1
|
Cheng H, Liu Y, Cheng M, Li W, Sun M, Tang Q, Ma J, Li P, Gong T. IDH2 regulates U2AF1 expression and hydroxymethylation in MDS patients. Biotechnol Genet Eng Rev 2024; 40:788-799. [PMID: 36942631 DOI: 10.1080/02648725.2023.2190953] [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: 02/06/2023] [Accepted: 03/06/2023] [Indexed: 03/23/2023]
Abstract
The expression of some genes regulated by their DNA methylation is involved in pathogenesis and disease progression of myelodysplastic syndrome (MDS), which is characterised by abnormal differentiation and development of myeloid cells. Therefore, it is significant for us to work on investigating what factors regulate U2AF1 expression and hydroxymethylation in MDS patients. However, the members of TET protein family can change 5-methylcytosine (5mC) into 5-hydroxymethylcytosine5-methyl cytosine (5hmC). In general, 5mC and 5hmC levels maintain dynamic equilibrium, and their imbalance is associated with the onset and progression of some tumors. In this study, the expression and 5mC and 5hmC levels of U2AF1 gene decreased significantly after the treatment by decitabine in Mutz-1 cells. The decreased degree of 5hmC is far greater than that of 5mC. IDH2 expression decreased significantly followed by U2AF1 5hmC levels. However, the expression of other hydroxymethylation-related genes such as IDH1, TET1 and TET2 also decreased, but the difference did not achieve significance. Compared with IDH2 or U2AF1 wild-type MDS patients, U2AF1 expression and 5hmC level in patients with these two gene mutations were both significantly reduced.
Collapse
Affiliation(s)
- Huanchen Cheng
- Institute of Harbin Hematology & Oncology, The First Hospital of Harbin, Harbin, Heilongjiang, China
| | - Yu Liu
- Institute of Harbin Hematology & Oncology, The First Hospital of Harbin, Harbin, Heilongjiang, China
| | - Mei Cheng
- Institute of Harbin Hematology & Oncology, The First Hospital of Harbin, Harbin, Heilongjiang, China
| | - Wei Li
- Institute of Harbin Hematology & Oncology, The First Hospital of Harbin, Harbin, Heilongjiang, China
| | - Meng Sun
- Institute of Harbin Hematology & Oncology, The First Hospital of Harbin, Harbin, Heilongjiang, China
| | - Qinghua Tang
- Institute of Harbin Hematology & Oncology, The First Hospital of Harbin, Harbin, Heilongjiang, China
| | - Jun Ma
- Institute of Harbin Hematology & Oncology, The First Hospital of Harbin, Harbin, Heilongjiang, China
| | - Pu Li
- Institute of Harbin Hematology & Oncology, The First Hospital of Harbin, Harbin, Heilongjiang, China
| | - Tiejun Gong
- Institute of Harbin Hematology & Oncology, The First Hospital of Harbin, Harbin, Heilongjiang, China
| |
Collapse
|
2
|
Smith-Díaz CC, Magon NJ, McKenzie JL, Hampton MB, Vissers MCM, Das AB. Ascorbate Inhibits Proliferation and Promotes Myeloid Differentiation in TP53-Mutant Leukemia. Front Oncol 2021; 11:709543. [PMID: 34497762 PMCID: PMC8419345 DOI: 10.3389/fonc.2021.709543] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 08/02/2021] [Indexed: 11/26/2022] Open
Abstract
Loss-of-function mutations in the DNA demethylase TET2 are associated with the dysregulation of hematopoietic stem cell differentiation and arise in approximately 10% of de novo acute myeloid leukemia (AML). TET2 mutations coexist with other mutations in AML, including TP53 mutations, which can indicate a particularly poor prognosis. Ascorbate can function as an epigenetic therapeutic in pathological contexts involving heterozygous TET2 mutations by restoring TET2 activity. How this response is affected when myeloid leukemia cells harbor mutations in both TET2 and TP53 is unknown. Therefore, we examined the effects of ascorbate on the SKM-1 AML cell line that has mutated TET2 and TP53. Sustained treatment with ascorbate inhibited proliferation and promoted the differentiation of these cells. Furthermore, ascorbate treatment significantly increased 5-hydroxymethylcytosine, suggesting increased TET activity as the likely mechanism. We also investigated whether ascorbate affected the cytotoxicity of Prima-1Met, a drug that reactivates some p53 mutants and is currently in clinical trials for AML. We found that the addition of ascorbate had a minimal effect on Prima-1Met–induced cytotoxicity, with small increases or decreases in cytotoxicity being observed depending on the timing of treatment. Collectively, these data suggest that ascorbate could exert a beneficial anti-proliferative effect on AML cells harboring both TET2 and TP53 mutations whilst not interfering with targeted cytotoxic therapies such as Prima-1Met.
