department of Internal Medicine, Division of Hematology, Faculty of Medicine, 37502Akdeniz University, Antalya, Turkey
| |
Collapse
|
7
|
Lee P, Yim R, Yung Y, Chu HT, Yip PK, Gill H. Molecular Targeted Therapy and Immunotherapy for Myelodysplastic Syndrome. Int J Mol Sci 2021; 22:10232. [PMID: 34638574 PMCID: PMC8508686 DOI: 10.3390/ijms221910232] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/17/2021] [Accepted: 09/21/2021] [Indexed: 12/22/2022] Open
Abstract
Myelodysplastic syndrome (MDS) is a heterogeneous, clonal hematological disorder characterized by ineffective hematopoiesis, cytopenia, morphologic dysplasia, and predisposition to acute myeloid leukemia (AML). Stem cell genomic instability, microenvironmental aberrations, and somatic mutations contribute to leukemic transformation. The hypomethylating agents (HMAs), azacitidine and decitabine are the standard of care for patients with higher-risk MDS. Although these agents induce responses in up to 40-60% of patients, primary or secondary drug resistance is relatively common. To improve the treatment outcome, combinational therapies comprising HMA with targeted therapy or immunotherapy are being evaluated and are under continuous development. This review provides a comprehensive update of the molecular pathogenesis and immune-dysregulations involved in MDS, mechanisms of resistance to HMA, and strategies to overcome HMA resistance.
Collapse
Affiliation(s)
| | | | | | | | | | - Harinder Gill
- Division of Haematology, Medical Oncology and Haemopoietic Stem Cell Transplantation, Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (P.L.); (R.Y.); (Y.Y.); (H.-T.C.); (P.-K.Y.)
| |
Collapse
|
8
|
Sampath S, Misra P, Yadav SK, Sharma S, Somasundaram V. A study on DNA methylation status in promoter region of p15 gene in patients of acute myeloid leukemia and myelodysplastic syndrome. Med J Armed Forces India 2021; 77:337-342. [PMID: 34305288 PMCID: PMC8282539 DOI: 10.1016/j.mjafi.2021.04.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 04/22/2021] [Indexed: 10/21/2022] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS) are a spectrum of hematological malignancies with a multistep process of accumulated genetic and epigenetic alterations. DNA methylation is most extensively studied epigenetic alteration in malignancies. Recent research studies in the field have brought out translational implications of promoter methylation of tumor suppressor gene p15 in tumors. Therefore, we studied the role of DNA Methylation of p15 gene in AML and MDS. METHODS The study was carried out in 41 consecutive AML/MDS cases reporting to hematological OPD of a tertiary care center along with 25 age and sex-matched healthy controls. The methylation status in the promoter region of the p15 gene was assessed by methylation-specific PCR (MSP) from blood samples after ethical approval and informed consent of the patients and controls. The association of methylation status was studied with clinical presentations, AML subtypes, and cytogenetics using Chi-square test/Fisher's exact test tools. RESULTS A total of 41 cases included in the study comprised 33 cases of AML and 08 cases of MDS with an age range between 06 months and 82 years. Of the 41 cases, 29 revealed promoter methylation of the p15 gene, which compared to healthy controls was found statistically significant (p < 0.001). The methylation status did not significantly correlate with AML subtypes or the cytogenetic abnormalities detected in cases. CONCLUSION The outcome of the study indicates p15 promoter DNA methylation in cases of AML and MDS may identify those individuals who might benefit from the targeted therapeutic approaches.
Collapse
Affiliation(s)
- Sangeetha Sampath
- Professor (Biochemistry), Command Hospital (Air Force), Bengaluru, India
| | - Pratibha Misra
- Professor & Head, Department of Biochemistry, Armed Forces Medical College, Pune, India
| | | | - Sanjeevan Sharma
- Senior Advisor (Medicine & Hematology), Command Hospital (Central Command), Lucknow, India
| | | |
Collapse
|
9
|
Wang XX, Jia HJ, Lv YR, Sun HH, Wei XL, Tan JY, Jing ZZ. A Luciferase-EGFP Reporter System for the Evaluation of DNA Methylation in Mammalian Cells. Mol Biol 2021; 55:742-751. [PMID: 34226765 PMCID: PMC8244672 DOI: 10.1134/s0026893321040099] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 10/22/2020] [Accepted: 11/03/2020] [Indexed: 11/23/2022]
Abstract
DNA methylation is an essential epigenetic modification involved in numerous biological processes. Here, we present a cell-based system pLTR-Luc2P-EGFP for evaluation of DNA methylation in mammalian cells. In this system, the expression of reporter gene luciferase2P (Luc2P)-EGFP is under the control of HIV-1 promoter 5' long terminal repeat (LTR), which contains multiple CpG sites. Once these sites are methylated, the expression of Luc2P-EGFP is turned off, which may be visualized under fluorescence microscopy, with quantification performed in luciferase activity assay. As a proof of principle, pLTR-Luc2P-EGFP was methylated in vitro, and transfected into 293T cells, where the reduction of Luc2P-EGFP expression was confirmed. Premixed reporter DNA samples with the methylation levels varying from 0 to 100% were used for quantitative measurements of DNA methylation. The resulting standard curves indicated the accuracy of luciferase activity exceeding that of the Western blotting against EGFP. The Bland–Altman analysis showed that data from luciferase activity assay were in good agreement with the actual DNA methylation levels. In summary, we have established a reporter system coupled with reliable detection technique capable of efficient quantifying the changes in methylation in mammalian cells. This system may be utilized as a high throughput screening tool for identifying molecules that modulate DNA methylation.
Collapse
Affiliation(s)
- X X Wang
- School of Public Health, Lanzhou University, 730000 Lanzhou, China
| | - H J Jia
- State Key Laboratory of Veterinary of Etiological Biology, Key Laboratory of Veterinary Public Health of Agricultural Ministry, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 730000 Lanzhou, China
| | - Y R Lv
- School of Public Health, Lanzhou University, 730000 Lanzhou, China.,State Key Laboratory of Veterinary of Etiological Biology, Key Laboratory of Veterinary Public Health of Agricultural Ministry, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 730000 Lanzhou, China
| | - H H Sun
- School of Public Health, Lanzhou University, 730000 Lanzhou, China.,State Key Laboratory of Veterinary of Etiological Biology, Key Laboratory of Veterinary Public Health of Agricultural Ministry, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 730000 Lanzhou, China
| | - X L Wei
- Institute of Immunology, School of Basic Medical Sciences, Lanzhou University, 730000 Lanzhou, China
| | - J Y Tan
- Institute of Immunology, School of Basic Medical Sciences, Lanzhou University, 730000 Lanzhou, China
| | - Z Z Jing
- State Key Laboratory of Veterinary of Etiological Biology, Key Laboratory of Veterinary Public Health of Agricultural Ministry, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 730000 Lanzhou, China
| |
Collapse
|
10
|
Brindley EC, Papoin J, Kennedy L, Robledo RF, Ciciotte SL, Kalfa TA, Peters LL, Blanc L. Rasa3 regulates stage-specific cell cycle progression in murine erythropoiesis. Blood Cells Mol Dis 2021; 87:102524. [PMID: 33341069 PMCID: PMC7856249 DOI: 10.1016/j.bcmd.2020.102524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 11/25/2020] [Indexed: 10/22/2022]
Abstract
Inherited bone marrow failure syndromes (IBMFS) are heterogeneous disorders characterized by dysregulated hematopoiesis in various lineages, developmental anomalies, and predisposition to malignancy. The scat (severe combined anemia and thrombocytopenia) mouse model is a model of IBMFS with a phenotype of pancytopenia cycling through crises and remission. Scat carries an autosomal recessive missense mutation in Rasa3 that results in RASA3 mislocalization and loss of function. RASA3 functions as a Ras-GTPase activating protein (GAP), and its loss of function in scat results in increased erythroid RAS activity and reactive oxygen species (ROS) and altered erythroid cell cycle progression, culminating in delayed terminal erythroid differentiation. Here we sought to further resolve the erythroid cell cycle defect in scat through ex vivo flow cytometric analyses. These studies revealed a specific G0/G1 accumulation in scat bone marrow (BM) polychromatophilic erythroblasts and scat BM Ter119-/c-KIT+/CD71lo/med progenitors, with no changes evident in equivalent scat spleen populations. Systematic analyses of RNAseq data from megakaryocyte-erythroid progenitors (MEPs) in scat crisis vs. scat partial remission reveal altered expression of genes involved in the G1-S checkpoint. Together, these data indicate a precise, biphasic role for RASA3 in regulating the cell cycle during erythropoiesis with relevance to hematopoietic disease progression.
Collapse
Affiliation(s)
- Elena C Brindley
- Department of Molecular Medicine and Pediatrics, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA; Laboratory of Developmental Erythropoiesis, Les Nelkin Memorial Laboratory of Pediatric Oncology, Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
| | - Julien Papoin
- Laboratory of Developmental Erythropoiesis, Les Nelkin Memorial Laboratory of Pediatric Oncology, Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
| | - Lauren Kennedy
- Laboratory of Developmental Erythropoiesis, Les Nelkin Memorial Laboratory of Pediatric Oncology, Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
| | | | | | - Theodosia A Kalfa
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 05229, USA
| | | | - Lionel Blanc
- Department of Molecular Medicine and Pediatrics, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA; Laboratory of Developmental Erythropoiesis, Les Nelkin Memorial Laboratory of Pediatric Oncology, Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA.
| |
Collapse
|
11
|
Fenaux P, Haase D, Santini V, Sanz GF, Platzbecker U, Mey U. Myelodysplastic syndromes: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up †☆. Ann Oncol 2021; 32:142-156. [PMID: 33221366 DOI: 10.1016/j.annonc.2020.11.002] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/30/2020] [Accepted: 11/02/2020] [Indexed: 12/14/2022] Open
Affiliation(s)
- P Fenaux
- Service d'Hématologie Clinique, Groupe Francophone des Myélodysplasies (GFM), Department of Hematology, Hôpital St. Louis (Assistance Publique, Hôpitaux de Paris), Paris, France; Paris 7 University, Paris, France
| | - D Haase
- Department of Hematology and Medical Oncology, University Medical Center, Göttingen, Germany
| | - V Santini
- MDS Unit, Haematology, AOU Careggi, University of Florence, Florence, Italy
| | - G F Sanz
- Department of Haematology, Hospital Universitario La Fe, Valencia, Spain; Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - U Platzbecker
- Department of Hematology and Cellular Therapy, Medical Clinic and Policlinic 1, University Hospital Leipzig, Germany
| | - U Mey
- Department of Oncology and Haematology, Kantonsspital Graubuenden, Chur, Switzerland
| |
Collapse
|
12
|
Borchiellini M, Ummarino S, Di Ruscio A. The Bright and Dark Side of DNA Methylation: A Matter of Balance. Cells 2019; 8:cells8101243. [PMID: 31614870 PMCID: PMC6830319 DOI: 10.3390/cells8101243] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 10/06/2019] [Accepted: 10/10/2019] [Indexed: 12/11/2022] Open
Abstract
DNA methylation controls several cellular processes, from early development to old age, including biological responses to endogenous or exogenous stimuli contributing to disease transition. As a result, minimal DNA methylation changes during developmental stages drive severe phenotypes, as observed in germ-line imprinting disorders, while genome-wide alterations occurring in somatic cells are linked to cancer onset and progression. By summarizing the molecular events governing DNA methylation, we focus on the methods that have facilitated mapping and understanding of this epigenetic mark in healthy conditions and diseases. Overall, we review the bright (health-related) and dark (disease-related) side of DNA methylation changes, outlining how bulk and single-cell genomic analyses are moving toward the identification of new molecular targets and driving the development of more specific and less toxic demethylating agents.
Collapse
Affiliation(s)
- Marta Borchiellini
- Department of Health Sciences, University of Eastern Piedmont, 28100 Novara, Italy.
- Department of Translational Medicine, University of Eastern Piedmont, 28100 Novara, Italy.
| | - Simone Ummarino
- Harvard Medical School Initiative for RNA Medicine, Harvard Medical School, Boston, MA 02115, USA.
| | - Annalisa Di Ruscio
- Department of Translational Medicine, University of Eastern Piedmont, 28100 Novara, Italy.
- Harvard Medical School Initiative for RNA Medicine, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
13
|
Nejati R, Wei S, Uzzo RG, Poureghbali S, Pei J, Talarchek JN, Ruth K, Dulaimi E, Kutikov A, Testa JR, Al-Saleem T. Monosomy of Chromosome 9 Is Associated With Higher Grade, Advanced Stage, and Adverse Outcome in Clear-cell Renal Cell Carcinoma. Clin Genitourin Cancer 2019; 18:56-61. [PMID: 31648964 DOI: 10.1016/j.clgc.2019.09.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/21/2019] [Accepted: 09/10/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND Clear-cell renal cell carcinoma (ccRCC) is one of the most common malignancies in humans and is usually associated with poor outcomes. Cancers are considered to be genetic diseases. Therefore, a better understanding of genetic alterations that are related to disease progression or poor prognosis can help to more precisely identify high-risk patients and treat them more effectively. The aim of this study was to examine the frequency of whole chromosome 9 loss (monosomy of chromosome 9) and its prognostic value in patients with ccRCC. MATERIALS AND METHODS Single nucleotide polymorphism-based chromosome microarray (CMA) analysis was performed on 103 resected specimens from patients with ccRCC who had undergone partial or radical nephrectomy between January 2002 and March 2017 at Fox Chase Cancer Center. Monosomy 9 was correlated with clinicopathologic parameters and recurrence-free survival. RESULTS Chromosome 9 loss was detected in 31 (30%) of 103 tumors. Tumors with chromosome 9 loss had higher histologic grade (3 and 4; P < .001) and pathologic stage (P < .001). In 59 patients with non-metastatic ccRCC, chromosome 9 loss was also associated with higher recurrence rate and shorter recurrence-free survival (RFS) (12-month RFS, 77.8%; 95% confidence interval, 36.5%-93.9% for chromosome 9 loss vs. 95.7%; 95% confidence interval, 84.0%-98.9% for no loss; P = .002). CONCLUSIONS Chromosome 9 loss was found in 30% of patients with ccRCC and correlated with higher grade, advanced stage, and shorter RFS in patients with Stage I to III ccRCC.
Collapse
Affiliation(s)
- Reza Nejati
- Department of Pathology, Fox Chase Cancer Center, Temple Health System, Philadelphia, PA.
| | - Shuanzeng Wei
- Department of Pathology, Fox Chase Cancer Center, Temple Health System, Philadelphia, PA
| | - Robert G Uzzo
- Division of Urologic Oncology, Fox Chase Cancer Center, Temple Health System, Philadelphia, PA
| | - Sahar Poureghbali
- Department of Pathology, Fox Chase Cancer Center, Temple Health System, Philadelphia, PA
| | - Jianming Pei
- Genomics Facility, Fox Chase Cancer Center, Temple Health System, Philadelphia, PA
| | | | - Karen Ruth
- Biostatistics and Bioinformatics Facility, Fox Chase Cancer Center, Temple Health System, Philadelphia, PA
| | - Essel Dulaimi
- Department of Pathology, Fox Chase Cancer Center, Temple Health System, Philadelphia, PA
| | - Alexander Kutikov
- Division of Urologic Oncology, Fox Chase Cancer Center, Temple Health System, Philadelphia, PA
| | - Joseph R Testa
- Cancer Biology Program, Fox Chase Cancer Center, Temple Health System, Philadelphia, PA
| | - Tahseen Al-Saleem
- Department of Pathology, Fox Chase Cancer Center, Temple Health System, Philadelphia, PA
| |
Collapse
|
14
|
Sharma ND, Nickl CK, Kang H, Ornatowski W, Brown R, Ness SA, Loh ML, Mullighan CG, Winter SS, Hunger SP, Cannon JL, Matlawska‐Wasowska K. Epigenetic silencing of SOCS5 potentiates JAK-STAT signaling and progression of T-cell acute lymphoblastic leukemia. Cancer Sci 2019; 110:1931-1946. [PMID: 30974024 PMCID: PMC6549933 DOI: 10.1111/cas.14021] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 04/03/2019] [Accepted: 04/08/2019] [Indexed: 01/12/2023] Open
Abstract
Activating mutations in cytokine receptors and transcriptional regulators govern aberrant signal transduction in T-cell lineage acute lymphoblastic leukemia (T-ALL). However, the roles played by suppressors of cytokine signaling remain incompletely understood. We examined the regulatory roles of suppressor of cytokine signaling 5 (SOCS5) in T-ALL cellular signaling networks and leukemia progression. We found that SOCS5 was differentially expressed in primary T-ALL and its expression levels were lowered in HOXA-deregulated leukemia harboring KMT2A gene rearrangements. Here, we report that SOCS5 expression is epigenetically regulated by DNA methyltransferase-3A-mediated DNA methylation and methyl CpG binding protein-2-mediated histone deacetylation. We show that SOCS5 negatively regulates T-ALL cell growth and cell cycle progression but has no effect on apoptotic cell death. Mechanistically, SOCS5 silencing induces activation of JAK-STAT signaling, and negatively regulates interleukin-7 and interleukin-4 receptors. Using a human T-ALL murine xenograft model, we show that genetic inactivation of SOCS5 accelerates leukemia engraftment and progression, and leukemia burden. We postulate that SOCS5 is epigenetically deregulated in T-ALL and serves as an important regulator of T-ALL cell proliferation and leukemic progression. Our results link aberrant downregulation of SOCS5 expression to the enhanced activation of the JAK-STAT and cytokine receptor-signaling cascade in T-ALL.
Collapse
Affiliation(s)
- Nitesh D. Sharma
- Department of PediatricsUniversity of New Mexico Health Sciences CenterAlbuquerqueNM
| | - Christian K. Nickl
- Department of PediatricsUniversity of New Mexico Health Sciences CenterAlbuquerqueNM
| | - Huining Kang
- Department of Internal MedicineUniversity of New Mexico Comprehensive Cancer CenterAlbuquerqueNM
| | - Wojciech Ornatowski
- Department of PathologyUniversity of New Mexico Comprehensive Cancer CenterAlbuquerqueNM
| | - Roger Brown
- Department of Internal MedicineUniversity of New Mexico Comprehensive Cancer CenterAlbuquerqueNM
| | - Scott A. Ness
- Department of Internal MedicineUniversity of New Mexico Comprehensive Cancer CenterAlbuquerqueNM
| | - Mignon L. Loh
- Department of PediatricsBenioff Children's HospitalUniversity of California at San FranciscoSan FranciscoCA
| | | | - Stuart S. Winter
- Children's Minnesota Research Institute and Cancer and Blood Disorders ProgramChildren's MinnesotaMinneapolisMN
| | - Stephen P. Hunger
- Department of Pediatrics and the Center for Childhood Cancer ResearchChildren's Hospital of PhiladelphiaPerelman School of Medicine at the University of PennsylvaniaPhiladelphiaPA
| | - Judy L. Cannon
- Department of PathologyUniversity of New Mexico Comprehensive Cancer CenterAlbuquerqueNM
- Department of Molecular Genetics and MicrobiologyUniversity of New Mexico Health Sciences CenterAlbuquerqueNMUSA
| | | |
Collapse
|
15
|
Pinel C, Prainsack B, McKevitt C. Markers as mediators: A review and synthesis of epigenetics literature. BIOSOCIETIES 2019; 13:276-303. [PMID: 31105763 PMCID: PMC6520226 DOI: 10.1057/s41292-017-0068-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Epigenetics, the study of the processes that control gene expression without a change in DNA sequence, highlights the importance of environmental factors in gene regulation. This paper maps the terrain of epigenetics and identifies four main research subfields: gene expression; molecular epigenetics; clinical epigenetics and epigenetic epidemiology. Within and across these fields, we analyse of what is conceptualised as environment and demonstrate the variable ways authors understand epigenetics environments. Then, following an analysis of the discursive strategies employed by epigenetics researchers, we demonstrate how authors portray the interactions between genes, epigenetics, and environment as relationships linking the outside (where the environment is located) with the inside (where the genes are located). We argue that authors assign specific roles to each actor: the environment as the active player initiating the relationship, the genes as recipients, and epigenetics as mediators between environment and genes. Framed as mediators, epigenetic markers can be understood as enablers of communication between environment and genome, capable of processing and organising signals so as to regulate the interactions between the actors of epigenetic relationships. This finding complicates the observation by social science scholars that the interactions between environment and genes can be understood through the concept of signal.