Collapse
Affiliation(s)
- Carlos C Smith-Díaz
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Nicholas J Magon
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Judith L McKenzie
- Haematology Research Group, Christchurch Hospital and Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Mark B Hampton
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Margreet C M Vissers
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Andrew B Das
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| |
Collapse
|
3
|
Štefík P, Annušová A, Lakatoš B, Elefantová K, Čepcová L, Hofbauerová M, Kálosi A, Jergel M, Majková E, Šiffalovič P. Targeting acute myeloid leukemia cells by CD33 receptor-specific MoS 2-based nanoconjugates. Biomed Mater 2021; 16. [PMID: 34280914 DOI: 10.1088/1748-605x/ac15b1] [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] [Received: 03/25/2021] [Accepted: 07/19/2021] [Indexed: 11/12/2022]
Abstract
Acute myeloid leukemia (AML) is a highly aggressive type of cancer caused by the uncontrolled proliferation of undifferentiated myeloblasts, affecting the bone marrow and blood. Systemic chemotherapy is considered the primary treatment strategy; unfortunately, healthy cells are also affected to a large extent, leading to severe side effects of this treatment. Targeted drug therapies are becoming increasingly popular in modern medicine, as they bypass normal tissues and cells. Two-dimensional MoS2-based nanomaterials have attracted attention in the biomedical field as promising agents for cancer diagnosis and therapy. Cancer cells typically (over)express distinctive cytoplasmic membrane-anchored or -spanning protein-based structures (e.g., receptors, enzymes) that distinguish them from healthy, non-cancerous cells. Targeting cancer cells via tumor-specific markers using MoS2-based nanocarriers loaded with labels or drugs can significantly improve specificity and reduce side effects of such treatment. SKM-1 is an established AML cell line that has been employed in various bio-research applications. However, to date, it has not been used as the subject of studies on selective cancer targeting by inorganic nanomaterials. Here, we demonstrate an efficient targeting of AML cells using MoS2nanoflakes prepared by a facile exfoliation route and functionalized with anti-CD33 antibody that binds to CD33 receptors expressed by SKM-1 cells. Microscopic analyses by confocal laser scanning microscopy supplemented by label-free confocal Raman microscopy proved that (anti-CD33)-MoS2conjugates were present on the cell surface and within SKM-1 cells, presumably having been internalized via CD33-mediated endocytosis. Furthermore, the cellular uptake of SKM-1 specific (anti-CD33)-MoS2conjugates assessed by flow cytometry analysis was significantly higher compared with the cellular uptake of SKM-1 nonspecific (anti-GPC3)-MoS2conjugates. Our results indicate the importance of appropriate functionalization of MoS2nanomaterials by tumor-recognizing elements that significantly increase their specificity and hence suggest the utilization of MoS2-based nanomaterials in the diagnosis and therapy of AML.