Collapse
Affiliation(s)
- Clémence Pinel
- School of Population Sciences and Health Services Research, King’s College London, UK
| | - Barbara Prainsack
- Department of Political Science, University of Vienna, Austria
- Department of Global Health & Social Medicine, King’s College London, UK
| | - Christopher McKevitt
- School of Population Sciences and Health Services Research, King’s College London, UK
| |
Collapse
|
16
|
Herrera-Moreno JF, Medina-Díaz IM, Bernal-Hernández YY, Ramos KS, Alvarado-Cruz I, Quintanilla-Vega B, González-Arias CA, Barrón-Vivanco BS, Rojas-García AE. Modified CDKN2B (p15) and CDKN2A (p16) DNA methylation profiles in urban pesticide applicators. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2019; 26:15124-15135. [PMID: 30924039 DOI: 10.1007/s11356-019-04658-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 02/19/2019] [Indexed: 06/09/2023]
Abstract
Gene-specific changes in DNA methylation by pesticides in occupationally exposed populations have not been studied extensively. Of particular concern are changes in the methylation profile of tumor-suppressor, such as CDKN2B and CDKN2A, genes involved in oncogenesis. The aim of this study was to evaluate the methylation profiles of CDKN2B and CDKN2A genes in urban pesticide applicators and their relationship with occupational exposure to pesticides. A cross-sectional study was conducted in 186 urban pesticide applicators (categorized as high or moderate exposures) and 102 participants without documented occupational exposures to pesticides. Acute and chronic pesticide exposures were evaluated by direct measurement of urinary dialkylphosphates, organophosphate metabolites, and a structured questionnaire, respectively. Anthropometric characteristics, diet, clinical histories, and other variables were estimated through a validated self-reported survey. DNA methylation was determined by pyrosequencing of bisulfite-treated DNA. Decreased DNA methylation of the CDKN2B gene was observed in pesticide-exposed groups compared to the non-exposed group. In addition, increased methylation of the CDKN2A promoter was observed in the moderate-exposure group compared to the non-exposed group. Bivariate analysis showed an association between CDKN2B methylation and pesticide exposure, general characteristics, smoking status, and micronutrients, while changes in CDKN2A methylation were associated with pesticide exposure, sex, educational level, body mass index, smoking status, supplement intake, clinical parameters, and caffeine consumption. These data suggest that pesticide exposure modifies the methylation pattern of CDKN2B and CDKN2A genes and raise important questions about the role that these changes may play in the regulation of cell cycle activities, senescence, and aging.
Collapse
Affiliation(s)
- José Francisco Herrera-Moreno
- Laboratorio de Contaminación y Toxicología Ambiental, Secretaría de Investigación y Posgrado, Universidad Autónoma de Nayarit, 63155, Ciudad de la Cultura s/n. Col. Centro, C.P. 63000, Tepic, Nayarit, Mexico
- Posgrado en Ciencias Biológico Agropecuarias, Unidad Académica de Agricultura, Km. 9 Carretera Tepic-Compostela, Xalisco, Nayarit, Mexico
| | - Irma Martha Medina-Díaz
- Laboratorio de Contaminación y Toxicología Ambiental, Secretaría de Investigación y Posgrado, Universidad Autónoma de Nayarit, 63155, Ciudad de la Cultura s/n. Col. Centro, C.P. 63000, Tepic, Nayarit, Mexico
| | - Yael Yvette Bernal-Hernández
- Laboratorio de Contaminación y Toxicología Ambiental, Secretaría de Investigación y Posgrado, Universidad Autónoma de Nayarit, 63155, Ciudad de la Cultura s/n. Col. Centro, C.P. 63000, Tepic, Nayarit, Mexico
| | - Kenneth S Ramos
- Department of Medicine, Division of Clinical Support and Data Analytics, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - Isabel Alvarado-Cruz
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de México, Mexico
| | - Betzabet Quintanilla-Vega
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de México, Mexico
| | - Cyndia Azucena González-Arias
- Laboratorio de Contaminación y Toxicología Ambiental, Secretaría de Investigación y Posgrado, Universidad Autónoma de Nayarit, 63155, Ciudad de la Cultura s/n. Col. Centro, C.P. 63000, Tepic, Nayarit, Mexico
| | - Briscia Socorro Barrón-Vivanco
- Laboratorio de Contaminación y Toxicología Ambiental, Secretaría de Investigación y Posgrado, Universidad Autónoma de Nayarit, 63155, Ciudad de la Cultura s/n. Col. Centro, C.P. 63000, Tepic, Nayarit, Mexico
| | - Aurora Elizabeth Rojas-García
- Laboratorio de Contaminación y Toxicología Ambiental, Secretaría de Investigación y Posgrado, Universidad Autónoma de Nayarit, 63155, Ciudad de la Cultura s/n. Col. Centro, C.P. 63000, Tepic, Nayarit, Mexico.
| |
Collapse
|
17
|
Poh W, Dilley RL, Moliterno AR, Maciejewski JP, Pratz KW, McDevitt MA, Herman JG. BRCA1 Promoter Methylation Is Linked to Defective Homologous Recombination Repair and Elevated miR-155 to Disrupt Myeloid Differentiation in Myeloid Malignancies. Clin Cancer Res 2019; 25:2513-2522. [PMID: 30692098 DOI: 10.1158/1078-0432.ccr-18-0179] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Revised: 10/04/2018] [Accepted: 01/16/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE Defective homologous recombination (HR) has been reported in multiple myeloid disorders, suggesting a shared dysregulated pathway in these diverse malignancies. Because targeting HR-defective cancers with PARP inhibition (PARPi) has yielded clinical benefit, improved understanding of HR defects is needed to implement this treatment modality. EXPERIMENTAL DESIGN We used an ex vivo irradiation-based assay to evaluate HR repair, HR gene promoter methylation, and mRNA expression in primary myeloid neoplastic cells. In vitro BRCA1 gene silencing was achieved to determine the consequences on HR repair, sensitivity to PARPi, and expression of miR-155, an oncogenic miRNA. RESULTS Impaired HR repair was frequently detected in myeloid neoplasm samples (9/21, 43%) and was linked to promoter methylation-mediated transcriptional repression of BRCA1, which was not observed for other members of the HR pathway (BRCA2, ATM, ATR, FANC-A). In vitro BRCA1 knockdown increased sensitivity to PARP inhibition, and BRCA1 expression is inversely correlated with miR-155 expression, a finding reproduced in vitro with BRCA1 knockdown. Increased miR-155 was associated with PU.1 and SHIP1 repression, known myeloid differentiation factors that are frequently downregulated during leukemic transformation. CONCLUSIONS This study demonstrates frequent defective HR, associated with BRCA1 epigenetic silencing, in a broad range of myeloid neoplasms. The increased prevalence of BRCA1 promoter methylation, resulting in repressed BRCA1, may have an additional role in leukemogenesis by increasing miR-155 expression, which then inhibits transcription factors associated with normal myeloid differentiation. Further study of HR defects may facilitate the identification of HR-defective myeloid neoplasms sensitive to PARPi.
Collapse
Affiliation(s)
- Weijie Poh
- Graduate Program in Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland.,Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
| | - Robert L Dilley
- Division of Hematology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Alison R Moliterno
- Division of Hematology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jaroslaw P Maciejewski
- Translational Hematology and Oncology Research, Cleveland Clinic/Taussig Cancer Institute, Cleveland, Ohio
| | - Keith W Pratz
- Division of Hematological Malignancy, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
| | - Michael A McDevitt
- Division of Hematology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Division of Hematological Malignancy, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
| | - James G Herman
- Graduate Program in Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland. .,Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland.,Division of Hematology/Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania
| |
Collapse
|
18
|
Analysis of the Methylation Status of CpG Sites Within Cancer-Related Genes in Equine Sarcoids. ANNALS OF ANIMAL SCIENCE 2018. [DOI: 10.2478/aoas-2018-0033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Abstract
In the recent years, particular attention was given to the research aimed at optimizing the use of tumour epigenetic markers. One of the best known epigenetic changes associated with the process of carcinogenesis is aberrant DNA methylation. The aim of the present research was to evaluate the methylation profile of genes potentially important in the diagnosis and/or prognosis of equine sarcoids, the most commonly detected skin tumours in Equidae. The methylation status of potential promoter sequences of nine genes: APC, CCND2, CDKN2B, DCC, RARβ, RASSF1, RASSF5, THBS1 and TRPM1, was determined using bisulfite sequencing polymerase chain reaction (BSP-CR). The results of this study did not reveal any changes in the level of DNA methylation in the analysed group of candidate genes between the tumour and healthy tissues. Despite numerous reports describing the aberrant methylation of the promoters of the analysed genes in human cancers, the data obtained did not confirm the existence of such relationships in the examined tumour tissues, which excludes the possibility of using these genes for the diagnosis of the equine sarcoid.
Collapse
|
19
|
Merkerova MD, Remesova H, Krejcik Z, Loudova N, Hrustincova A, Szikszai K, Cermak J, Jonasova A, Belickova M. Relationship between Altered miRNA Expression and DNA Methylation of the DLK1-DIO3 Region in Azacitidine-Treated Patients with Myelodysplastic Syndromes and Acute Myeloid Leukemia with Myelodysplasia-Related Changes. Cells 2018; 7:cells7090138. [PMID: 30223454 PMCID: PMC6162661 DOI: 10.3390/cells7090138] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 09/05/2018] [Accepted: 09/11/2018] [Indexed: 12/22/2022] Open
Abstract
The DLK1–DIO3 region contains a large miRNA cluster, the overexpression of which has previously been associated with myelodysplastic syndromes (MDS). To reveal whether this overexpression is epigenetically regulated, we performed an integrative analysis of miRNA/mRNA expression and DNA methylation of the regulatory sequences in the region (promoter of the MEG3 gene) in CD34+ bone marrow cells from the patients with higher-risk MDS and acute myeloid leukemia with myelodysplasia-related changes (AML-MRC), before and during hypomethylating therapy with azacytidine (AZA). Before treatment, 50% of patients showed significant miRNA/mRNA overexpression in conjunction with a diagnosis of AML-MRC. Importantly, increased level of MEG3 was associated with poor outcome. After AZA treatment, the expression levels were reduced and were closer to those seen in the healthy controls. In half of the patients, we observed significant hypermethylation in a region preceding the MEG3 gene that negatively correlated with expression. Interestingly, this hypermethylation (when found before treatment) was associated with longer progression-free survival after therapy initiation. However, neither expression nor methylation status were associated with future responsiveness to AZA treatment. In conclusion, we correlated expression and methylation changes in the DLK1–DIO3 region, and we propose a complex model for regulation of this region in myelodysplasia.
Collapse
Affiliation(s)
| | - Hana Remesova
- Institute of Hematology and Blood Transfusion, U Nemocnice 1, 128 20 Prague 2, Czech Republic.
| | - Zdenek Krejcik
- Institute of Hematology and Blood Transfusion, U Nemocnice 1, 128 20 Prague 2, Czech Republic.
| | - Nikoleta Loudova
- Institute of Hematology and Blood Transfusion, U Nemocnice 1, 128 20 Prague 2, Czech Republic.
| | - Andrea Hrustincova
- Institute of Hematology and Blood Transfusion, U Nemocnice 1, 128 20 Prague 2, Czech Republic.
| | - Katarina Szikszai
- Institute of Hematology and Blood Transfusion, U Nemocnice 1, 128 20 Prague 2, Czech Republic.
| | - Jaroslav Cermak
- Institute of Hematology and Blood Transfusion, U Nemocnice 1, 128 20 Prague 2, Czech Republic.
| | - Anna Jonasova
- General University Hospital, 128 08 Prague, Czech Republic.
| | - Monika Belickova
- Institute of Hematology and Blood Transfusion, U Nemocnice 1, 128 20 Prague 2, Czech Republic.
| |
Collapse
|
20
|
Alteration of Epigenetic Regulation by Long Noncoding RNAs in Cancer. Int J Mol Sci 2018; 19:ijms19020570. [PMID: 29443889 PMCID: PMC5855792 DOI: 10.3390/ijms19020570] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 02/13/2018] [Accepted: 02/13/2018] [Indexed: 02/06/2023] Open
Abstract
Long noncoding RNAs (lncRNAs) are important regulators of the epigenetic status of the human genome. Besides their participation to normal physiology, lncRNA expression and function have been already associated to many diseases, including cancer. By interacting with epigenetic regulators and by controlling chromatin topology, their misregulation may result in an aberrant regulation of gene expression that may contribute to tumorigenesis. Here, we review the functional role and mechanisms of action of lncRNAs implicated in the aberrant epigenetic regulation that has characterized cancer development and progression.