Collapse
Affiliation(s)
- Pavol Štefík
- Institute of Biochemistry and Microbiology, Faculty of Chemical and Food Technology, Slovak University of Technology, Radlinského 9, 81237 Bratislava, Slovakia
| | - Adriana Annušová
- Institute of Physics, Slovak Academy of Sciences, Dúbravská cesta 9, 84511 Bratislava, Slovakia.,Centre for Advanced Materials Application, Slovak Academy of Sciences, Dúbravská cesta 9, 84511 Bratislava, Slovakia
| | - Boris Lakatoš
- Institute of Biochemistry and Microbiology, Faculty of Chemical and Food Technology, Slovak University of Technology, Radlinského 9, 81237 Bratislava, Slovakia
| | - Katarína Elefantová
- Institute of Biochemistry and Microbiology, Faculty of Chemical and Food Technology, Slovak University of Technology, Radlinského 9, 81237 Bratislava, Slovakia
| | - Lucia Čepcová
- Institute of Biochemistry and Microbiology, Faculty of Chemical and Food Technology, Slovak University of Technology, Radlinského 9, 81237 Bratislava, Slovakia
| | - Monika Hofbauerová
- Institute of Physics, Slovak Academy of Sciences, Dúbravská cesta 9, 84511 Bratislava, Slovakia
| | - Anna Kálosi
- Centre for Advanced Materials Application, Slovak Academy of Sciences, Dúbravská cesta 9, 84511 Bratislava, Slovakia
| | - Matej Jergel
- Institute of Physics, Slovak Academy of Sciences, Dúbravská cesta 9, 84511 Bratislava, Slovakia.,Centre for Advanced Materials Application, Slovak Academy of Sciences, Dúbravská cesta 9, 84511 Bratislava, Slovakia
| | - Eva Majková
- Institute of Physics, Slovak Academy of Sciences, Dúbravská cesta 9, 84511 Bratislava, Slovakia.,Centre for Advanced Materials Application, Slovak Academy of Sciences, Dúbravská cesta 9, 84511 Bratislava, Slovakia
| | - Peter Šiffalovič
- Institute of Physics, Slovak Academy of Sciences, Dúbravská cesta 9, 84511 Bratislava, Slovakia.,Centre for Advanced Materials Application, Slovak Academy of Sciences, Dúbravská cesta 9, 84511 Bratislava, Slovakia
| |
Collapse
|
4
|
Zhu Y, Song D, Guo J, Jin J, Tao Y, Zhang Z, Xu F, He Q, Li X, Chang C, Wu L. U2AF1 mutation promotes tumorigenicity through facilitating autophagy flux mediated by FOXO3a activation in myelodysplastic syndromes. Cell Death Dis 2021; 12:655. [PMID: 34183647 PMCID: PMC8238956 DOI: 10.1038/s41419-021-03573-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/23/2021] [Accepted: 03/01/2021] [Indexed: 12/16/2022]
Abstract
Mutations in the U2 small nuclear RNA auxiliary factor 1 (U2AF1) gene are the common feature of a major subset in myelodysplastic syndromes (MDS). However, the genetic landscape and molecular pathogenesis of oncogenic U2AF1S34F mutation in MDS are not totally understood. We performed comprehensive analysis for prognostic significance of U2AF1 mutations in acute myeloid leukemia (AML) cohort based on The Cancer Genome Atlas (TCGA) database. Functional analysis of U2AF1S34F mutation was performed in vitro. Differentially expressed genes (DEGs) and significantly enriched pathways were identified by RNA sequencing. The forkhead box protein O3a (FOXO3a) was investigated to mediate the function of U2AF1S34F mutation in cell models using lentivirus. Chromatin immunoprecipitation, immunoblotting analyses, and immunofluorescence assays were also conducted. U2AF1 mutations were associated with poor prognosis in MDS and AML samples, which significantly inhibited cell proliferation and induced cellular apoptosis in cell models. Our data identified that U2AF1-mutant cell lines undergo FOXO3a-dependent apoptosis and NLRP3 inflammasome activation, which induces pyroptotic cell death. Particularly, an increase in the level of FOXO3a promoted the progression of MDS in association with restored autophagy program leading to NLRP3 inflammasome activation in response to U2AF1S34F mutation. Based on the result that U2AF1S34F mutation promoted the transcriptional activity of Bim through upregulating FOXO3a with transactivation of cell cycle regulators p21Cip1 and p27Kip1, FOXO3a, a potentially cancer-associated transcription factor, was identified as the key molecule on which these pathways converge. Overall, our studies provide new insights that U2AF1S34F mutation functions the crucial roles in mediating MDS disease progression via FOXO3a activation, and demonstrate novel targets of U2AF1 mutations to the pathogenesis of MDS.