Collapse
|
21
|
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: 9] [Impact Index Per Article: 1.5] [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
|
22
|
Behzad MM, Shahrabi S, Jaseb K, Bertacchini J, Ketabchi N, Saki N. Aberrant DNA Methylation in Chronic Myeloid Leukemia: Cell Fate Control, Prognosis, and Therapeutic Response. Biochem Genet 2018; 56:149-175. [DOI: 10.1007/s10528-018-9841-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 01/18/2018] [Indexed: 01/24/2023]
|
23
|
Chaber R, Gurgul A, Wróbel G, Haus O, Tomoń A, Kowalczyk J, Szmatoła T, Jasielczuk I, Rybka B, Ryczan-Krawczyk R, Duszeńko E, Stąpor S, Ciebiera K, Paszek S, Potocka N, Arthur CJ, Zawlik I. Whole-genome DNA methylation characteristics in pediatric precursor B cell acute lymphoblastic leukemia (BCP ALL). PLoS One 2017; 12:e0187422. [PMID: 29125853 PMCID: PMC5695275 DOI: 10.1371/journal.pone.0187422] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 10/19/2017] [Indexed: 11/21/2022] Open
Abstract
In addition to genetic alterations, epigenetic abnormalities have been shown to underlie the pathogenesis of acute lymphoblastic leukemia (ALL)—the most common pediatric cancer. The purpose of this study was to characterize the whole genome DNA methylation profile in children with precursor B-cell ALL (BCP ALL) and to compare this profile with methylation observed in normal bone marrow samples. Additional efforts were made to correlate the observed methylation patterns with selected clinical features. We assessed DNA methylation from bone marrow samples obtained from 38 children with BCP ALL at the time of diagnosis along with 4 samples of normal bone marrow cells as controls using Infinium MethylationEPIC BeadChip Array. Patients were diagnosed and stratified into prognosis groups according to the BFM ALL IC 2009 protocol. The analysis of differentially methylated sites across the genome as well as promoter methylation profiles allowed clear separation of the leukemic and control samples into two clusters. 86.6% of the promoter-associated differentially methylated sites were hypermethylated in BCP ALL. Seven sites were found to correlate with the BFM ALL IC 2009 high risk group. Amongst these, one was located within the gene body of the MBP gene and another was within the promoter region- PSMF1 gene. Differentially methylated sites that were significantly related with subsets of patients with ETV6-RUNX1 fusion and hyperdiploidy. The analyzed translocations and change of genes’ sequence context does not affect methylation and methylation seems not to be a mechanism for the regulation of expression of the resulting fusion genes.
Collapse
Affiliation(s)
- Radosław Chaber
- Institute of Nursing and Health Sciences, Faculty of Medicine, University of Rzeszow, Rzeszow, Poland
| | - Artur Gurgul
- National Research Institute of Animal Production, Laboratory of Genomics, Balice, Poland
| | - Grażyna Wróbel
- Department of Paediatric Bone Marrow Transplantation, Oncology and Hematology, Medical University of Wroclaw, Wroclaw, Poland
| | - Olga Haus
- Department of Clinical Genetics, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Bydgoszcz, Nicolaus Copernicus University in Torun, Torun, Poland
| | - Anna Tomoń
- Institute of Nursing and Health Sciences, Faculty of Medicine, University of Rzeszow, Rzeszow, Poland
| | - Jerzy Kowalczyk
- Department of Pediatric, Hematology, Oncology and Bone Marrow Transplantation, Medical University of Lublin, Lublin, Poland
| | - Tomasz Szmatoła
- National Research Institute of Animal Production, Laboratory of Genomics, Balice, Poland
| | - Igor Jasielczuk
- National Research Institute of Animal Production, Laboratory of Genomics, Balice, Poland
| | - Blanka Rybka
- Department of Paediatric Bone Marrow Transplantation, Oncology and Hematology, Medical University of Wroclaw, Wroclaw, Poland
| | - Renata Ryczan-Krawczyk
- Department of Paediatric Bone Marrow Transplantation, Oncology and Hematology, Medical University of Wroclaw, Wroclaw, Poland
| | - Ewa Duszeńko
- Department of Hematology, Medical University, Wroclaw, Poland
| | - Sylwia Stąpor
- Department of Hematology, Medical University, Wroclaw, Poland
| | | | - Sylwia Paszek
- Centre for Innovative Research in Medical and Natural Sciences, Laboratory of Molecular Biology, Faculty of Medicine, University of Rzeszow, Rzeszow, Poland
| | - Natalia Potocka
- Centre for Innovative Research in Medical and Natural Sciences, Laboratory of Molecular Biology, Faculty of Medicine, University of Rzeszow, Rzeszow, Poland
| | | | - Izabela Zawlik
- Centre for Innovative Research in Medical and Natural Sciences, Laboratory of Molecular Biology, Faculty of Medicine, University of Rzeszow, Rzeszow, Poland
- Department of Genetics, Institution of Experimental and Clinical Medicine, University of Rzeszow, Rzeszow, Poland
- * E-mail:
| |
Collapse
|
24
|
Udayakumar AM, Fawaz N, Pathare A, Asraf S, Al-Huneini M, Al-Farsi K, Al-Kindi S, Al-Khabouri M. First Cytogenetic Profile of Omani Patients with de novo Myelodysplastic Syndromes: Comparison with data from Asia, Africa, Europe and North and South America. Sultan Qaboos Univ Med J 2017; 17:e286-e292. [PMID: 29062550 DOI: 10.18295/squmj.2017.17.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 12/01/2016] [Accepted: 04/06/2017] [Indexed: 11/16/2022] Open
Abstract
Clonal cytogenetic abnormalities have been reported among 30-80% of patients with myelodysplastic syndromes (MDS); however, 20-70% of patients with MDS show a normal karyotype that may nevertheless harbour a cryptic genetic alteration. Earlier reports have suggested that the distribution of specific chromosomal aberrations varies among Western and Asian countries, with geographical and ethnic differences in the frequency of specific chromosomal aberrations. This article compared the cytogenetic data of 36 adult Omani patients with MDS to previously reported data from other populations. Differences were noted between the percentages of clonal aberrations and the median age of Omani subjects at presentation in comparison to individuals of different ethnicities and from various geographical locations. To the best of the authors' knowledge, this is the first report to describe the cytogenetic data of patients with MDS from Oman.
Collapse
Affiliation(s)
- Achandira M Udayakumar
- Departments of Genetics, College of Medicine & Health Sciences, Sultan Qaboos University, Muscat, Oman.,Department of Malignancy Cytogenetics & Molecular Pathology, Hammersmith Hospital, Imperial College Healthcare National Health Service Trust, London, UK
| | - Nagla Fawaz
- Departments of Genetics Haematology, College of Medicine & Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Anil Pathare
- Department of Haematology, Sultan Qaboos University Hospital, Muscat, Oman
| | - Shakila Asraf
- Department of Haematology, Sultan Qaboos University Hospital, Muscat, Oman
| | | | - Khalil Al-Farsi
- Department of Haematology, Sultan Qaboos University Hospital, Muscat, Oman
| | - Salam Al-Kindi
- Departments of Genetics Haematology, College of Medicine & Health Sciences, Sultan Qaboos University, Muscat, Oman
| | | |
Collapse
|
25
|
Zhou X, Yuan P, Liu Q, Liu Z. LncRNA MEG3 Regulates Imatinib Resistance in Chronic Myeloid Leukemia via Suppressing MicroRNA-21. Biomol Ther (Seoul) 2017; 25:490-496. [PMID: 28190319 PMCID: PMC5590792 DOI: 10.4062/biomolther.2016.162] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 10/07/2016] [Accepted: 11/15/2016] [Indexed: 12/04/2022] Open
Abstract
Imatinib resistance has become a major clinical problem for chronic myeloid leukemia. The aim of the present study was to investigate the involvement of MEG3, a lncRNA, in imatinib resistance and demonstrate its underlying mechanisms. RNAs were extracted from CML patients’ peripheral blood cells and human leukemic K562 cells, and the expression of MEG3 was measured by RT-qPCR. Cell proliferation and cell apoptosis were evaluated. Western blotting was used to measure the protein expression of several multidrug resistant transporters. Luciferase reporter assay was performed to determine the binding between MEG3 and miR-21. Our results showed that MEG3 was significantly decreased in imatinib-resistant CML patients and imatinib-resistant K562 cells. Overexpression of MEG3 in imatinib-resistant K562 cells markedly decreased cell proliferation, increased cell apoptosis, reversed imatinib resistance, and reduced the expression of MRP1, MDR1, and ABCG2. Interestingly, MEG3 binds to miR-21. MEG3 and miR-21 were negatively correlated in CML patients. In addition, miR-21 mimics reversed the phenotype of MEG3-overexpression in imatinib-resistant K562 cells. Taken together, MEG3 is involved in imatinib resistance in CML and possibly contributes to imatinib resistance through regulating miR-21, and subsequent cell proliferation, apoptosis and expression of multidrug resistant transporters.
Collapse
Affiliation(s)
- Xiangyu Zhou
- Department of Vascular and Thyroid, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Ping Yuan
- Department of Neurology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Qi Liu
- Department of Pediatrics, Nanchong Central Hospital, Nanchong, Sichuan 637000, China
| | - Zhiqiang Liu
- Department of Lymphoma and Myeloma, University of Texas MD Anderson Cancer Center, Houston, Texas 77054, USA
| |
Collapse
|
26
|
Bhattacharya P, Patel TN. Microsatellite Instability and Promoter Hypermethylation of DNA repair genes in Hematologic Malignancies: a forthcoming direction toward diagnostics. ACTA ACUST UNITED AC 2017; 23:77-82. [PMID: 28728506 DOI: 10.1080/10245332.2017.1354428] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVE The objective of our review is to highlight the significance of microsatellite hypervariation in diagnostics of hematologic malignancies. METHODS For the past few decades, extensive experiments in cancer research have explored all the possible pathways and a number of deleterious mutations that either make the tumor suppressor genes (TSGs) dysfunctional or cause the proto-oncogenes to behave abnormally by changing the cellular phenotype hence rendering disease. To prevent the deleterious effects of mutations and to protect the genomic integrity, our system possesses multiple repair mechanisms. DNA Mismatch Repair (MMR) and Direct Reversal of Damage (DRD) are two repair mechanisms which help in removal of faulty base pairs and alkyl adduct formation respectively to avoid long term effects of toxicity, tumorigenesis and mutagenesis. There are nine major MMR genes - MutS homolog (MSH2, MSH3, MSH4, MSH5, MSH6), MutL homolog (MLH1, MLH3), human post-meiotic segregation genes (PMS1, PMS2), and three major damage reversal genes - O6-methylguanine-DNA-methyltransferase (MGMT), ABH2 and DEPC1. RESULTS Any malfunction in DNA repair machinery can cause microsatellite instability (MSI), a form of genomic abnormality with hyper mutable repeats that is directly associated with cancer. Microsatellites are short, repetitive sequences, non-randomly distributed and localized in 3'-UTR (Untranslated Region), introns, coding regions and promoters. Besides MSI, evidence on promoter hypermethylation of selected repair genes also points toward a prominent reason for cancer initiation and progression. CONCLUSION The presence of specific microsatellite marker hyper-mutability and consistent promoter hypermethylation in leukemia or lymphoma can be considered as a part of routine diagnostic test in clinical laboratories.