Collapse
Affiliation(s)
- Yuqian Zhu
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Dandan Song
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Juan Guo
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Jiacheng Jin
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Ying Tao
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Zheng Zhang
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Feng Xu
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Qi He
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Xiao Li
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Chunkang Chang
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Lingyun Wu
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.
| |
Collapse
|
5
|
Luo J, Mu J, Zhang M, Zhao B, Liu L. SPAG6-silencing enhances decitabine-induced apoptosis and demethylation of PTEN in SKM-1 cells and in a xenograft mouse model. Leuk Lymphoma 2021; 62:2242-2252. [PMID: 33843428 DOI: 10.1080/10428194.2021.1913148] [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: 12/12/2022]
Abstract
Myelodysplastic syndromes (MDS) are a group of malignant diseases that are characterized by disordered hematopoiesis with a high risk of transforming into leukemia. In the present study, SPAG6-knockdown and decitabine (DAC) treatment resulted in a decreased DNA methyltransferases and methyl-CpG-binding domain protein expression. In addition, DAC and LBH589 were shown to promote apoptosis in SKM-1 cells, and SPAG6-knockdown to enhance the pro-apoptotic effect of DAC. DAC could reduce PTEN methylation and increase PTEN expression in SKM-1 cells. SPAG6-knockdown and LBH589 treatment could increase DAC-mediated demethylation of PTEN promoter. Finally, a mouse model was constructed, and an enhanced efficacy of DAC following SPAG6-knockdown was confirmed in vivo. In conclusion, DAC-mediated apoptosis and PTEN promoter demethylation may be synergistically enhanced by SPAG6-silencing. Therefore, in the present study it was indicated that SPAG6 may be a potential target for demethylation therapy in MDS.
Collapse
Affiliation(s)
- Jie Luo
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiao Mu
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Meng Zhang
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Beibei Zhao
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lin Liu
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
6
|
Wang MJ, Liu WY, Wang XY, Li YM, Xiao HY, Quan RC, Huang G, Hu XM. Autophagy Gene Panel-Based Prognostic Model in Myelodysplastic Syndrome. Front Oncol 2021; 10:606928. [PMID: 33614490 PMCID: PMC7894207 DOI: 10.3389/fonc.2020.606928] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 11/19/2020] [Indexed: 01/18/2023] Open
Abstract
Abnormal autophagy is related to the pathogenesis and clinical symptoms of myelodysplastic syndrome (MDS). However, the effect of autophagy-related genes (ARGs) on the prognosis of MDS remains unclear. Here, we examined the expression profile of 108 patients with MDS from the GSE58831 dataset, and identified 22 genes that were significantly associated with overall survival. Among them, seven ARGs were screened and APIs were calculated for all samples based on the expression of the seven ARGs, and then, MDS patients were categorized into high- and low-risk groups based on the median APIs. The overall survival of patients with high-risk scores based on these seven ARGs was shorter than patients with low-risk scores in both the training cohort (P = 2.851e-06) and the validation cohort (P = 9.265e-03). Additionally, API showed an independent prognostic indicator for survival in the training samples [hazard ratio (HR) = 1.322, 95% confidence interval (CI): 1.158–1.51; P < 0.001] and the validation cohort (HR = 1.05, 95% CI: 1–1.1; P < 0.01). The area under the receiver operating characteristic curve (AUROC) of API and IPSS were 43.0137 and 66.0274 in the training cohorts and the AUC of the validation cohorts were 41.5361 and 72.0219. Our data indicate these seven ARGs can predict prognosis in patients with MDS and could guide individualized treatment.
Collapse
Affiliation(s)
- Ming-Jing Wang
- Department of Hematology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.,Graduate School, China Academy of Chinese Medical Sciences, Beijing, China
| | - Wei-Yi Liu
- Department of Hematology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xue-Ying Wang
- Department of Hematology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.,Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Yu-Meng Li
- Department of Hematology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.,Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Hai-Yan Xiao
- Department of Hematology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ri-Cheng Quan
- Department of Hematology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Gang Huang
- Divisions of Pathology and Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Xiao-Mei Hu
- Department of Hematology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
7
|
Zhang Z, Jia Y, Xv F, Song LX, Shi L, Guo J, Chang CK. Decitabine Induces Change of Biological Traits in Myelodysplastic Syndromes via FOXO1 Activation. Front Genet 2021; 11:603956. [PMID: 33584800 PMCID: PMC7873873 DOI: 10.3389/fgene.2020.603956] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 12/04/2020] [Indexed: 11/26/2022] Open
Abstract
Decitabine (DAC) is considered to be a profound global DNA demethylation, which can induce the re-expression of silenced tumor suppressor genes. Little is known about the function of tumor suppressor gene FOXO1 in myelodysplastic syndromes (MDS). To address this issue, the study firstly investigated differentially expressed genes (DEGs) for DAC treatment in MDS cell lines, then explored the role of FOXO1 through silencing its expression before DAC treatment in MDS. The results showed that FOXO1 exists in a hyperphosphorylated, inactive form in MDS-L cells. DAC treatment both induces FOXO1 expression and reactivates the protein in its low phosphorylation level. Additionally, the results also demonstrated that this FOXO1 activation is responsible for the DAC-induced apoptosis, cell cycle arrest, antigen differentiation, and immunoregulation in MDS-L cells. We also demonstrated DAC-induced FOXO1 activation upregulates anti-tumor immune response in higher-risk MDS specimens. Collectively, these results suggest that DAC induces FOXO1 activation, which plays an important role in anti-MDS tumors.