Collapse
Affiliation(s)
- Priyanjali Bhattacharya
- a Department of Integrative Biology , Vellore Institute of Technology , Vellore , Tamil Nadu , India
| | - Trupti N Patel
- a Department of Integrative Biology , Vellore Institute of Technology , Vellore , Tamil Nadu , India
| |
Collapse
|
27
|
Yang X, Yang L, Dai W, Ye B. Role of p14ARF and p15INK4B promoter methylation in patients with lung cancer: a systematic meta-analysis. Onco Targets Ther 2016; 9:6977-6985. [PMID: 27956841 PMCID: PMC5113922 DOI: 10.2147/ott.s117161] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Background The cyclin-dependent kinase inhibitors p14ARF and p15INK4B are tumor suppressor genes that have been reported to be silenced through promoter methylation in many human cancers. However, the strength of association between p14ARF or p15INK4B promoter methylation and lung cancer remains unclear. Thus, we first determined whether p14ARF and p15INK4B promoter methylation played a key role in the carcinogenesis of lung cancer. Methods Eligible studies were selected from the online electronic databases. The pooled odds ratios or hazard ratios and 95% confidence intervals were calculated and summarized. Results Finally, 12 studies with 625 lung cancer samples and 488 nontumor samples were included under the fixed-effects model. The pooled odds ratio showed that p14ARF promoter methylation was observed to be significantly higher in non-small-cell lung cancer (NSCLC) than in nontumor samples (P<0.001). No significant correlation was found between p15INK4B promoter methylation and lung cancer (P=0.27). Subgroup analysis of ethnicity revealed that p14ARF promoter methylation was significantly related to the risk of NSCLC in Asian and Caucasian populations. Subgroup analysis of sample type demonstrated that p14ARF promoter methylation was correlated with the risk of NSCLC in tissue samples (P<0.001), but not in bronchoalveolar lavage fluid and blood samples. P14ARF promoter methylation from one study was not significantly correlated with overall survival of patients with NSCLC. Promoter methylation of p14ARF and p15INK4B was not correlated with clinicopathological characteristics, such as gender status, smoking status, tumor differentiation, and tumor stage (P>0.05). Conclusion Our findings suggested that p14ARF promoter methylation may play an important role in the carcinogenesis of lung cancer, but not p15INK4B promoter methylation. Promoter methylation of p14ARF and p15INK4B was not associated with clinicopathological parameters. However, more extensive large-scale studies are essential to further validate our study.
Collapse
Affiliation(s)
- Xinmei Yang
- Department of Oncology, the First Affiliated Hospital of Jiaxing University, Jiaxing
| | - Lei Yang
- Cancer Center, The First Hospital of Jilin University, Changchun
| | - Wanrong Dai
- Department of Pharmacy, The First Affiliated Hospital, Zhejiang University, Zhejiang
| | - Bo Ye
- Department of Thoracic Surgery, Hangzhou Red Cross Hospital, Hangzhou, People's Republic of China
| |
Collapse
|
28
|
Aberrant DNA methylation of acute myeloid leukemia and colorectal cancer in a Chinese pedigree with a MLL3 germline mutation. Tumour Biol 2016; 37:12609-12618. [PMID: 27405564 DOI: 10.1007/s13277-016-5130-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 06/29/2016] [Indexed: 12/14/2022] Open
Abstract
Unlike genetic aberrations, epigenetic alterations do not modify the deoxyribonucleic acid (DNA) coding sequence and can be reversed pharmacologically. Identifying a particular epigenetic alteration such as abnormal DNA methylation may provide better understanding of cancers and improve current therapy. In a Chinese pedigree with colorectal carcinoma and acute myeloid leukemia, we examined the genome-wide DNA methylation level of cases and explored the role of methylation in pathogenesis and progression. DNA methylation status in the four cases, which all harbor a MLL3 germline mutation, differed from that of the normal control, and hypermethylation was more prevalent. Also, more CpG sites were hypermethylated in the acute-phase AML patient than in the AML patient in remission. Fifty-nine hyper- or hypomethylated genes were identified as common to all four cases. Genome-wide DNA methylation analysis demonstrated that differentially methylated sites among acute myeloid leukemia and colorectal carcinoma cases and the control were in both promoters (CpG island) and gene body regions (shelf/shore areas). Hypermethylation was more prevalent in cancer cases. The study supports the suggestion that the level of DNA methylation changes in AML progression.
Collapse
|
29
|
Xia Y, Hong Q, Chen X, Ye H, Fang L, Zhou A, Gao Y, Jiang D, Duan S. APC2 and CYP1B1 methylation changes in the bone marrow of acute myeloid leukemia patients during chemotherapy. Exp Ther Med 2016; 12:3047-3052. [PMID: 27882114 DOI: 10.3892/etm.2016.3719] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 08/23/2016] [Indexed: 11/06/2022] Open
Abstract
Aberrant promoter DNA methylation is a major mechanism of leukemogenesis in hematologic malignancies, including acute myeloid leukemia (AML). However, the association between promoter methylation with chemotherapeutic outcomes remains unknown. In the present study, bone marrow samples were collected prior to and following chemotherapy in 30 AML patients. Methylation-specific polymerase chain reaction technology was used to examine the promoter methylation status of adenomatous polyposis col 2 (APC2) and cytochrome P450 family 1 subfamily B polypeptide 1 (CYP1B1). The results revealed no change in the methylation status of the APC2 promoter in patients following various chemotherapy regimens. However, the methylation status of the CYP1B1 promoter changed in response to 6 different chemotherapy regimens. AML patients of the M3 subtype displayed an induction of the CYP1B1 promoter methylation levels more frequently (57.1%) than patients affected by the other subtypes (M1: 33.3%; M2: 12.5%; M4: 16.7%; M5: 0% and M6: 0%). In addition, a higher frequency of male patients (4/13) exhibited modulation of the CYP1B1 promoter methylation status compared with female patients (3/17). Furthermore, of five AML patients with a poor prognosis, two exhibited changes leading to CYP1B1 hypomethylation and two leading to CYP1B1 hypermethylation. By contrast, three other patients exhibited hypermethylation changes along with remission. This may be explained by the different chemotherapy regimens used to treat these patients or by other unknown factors. The present study revealed that CYP1B1 promoter methylation was induced during chemotherapy, whereas the APC2 promoter remained hemimethylated. Furthermore, the changes in CYP1B1 methylation were dependent on the AML subtypes and the gender of the patients.
Collapse
Affiliation(s)
- Yongming Xia
- Department of Hematology, Yuyao People's Hospital, Ningbo University Yangming Affiliated Hospital, Yuyao, Zhejiang 315400, P.R. China
| | - Qingxiao Hong
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Xiaoying Chen
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Huadan Ye
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Lili Fang
- Department of Hematology, Yuyao People's Hospital, Ningbo University Yangming Affiliated Hospital, Yuyao, Zhejiang 315400, P.R. China
| | - Annan Zhou
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Yuting Gao
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Danjie Jiang
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Shiwei Duan
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| |
Collapse
|
30
|
Mai H, Liu X, Chen Y, Li C, Cao L, Chen X, Chen S, Liu G, Wen F. Hypermethylation of p15 gene associated with an inferior poor long-term outcome in childhood acute lymphoblastic leukemia. J Cancer Res Clin Oncol 2016; 142:497-504. [PMID: 26501552 DOI: 10.1007/s00432-015-2063-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 10/19/2015] [Indexed: 12/19/2022]
Abstract
PURPOSE To quantitate methylation of the CpG island of the promoter region of the p15 gene in childhood acute lymphoblastic leukemia (ALL) and explore its effect on prognosis. METHODS We assessed methylation of the CpG island on the p15 gene in bone marrow mononuclear cells in 93 ALL cases and in a control group of 20 children with idiopathic thrombocytopenia (ITP) by restriction enzyme Eco52I digestion combined with polymerase chain reaction techniques. We explored the effect of varying levels of methylation on event-free survival (EFS). RESULTS The mean methylation level was 25 % in de novo ALL and significantly higher than the control group 2 %, P < 0.01). Forty-two percent of cases (39/93) had hypermethylation (level over 10 %). Fifty-seven percent (12/21) and 38 % (27/72) T- and precursor-B ALL patients had hypermethylation (not significant). For all patients, the 8-year EFS was (83 ± 4) %, standard risk (91 ± 4) %, intermediate risk (IR) (82 ± 5) %, and high risk (HR) (43 ± 19) % (χ(2) = 11.58, P < 0.01). Hypermethylation was associated with a lower 8-year EFS (71 ± 7 vs. 91 ± 4 %, P = 0.02) in univariate analyses. CONCLUSIONS Children with ALL have higher levels of p15 CpG island methylation than a control group of children with ITP. Among children with ALL, hypermethylation was associated with inferior EFS. Higher levels of p15 CpG island methylation may be a poor prognostic marker in childhood ALL.
Collapse
|
31
|
Hong Q, Chen X, Ye H, Zhou A, Gao Y, Jiang D, Wu X, Tian B, Chen Y, Wang M, Xie J, Xia Y, Duan S. Association between the methylation status of the MGMT promoter in bone marrow specimens and chemotherapy outcomes of patients with acute myeloid leukemia. Oncol Lett 2016; 11:2851-2856. [PMID: 27073563 DOI: 10.3892/ol.2016.4317] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 02/04/2016] [Indexed: 12/13/2022] Open
Abstract
The O(6)-methylguanine-DNA methyltransferase (MGMT) gene is a tumor suppressor gene that is associated with the risk of developing acute myeloid leukemia (AML). However, the association between the methylation status of the MGMT promoter and the chemotherapeutic outcomes of patients with AML remains unknown. In the present study, 30 bone marrow samples derived from patients with AML were collected prior and subsequent to chemotherapy. The methylation status of the MGMT promoter in the bone marrow specimens was determined by methylation-specific polymerase chain reaction. The results indicated that the methylation status of the MGMT promoter was influenced by different chemotherapeutic regimens. The MGMT methylation status of M4 patients (3 out of 6) were more chemosensitive, compared with that of patients with other AML subtypes (M1, 1 out of 3; M2, 0 out of 8; M3, 3 out of 7; M5, 0 out of 3; and M6, 1 out of 3). Age-based analysis revealed that the group aged ≤60 years (7 out of 24 patients) exhibited more methylation changes than patients aged >60 years (1 out of 6). Male patients (4 out of 13) were more susceptible to chemotherapy-induced methylation changes than female patients (4 out of 17). Thus, the methylation status of the MGMT promoter may serve as a potential biomarker to predict the therapeutic outcomes in male AML patients. However, further studies in larger sample sets are required to confirm the present findings.
Collapse
Affiliation(s)
- Qingxiao Hong
- Department of Biochemistry and Molecular Biology, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Xiaoying Chen
- Department of Biochemistry and Molecular Biology, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Huadan Ye
- Department of Biochemistry and Molecular Biology, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Annan Zhou
- Department of Biochemistry and Molecular Biology, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Yuting Gao
- Department of Biochemistry and Molecular Biology, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Danjie Jiang
- Department of Biochemistry and Molecular Biology, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Xiaodong Wu
- Department of Hematology, Yuyao People's Hospital, Yuyao, Zhejiang 315400, P.R. China
| | - Bingru Tian
- Department of Hematology, Yuyao People's Hospital, Yuyao, Zhejiang 315400, P.R. China
| | - Youfen Chen
- Department of Hematology, Yuyao People's Hospital, Yuyao, Zhejiang 315400, P.R. China
| | - Ming Wang
- Department of Hematology, Yuyao People's Hospital, Yuyao, Zhejiang 315400, P.R. China
| | - Jiping Xie
- Department of Hematology, Yuyao People's Hospital, Yuyao, Zhejiang 315400, P.R. China
| | - Yongming Xia
- Department of Hematology, Yuyao People's Hospital, Yuyao, Zhejiang 315400, P.R. China
| | - Shiwei Duan
- Department of Biochemistry and Molecular Biology, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| |
Collapse
|
32
|
Wilhelm T, Lipka DB, Witte T, Wierzbinska JA, Fluhr S, Helf M, Mücke O, Claus R, Konermann C, Nöllke P, Niemeyer CM, Flotho C, Plass C. Epigenetic silencing of AKAP12 in juvenile myelomonocytic leukemia. Epigenetics 2016; 11:110-9. [PMID: 26891149 DOI: 10.1080/15592294.2016.1145327] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
A-kinase anchor protein 12 (AKAP12) is a regulator of protein kinase A and protein kinase C signaling, acting downstream of RAS. Epigenetic silencing of AKAP12 has been demonstrated in different cancer entities and this has been linked to the process of tumorigenesis. Here, we used quantitative high-resolution DNA methylation measurement by MassARRAY to investigate epigenetic regulation of all three AKAP12 promoters (i.e., α, β, and γ) within a large cohort of juvenile myelomonocytic leukemia (JMML) patient samples. The AKAP12α promoter shows DNA hypermethylation in JMML samples, which is associated with decreased AKAP12α expression. Promoter methylation of AKAP12α correlates with older age at diagnosis, elevated levels of fetal hemoglobin and poor prognosis. In silico screening for transcription factor binding motifs around the sites of most pronounced methylation changes in the AKAP12α promoter revealed highly significant scores for GATA-2/-1 sequence motifs. Both transcription factors are known to be involved in the haematopoietic differentiation process. Methylation of a reporter construct containing this region resulted in strong suppression of AKAP12 promoter activity, suggesting that DNA methylation might be involved in the aberrant silencing of the AKAP12 promoter in JMML. Exposure to DNMT- and HDAC-inhibitors reactivates AKAP12α expression in vitro, which could potentially be a mechanism underlying clinical treatment responses upon demethylating therapy. Together, these data provide evidence for epigenetic silencing of AKAP12α in JMML and further emphasize the importance of dysregulated RAS signaling in JMML pathogenesis.
Collapse
Affiliation(s)
- Thomas Wilhelm
- a Division of Epigenomics and Cancer Risk Factors, German Cancer Research Center , Heidelberg , Germany
| | - Daniel B Lipka
- b Regulation of Cellular Differentiation Group, Division of Epigenomics and Cancer Risk Factors, German Cancer Research Center , Heidelberg , Germany
| | - Tania Witte
- a Division of Epigenomics and Cancer Risk Factors, German Cancer Research Center , Heidelberg , Germany
| | - Justyna A Wierzbinska
- a Division of Epigenomics and Cancer Risk Factors, German Cancer Research Center , Heidelberg , Germany.,b Regulation of Cellular Differentiation Group, Division of Epigenomics and Cancer Risk Factors, German Cancer Research Center , Heidelberg , Germany
| | - Silvia Fluhr
- c Department of Pediatrics and Adolescent Medicine , Division of Pediatric Hematology-Oncology, University of Freiburg Medical Center , Freiburg , Germany.,d Hermann Staudinger Graduate School, University of Freiburg , Freiburg , Germany
| | - Monika Helf
- b Regulation of Cellular Differentiation Group, Division of Epigenomics and Cancer Risk Factors, German Cancer Research Center , Heidelberg , Germany
| | - Oliver Mücke
- a Division of Epigenomics and Cancer Risk Factors, German Cancer Research Center , Heidelberg , Germany.,b Regulation of Cellular Differentiation Group, Division of Epigenomics and Cancer Risk Factors, German Cancer Research Center , Heidelberg , Germany
| | - Rainer Claus
- a Division of Epigenomics and Cancer Risk Factors, German Cancer Research Center , Heidelberg , Germany.,e Department of Medicine , Division of Hematology, Oncology and Stem Cell Transplantation, University of Freiburg Medical Center , Freiburg , Germany
| | - Carolin Konermann
- a Division of Epigenomics and Cancer Risk Factors, German Cancer Research Center , Heidelberg , Germany
| | - Peter Nöllke
- c Department of Pediatrics and Adolescent Medicine , Division of Pediatric Hematology-Oncology, University of Freiburg Medical Center , Freiburg , Germany
| | - Charlotte M Niemeyer
- c Department of Pediatrics and Adolescent Medicine , Division of Pediatric Hematology-Oncology, University of Freiburg Medical Center , Freiburg , Germany.,f German Cancer Consortium (DKTK)
| | - Christian Flotho
- c Department of Pediatrics and Adolescent Medicine , Division of Pediatric Hematology-Oncology, University of Freiburg Medical Center , Freiburg , Germany.,f German Cancer Consortium (DKTK)
| | - Christoph Plass
- a Division of Epigenomics and Cancer Risk Factors, German Cancer Research Center , Heidelberg , Germany.,f German Cancer Consortium (DKTK)
| |
Collapse
|
33
|
Zhang YY, Yao DM, Zhu XW, Zhou JD, Ma JC, Yang J, Wen XM, Guo H, Lin J, Qian J. DNMT3A intragenic hypomethylation is associated with adverse prognosis in acute myeloid leukemia. Leuk Res 2015; 39:1041-7. [DOI: 10.1016/j.leukres.2015.06.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 06/03/2015] [Accepted: 06/25/2015] [Indexed: 10/23/2022]
|
34
|
Li Y, Melnikov AA, Levenson V, Guerra E, Simeone P, Alberti S, Deng Y. A seven-gene CpG-island methylation panel predicts breast cancer progression. BMC Cancer 2015; 15:417. [PMID: 25986046 PMCID: PMC4438505 DOI: 10.1186/s12885-015-1412-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 05/01/2015] [Indexed: 12/31/2022] Open
Abstract
Background DNA methylation regulates gene expression, through the inhibition/activation of gene transcription of methylated/unmethylated genes. Hence, DNA methylation profiling can capture pivotal features of gene expression in cancer tissues from patients at the time of diagnosis. In this work, we analyzed a breast cancer case series, to identify DNA methylation determinants of metastatic versus non-metastatic tumors. Methods CpG-island methylation was evaluated on a 56-gene cancer-specific biomarker microarray in metastatic versus non-metastatic breast cancers in a multi-institutional case series of 123 breast cancer patients. Global statistical modeling and unsupervised hierarchical clustering were applied to identify a multi-gene binary classifier with high sensitivity and specificity. Network analysis was utilized to quantify the connectivity of the identified genes. Results Seven genes (BRCA1, DAPK1, MSH2, CDKN2A, PGR, PRKCDBP, RANKL) were found informative for prognosis of metastatic diffusion and were used to calculate classifier accuracy versus the entire data-set. Individual-gene performances showed sensitivities of 63–79 %, 53–84 % specificities, positive predictive values of 59–83 % and negative predictive values of 63–80 %. When modelled together, these seven genes reached a sensitivity of 93 %, 100 % specificity, a positive predictive value of 100 % and a negative predictive value of 93 %, with high statistical power. Unsupervised hierarchical clustering independently confirmed these findings, in close agreement with the accuracy measurements. Network analyses indicated tight interrelationship between the identified genes, suggesting this to be a functionally-coordinated module, linked to breast cancer progression. Conclusions Our findings identify CpG-island methylation profiles with deep impact on clinical outcome, paving the way for use as novel prognostic assays in clinical settings. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1412-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yan Li
- Rush University Medical Center, 653 W Congress Pkwy, Chicago, IL, 60612, USA.
| | | | - Victor Levenson
- US Biomarkers, Inc, 29 Buckingham Ln., Buffalo Grove, IL, 60089, USA. .,Currently at Center for Translational Research, Catholic Health Initiatives, Englewood, USA.
| | - Emanuela Guerra
- Unit of Cancer Pathology, CeSI, 'G. d'Annunzio' University Foundation, Via L. Polacchi 11, 66100, Chieti, Italy.
| | - Pasquale Simeone
- Unit of Cancer Pathology, CeSI, 'G. d'Annunzio' University Foundation, Via L. Polacchi 11, 66100, Chieti, Italy.
| | - Saverio Alberti
- Unit of Cancer Pathology, CeSI, 'G. d'Annunzio' University Foundation, Via L. Polacchi 11, 66100, Chieti, Italy. .,Department of Neuroscience, Imaging and Clinical Sciences, Unit of Physiology and Physiopathology, 'G. d'Annunzio' University, Via dei Vestini, 66100, Chieti, Italy.
| | - Youping Deng
- Rush University Medical Center, 653 W Congress Pkwy, Chicago, IL, 60612, USA.
| |
Collapse
|
35
|
Jafri M, Wake NC, Ascher DB, Pires DE, Gentle D, Morris MR, Rattenberry E, Simpson MA, Trembath RC, Weber A, Woodward ER, Donaldson A, Blundell TL, Latif F, Maher ER. Germline Mutations in the CDKN2B Tumor Suppressor Gene Predispose to Renal Cell Carcinoma. Cancer Discov 2015; 5:723-9. [DOI: 10.1158/2159-8290.cd-14-1096] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 04/09/2015] [Indexed: 11/16/2022]
|
36
|
Zheng H, Wang J, Zhou J, Wang P, Fu C, Wu D, Sun A, Qiu H, Jin Z, Han Y, Tang X, Ma X. [Retrospective efficacy analysis of decitabine bridging allogeneic hematopoietic stem cell transplantation on the treatment of myelodysplastic syndrome]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2015; 36:121-4. [PMID: 25778887 PMCID: PMC7342152 DOI: 10.3760/cma.j.issn.0253-2727.2015.02.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
目的 评价地西他滨桥接异基因造血干细胞移植(allo-HSCT)治疗骨髓增生异常综合征(MDS)的疗效及安全性。 方法 回顾性分析2010年7月至2013年12月于苏州大学附属第一医院血液科接受allo-HSCT的MDS患者的临床特征及疗效,随机抽取25例接受地西他滨桥接allo-HSCT的MDS患者为桥接组,以同期33例未接受地西他滨行allo-HSCT的MDS患者为对照组,观察患者疗效、总生存(OS)及移植物抗宿主病(GVHD)发生情况。 结果 桥接组患者移植前骨髓完全缓解率为64.0%(25例中16例),明显高于对照组的15.1%(33例中5例),差异有统计学意义(P<0.05);早期移植相关死亡率低于对照组(4.0%对18.2%),但差异无统计学意义(P=0.106)。桥接组移植相关死亡率及2年OS率分别为12.0%及83.0%,与对照组的30.3%及59.0%比较差异均有统计学意义(P值均<0.05)。桥接组14例患者发生急性GVHD(aGVHD),其中Ⅰ度7例、Ⅱ度3例、Ⅲ度4例;对照组16例患者发生aGVHD,其中Ⅰ度7例、Ⅱ度8例、Ⅲ度1例。 结论 地西他滨桥接allo-HSCT治疗MDS安全且有效。
Collapse
Affiliation(s)
- Huifei Zheng
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Jing Wang
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Jin Zhou
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Panfeng Wang
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Chengcheng Fu
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Depei Wu
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Aining Sun
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Huiying Qiu
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Zhengming Jin
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Yue Han
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Xiaowen Tang
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Xiao Ma
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| |
Collapse
|
37
|
Pellagatti A, Boultwood J. The molecular pathogenesis of the myelodysplastic syndromes. Eur J Haematol 2015; 95:3-15. [DOI: 10.1111/ejh.12515] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/01/2014] [Indexed: 02/07/2023]
Affiliation(s)
- Andrea Pellagatti
- Leukaemia & Lymphoma Research Molecular Haematology Unit; Nuffield Division of Clinical Laboratory Sciences; Radcliffe Department of Medicine; University of Oxford; Oxford UK
| | - Jacqueline Boultwood
- Leukaemia & Lymphoma Research Molecular Haematology Unit; Nuffield Division of Clinical Laboratory Sciences; Radcliffe Department of Medicine; University of Oxford; Oxford UK
| |
Collapse
|
38
|
Prognostic significance of DNMT3A mutations in patients with acute myeloid leukemia. Blood Cells Mol Dis 2015; 54:84-9. [DOI: 10.1016/j.bcmd.2014.07.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Accepted: 07/25/2014] [Indexed: 11/23/2022]
|
39
|
Wang B, Ji K, Wang Y, Li Y, Tang Y, Gu J, Cai L. Exposure to low dose cadmium enhances FL83B cells proliferation through down-regulation of caspase-8 by DNA hypermethylation. Toxicol Res (Camb) 2015. [DOI: 10.1039/c4tx00107a] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Cadmium (Cd) is classified as a human carcinogen probably associated with epigenetic change.
Collapse
Affiliation(s)
- Bo Wang
- Department of Pathology
- The Second Clinical Medical School of Inner Mongolia University for the Nationalities (Inner Mongolia Forestry General Hospital)
- Hulun Buir
- China
- Molecular Pathological Research Institute of Inner Mongolia University for Nationalities
| | - Kun Ji
- Department of Pathophysiology
- Shenyang Medical College
- Shenyang
- China
| | - Yue Wang
- Department of Pathophysiology
- Norman Bethune College of Medicine
- Jilin University
- Changchun
- China
| | - Yang Li
- Department of Pathophysiology
- Norman Bethune College of Medicine
- Jilin University
- Changchun
- China
| | - Yufeng Tang
- Department of Orthopedic trauma
- Shandong Provincial Qianfoshan Hospital
- Shandong University
- Jinan
- China
| | - Junlian Gu
- Department of Pathology
- Shandong Provincial Qianfoshan Hospital
- Shandong University
- Jinan
- China
| | - Lu Cai
- Departments of Pediatrics
- Radiation Oncology and Pharmacology & Toxicology
- University of Louisville
- Louisville
- USA
| |
Collapse
|
40
|
Robaina MCS, Faccion RS, Arruda VO, de Rezende LMM, Vasconcelos GM, Apa AG, Bacchi CE, Klumb CE. Quantitative analysis of CDKN2A methylation, mRNA, and p16(INK4a) protein expression in children and adolescents with Burkitt lymphoma: biological and clinical implications. Leuk Res 2014; 39:248-56. [PMID: 25542698 DOI: 10.1016/j.leukres.2014.11.023] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 11/24/2014] [Accepted: 11/25/2014] [Indexed: 12/22/2022]
Abstract
CDKN2A is a tumor suppressor gene critical in the cell cycle regulation. Little is known regarding the role of CDKN2A methylation in the pathogenesis of Burkitt lymphoma (BL). CDKN2A methylation was investigated using pyrosequencing in 51 tumor samples. p16(INK4a) mRNA and protein levels were measured using real-time PCR and immunohistochemistry, respectively. CDKN2A methylation was detectable in 72% cases. Nuclear expression of p16(INK4a) was not detected in 41% cases. There was an association between methylation and absence of CDKN2A mRNA (P=0.003). In conclusion, CDKN2A methylation occurs at a high frequency suggesting a role in BL pathogenesis and potential therapeutic implications.
Collapse
Affiliation(s)
- Marcela Cristina S Robaina
- Programa de Pesquisa em Hemato-Oncologia Molecular, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | - Roberta Soares Faccion
- Programa de Pesquisa em Hemato-Oncologia Molecular, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | - Viviane Oliveira Arruda
- Programa de Pesquisa em Hemato-Oncologia Molecular, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | | | - Gisele Moledo Vasconcelos
- Programa de Hematologia e Oncologia Pediátrica, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | | | | | - Claudete Esteves Klumb
- Programa de Pesquisa em Hemato-Oncologia Molecular, Instituto Nacional de Câncer, Rio de Janeiro, Brazil.
| |
Collapse
|
41
|
Tao YF, Hu SY, Lu J, Cao L, Zhao WL, Xiao PF, Xu LX, Li ZH, Wang NN, Du XJ, Sun LC, Zhao H, Fang F, Su GH, Li YH, Li YP, Xu YY, Ni J, Wang J, Feng X, Pan J. Zinc finger protein 382 is downregulated by promoter hypermethylation in pediatric acute myeloid leukemia patients. Int J Mol Med 2014; 34:1505-15. [PMID: 25319049 PMCID: PMC4214337 DOI: 10.3892/ijmm.2014.1966] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2014] [Accepted: 10/03/2014] [Indexed: 11/06/2022] Open
Abstract
Acute myeloid leukemia (AML) is the second-most common form of leukemia in children. Aberrant DNA methylation patterns are characteristic of AML. Zinc finger protein 382 (ZNF382) has been suggested to be a tumor suppressor gene possibly regulated by promoter hypermethylation in various types of human cancer. However, ZNF382 expression and methylation status in pediatric AML is unknown. In the present study, ZNF382 transcription levels were evaluated by quantitative reverse-transcription PCR. Methylation status was investigated by methylation-specific (MSP) PCR and bisulfate genomic sequencing (BGS). The prognostic significance of ZNF382 expression and promoter methylation was assessed in 105 cases of pediatric AML. The array data suggested that the ZNF382 promoter was hypermethylated in the AML cases examined. MSP PCR and BGS analysis revealed that ZNF382 was hypermethylated in leukemia cell lines. Furthermore, treatment with 5-aza-2'-deoxycytidine (5-Aza) upregulated ZNF382 expression in the selected leukemia cell lines. The aberrant methylation of ZNF382 was observed in 10% (2/20) of the control samples compared with 26.7% (28/105) of the AML samples. ZNF382 expression was significantly decreased in the 105 AML patients compared with the controls. Patients with ZNF382 methylation showed lower ZNF382 transcript levels compared with patients exhibiting no methylation. There were no significant differences in clinical characteristics or cytogenetic analysis between the patients with or without ZNF382 methylation. ZNF382 methylation correlated with minimal residual disease (MRD). Kaplan-Meier survival analysis revealed similar survival times in the samples with ZNF382 methylation, and multivariate analysis revealed that ZNF382 methylation was not an independent prognostic factor in pediatric AML. The epigenetic inactivation of ZNF382 by promoter hypermethylation can be observed in AML cell lines and pediatric AML samples. Therefore, our study suggests that ZNF382 may be considered a putative tumor suppressor gene in pediatric AML. However, further studies focusing on the mechanisms responsible for ZNF382 downregulation in pediatric leukemia are required.
Collapse
Affiliation(s)
- Yan-Fang Tao
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Shao-Yan Hu
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Jun Lu
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Lan Cao
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Wen-Li Zhao
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Pei-Fang Xiao
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Li-Xiao Xu
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Zhi-Heng Li
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Na-Na Wang
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Xiao-Juan Du
- Department of Gastroenterology, The 5th Hospital of Chinese PLA, Yinchuan, Ningxia, P.R. China
| | - Li-Chao Sun
- Department of Cell and Molecular Biology, Cancer Institute (Hospital), Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, P.R. China
| | - He Zhao
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Fang Fang
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Guang-Hao Su
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Yan-Hong Li
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Yi-Ping Li
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Yun-Yun Xu
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Jian Ni
- Translational Research Center, Second Hospital, The Second Clinical School, Nanjing Medical University, Nanjing, Jiangsu, P.R. China
| | - Jian Wang
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Xing Feng
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Jian Pan
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| |
Collapse
|
42
|
Fenaux P, Haase D, Sanz GF, Santini V, Buske C. Myelodysplastic syndromes: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 2014; 25 Suppl 3:iii57-69. [PMID: 25185242 DOI: 10.1093/annonc/mdu180] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Affiliation(s)
- P Fenaux
- Service d'Hématologie Clinique, Groupe Francophone des Myélodysplasies (GFM), Hôpital St Louis (Assistance Publique, Hôpitaux de Paris) and Paris 7 University, Paris, France
| | - D Haase
- Clinics of Hematology and Medical Oncology, University Medicine, Goettingen, Germany
| | - G F Sanz
- Department of Haematology, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - V Santini
- Functional Unit of Haematology, AOU Careggi, University of Florence, Firenze, Italy
| | - C Buske
- Comprehensive Cancer Center Ulm, Institute of Experimental Cancer Research, University Hospital, Ulm, Germany
| | | |
Collapse
|
43
|
Heinen MM, van den Brandt PA, Schouten LJ, Goldbohm RA, Schouten HC, Verhage BAJ. Dietary one-carbon nutrient intake and risk of lymphoid and myeloid neoplasms: results of the Netherlands cohort study. Cancer Epidemiol Biomarkers Prev 2014; 23:2153-64. [PMID: 25047896 DOI: 10.1158/1055-9965.epi-14-0136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Previous epidemiologic research suggests a protective role of one-carbon nutrients in carcinogenesis. Folate, however, may play a dual role in neoplasms development: protect early in carcinogenesis and promote carcinogenesis at a later stage. We prospectively examined associations between intake of total folate, methionine, riboflavin, vitamin B6, and risk of lymphoid and myeloid neoplasms (including subtypes) and investigated whether alcohol modified the effects of folate. METHODS The Netherlands Cohort Study consists of 120,852 individuals who completed a baseline questionnaire in 1986, including a 150-item food-frequency questionnaire. After 17.3 years of follow-up, 1,280 cases of lymphoid and 222 cases of myeloid neoplasms were available for analysis. RESULTS Intakes of folate, methionine, and riboflavin were not associated with lymphoid or myeloid neoplasms. For vitamin B6, a statistically significantly increased myeloid neoplasms risk was observed (highest vs. lowest quintile: HR = 1.87; 95% confidence intervals, 1.08-3.25). When analyzing by lymphoid and myeloid neoplasms subtypes, no clear associations were observed for most subtypes, with just a few increased risks for some subtypes and nutrients. Some risks became nonsignificant after excluding early cases. No interaction between alcohol and folate was observed. CONCLUSIONS We observed a few significant positive associations; however, some of these would be expected to arise due to chance alone. Furthermore, some risks became nonsignificant after excluding early cases. Therefore, we conclude that there is no association between one-carbon nutrient intake and risk of lymphoid and myeloid neoplasms. IMPACT This study contributes substantially to the limited and inconclusive evidence on the association with one-carbon nutrients.
Collapse
Affiliation(s)
- Mirjam M Heinen
- Department of Epidemiology, School for Oncology and Developmental Biology (GROW), Maastricht University Medical Centre+, Maastricht, the Netherlands
| | - Piet A van den Brandt
- Department of Epidemiology, School for Oncology and Developmental Biology (GROW), Maastricht University Medical Centre+, Maastricht, the Netherlands
| | - Leo J Schouten
- Department of Epidemiology, School for Oncology and Developmental Biology (GROW), Maastricht University Medical Centre+, Maastricht, the Netherlands
| | - R Alexandra Goldbohm
- Department of Prevention and Health, TNO Quality of Life, Leiden, the Netherlands
| | - Harry C Schouten
- Division of Hematology, Department of Internal Medicine, School for Oncology and Developmental Biology (GROW), Maastricht University Medical Centre+, Maastricht, the Netherlands
| | - Bas A J Verhage
- Department of Epidemiology, School for Oncology and Developmental Biology (GROW), Maastricht University Medical Centre+, Maastricht, the Netherlands.
| |
Collapse
|
44
|
Immortalization of T-cells is accompanied by gradual changes in CpG methylation resulting in a profile resembling a subset of T-cell leukemias. Neoplasia 2014; 16:606-15. [PMID: 25065939 PMCID: PMC4198827 DOI: 10.1016/j.neo.2014.07.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 07/01/2014] [Accepted: 07/03/2014] [Indexed: 01/10/2023] Open
Abstract
We have previously described gene expression changes during spontaneous immortalization of T-cells, thereby identifying cellular processes important for cell growth crisis escape and unlimited proliferation. Here, we analyze the same model to investigate the role of genome-wide methylation in the immortalization process at different time points pre-crisis and post-crisis using high-resolution arrays. We show that over time in culture there is an overall accumulation of methylation alterations, with preferential increased methylation close to transcription start sites (TSSs), islands, and shore regions. Methylation and gene expression alterations did not correlate for the majority of genes, but for the fraction that correlated, gain of methylation close to TSS was associated with decreased gene expression. Interestingly, the pattern of CpG site methylation observed in immortal T-cell cultures was similar to clinical T-cell acute lymphoblastic leukemia (T-ALL) samples classified as CpG island methylator phenotype positive. These sites were highly overrepresented by polycomb target genes and involved in developmental, cell adhesion, and cell signaling processes. The presence of non-random methylation events in in vitro immortalized T-cell cultures and diagnostic T-ALL samples indicates altered methylation of CpG sites with a possible role in malignant hematopoiesis.
Collapse
|
45
|
Zhang H, Li X, Ge L, Yang J, Sun J, Niu Q. Methylation of CpG island of p14(ARK), p15(INK4b) and p16(INK4a) genes in coke oven workers. Hum Exp Toxicol 2014; 34:191-7. [PMID: 24837742 DOI: 10.1177/0960327114533576] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
To detect the blood genomic DNA methylation in coke oven workers and find a possible early screening index for occupational lung cancer, 74 coke oven workers as the exposed group and 47 water pump workers as the controls were surveyed, and urine samples and peripheral blood mononuclear cells (PBMCs) were collected. Airborne benzo[a]pyrene (B[a]P) levels in workplace and urinary 1-hydroxypyrene (1-OH-Py) levels were determined by high-performance liquid chromatography. DNA damage of PBMCs and the p14(ARK), p15(INK4b) and p16(INK4a) gene CpG island methylation in the promoter region were detected by comet assay and methylation-specific polymerase chain reaction techniques, respectively. Results show that compared with the controls, concentration of airborne B[a]Ps was elevated in the coke plant, and urinary 1-OH-Py's level and DNA olive tail moment in comet assay were significantly increased in the coke oven workers, and p14(ARK), p15(INK4b) and p16(INK4a) gene methylation rates were also significantly increased. With the working years and urinary 1-OH-Py's level, the rates of p14(ARK) and p16(INK4a) gene methylation were significantly increased while that of p15(INK4b) gene methylation displayed no statistical change. We conclude that PBMCs' p14(ARK) and p16(INK4a) gene methylation may be used for screening and warning lung cancer in coke oven workers.
Collapse
Affiliation(s)
- H Zhang
- Department of Environmental Health, Shanxi Medical University, Taiyuan, China
| | - X Li
- Center of Disease Control and Prevention, Taiyuan Iron and Steel Company, Taiyuan, China
| | - L Ge
- Department of Preventive Medicine, Hebei North University, Zhangjiakou, China
| | - J Yang
- Department of Occupational Health, Shanxi Medical University, Taiyuan, China
| | - J Sun
- Center of Disease Control and Prevention, Taiyuan Iron and Steel Company, Taiyuan, China
| | - Q Niu
- Department of Occupational Health, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
46
|
Lin J, Chen Q, Yang J, Qian J, Deng ZQ, Qian W, Chen XX, Ma JC, Xiong DS, Ma YJ, An C, Tang CY. DDX43 promoter is frequently hypomethylated and may predict a favorable outcome in acute myeloid leukemia. Leuk Res 2014; 38:601-7. [DOI: 10.1016/j.leukres.2014.02.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 02/15/2014] [Accepted: 02/24/2014] [Indexed: 12/30/2022]
|
47
|
MicroRNA let-7a-3 gene methylation is associated with karyotyping, CEBPA promoter methylation, and survival in acute myeloid leukemia. Leuk Res 2014; 38:625-31. [DOI: 10.1016/j.leukres.2014.03.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Revised: 03/05/2014] [Accepted: 03/06/2014] [Indexed: 01/01/2023]
|
48
|
Therapy of chronic myeloid leukemia: twilight of the imatinib era? ISRN ONCOLOGY 2014; 2014:596483. [PMID: 24634785 PMCID: PMC3929284 DOI: 10.1155/2014/596483] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2013] [Accepted: 12/18/2013] [Indexed: 11/19/2022]
Abstract
Chronic myeloid leukemia (CML) results from the clonal expansion of pluripotent hematopoietic stem cells containing the active BCR/ABL fusion gene produced by a reciprocal translocation of the ABL1 gene to the BCR gene. The BCR/ABL protein displays a constitutive tyrosine kinase activity and confers on leukemic cells growth and proliferation advantage and resistance to apoptosis. Introduction of imatinib (IM) and other tyrosine kinase inhibitors (TKIs) has radically improved the outcome of patients with CML and some other diseases with BCR/ABL expression. However, a fraction of CML patients presents with resistance to this drug. Regardless of clinical profits of IM, there are several drawbacks associated with its use, including lack of eradication of the malignant clone and increasing relapse rate resulting from long-term therapy, resistance, and intolerance. Second and third generations of TKIs have been developed to break IM resistance. Clinical studies revealed that the introduction of second-generation TKIs has improved the overall survival of CML patients; however, some with specific mutations such as T315I remain resistant. Second-generation TKIs may completely replace imatinib in perspective CML therapy, and addition of third-generation inhibitors may overcome resistance induced by every form of point mutations.
Collapse
|
49
|
Abstract
The myelodysplastic syndrome (MDS) is a clonal hematologic disorder that frequently evolves to acute myeloid leukemia (AML). Its pathogenesis remains unclear, but mutations in epigenetic modifiers are common and the disease often responds to DNA methylation inhibitors. We analyzed DNA methylation in the bone marrow and spleen in two mouse models of MDS/AML, the NUP98-HOXD13 (NHD13) mouse and the RUNX1 mutant mouse model. Methylation array analysis showed an average of 512/3445 (14.9%) genes hypermethylated in NHD13 MDS, and 331 (9.6%) genes hypermethylated in RUNX1 MDS. Thirty-two percent of genes in common between the two models (2/3 NHD13 mice and 2/3 RUNX1 mice) were also hypermethylated in at least two of 19 human MDS samples. Detailed analysis of 41 genes in mice showed progressive drift in DNA methylation from young to old normal bone marrow and spleen; to MDS, where we detected accelerated age-related methylation; and finally to AML, which markedly extends DNA methylation abnormalities. Most of these genes showed similar patterns in human MDS and AML. Repeat element hypomethylation was rare in MDS but marked the transition to AML in some cases. Our data show consistency in patterns of aberrant DNA methylation in human and mouse MDS and suggest that epigenetically, MDS displays an accelerated aging phenotype.
Collapse
|
50
|
Chen Y, Wang J. A membrane-based near-infrared fluorescence assay for detecting DNA methylation and transcription. Anal Biochem 2013; 442:196-204. [DOI: 10.1016/j.ab.2013.07.035] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Revised: 07/11/2013] [Accepted: 07/25/2013] [Indexed: 11/25/2022]
|