Collapse
Affiliation(s)
- Zheng Zhang
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yan Jia
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Feng Xv
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Lu-Xi Song
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Lei Shi
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Juan Guo
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Chun-Kang Chang
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
8
|
Mpakou V, Spathis A, Bouhla A, Mpazani E, Papageorgiou S, Gkontopoulos K, Glezou E, Thomopoulos T, Foukas P, Pappa V. Synergistic inhibitory effects of low-dose decitabine in combination with bortezomib in the AML cell line Kasumi-1. Exp Ther Med 2021; 21:195. [PMID: 33488804 PMCID: PMC7812574 DOI: 10.3892/etm.2021.9628] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 11/17/2020] [Indexed: 11/29/2022] Open
Abstract
In the present study, the ability of the proteasome inhibitor bortezomib (BZ), an oxidative stress-inducing agent, to sensitize acute myeloid leukemia (AML) cells to decitabine (Dacogen®, DAC; a DNA methyltransferase inhibitor), in terms of cell viability and differentiation, was investigated. Kasumi-1 AML (M2) cells were treated with low-dose DAC (10, 50, 100, 200 or 400 nΜ), with or without BZ (10 nM). Apoptosis and the cell cycle were evaluated after 24 h of treatment through fluorescence-assisted cell sorting (FACS) with Annexin V/propidium iodide and DAPI staining, respectively. The expression levels of CD193, CD11b, CD13, CD14, CD15, CD16 and CD117 surface differentiation markers were evaluated by FACS after 6 days of treatment. The results indicated significant alterations in cell death and cell cycle phases in Kasumi-1 cells following DAC and BZ combination treatment compared to untreated cells and cells with single treatments. Low-dose DAC/BZ combinations significantly enhanced apoptosis and decreased the population of live Kasumi-1 cells, with 100 and 200 nM of DAC and 10 nM BZ appearing to have the most potent synergistic effect according to a combination index. Furthermore, cell cycle profiling revealed that DAC/BZ treatment synergistically led to G0/G1- and G2/M-phase arrest. By contrast, DAC appeared to promote monocytic and granulocytic differentiation of Kasumi-1 cells more effectively alone than in combination with BZ. BZ acted synergistically with low-dose DAC in vitro, leading to enhanced apoptosis and G0/G1- and G2/M-phase arrest in AML cells, hence prohibiting either DNA synthesis or mitosis. Although further in vivo investigation is necessary, these results provide a strong rationale for the implementation of a combination treatment with DAC and bortezomib in AML therapy, followed by DAC alone, which may achieve better clinical responses and possibly partially overcome the frequently encountered DAC resistance of patients with AML.
Collapse
Affiliation(s)
- Vassiliki Mpakou
- Second Department of Internal Medicine and Research Institute, National and Kapodistrian University of Athens, Attikon University General Hospital, Athens 12462, Greece
| | - Aris Spathis
- Second Department of Pathology, National and Kapodistrian University of Athens, Attikon University General Hospital, Athens 12462, Greece
| | - Anthi Bouhla
- Second Department of Internal Medicine and Research Institute, National and Kapodistrian University of Athens, Attikon University General Hospital, Athens 12462, Greece
| | - Efthimia Mpazani
- Second Department of Internal Medicine and Research Institute, National and Kapodistrian University of Athens, Attikon University General Hospital, Athens 12462, Greece
| | - Sotirios Papageorgiou
- Second Department of Internal Medicine and Research Institute, National and Kapodistrian University of Athens, Attikon University General Hospital, Athens 12462, Greece
| | - Konstantinos Gkontopoulos
- Second Department of Internal Medicine and Research Institute, National and Kapodistrian University of Athens, Attikon University General Hospital, Athens 12462, Greece
| | - Eirini Glezou
- Second Department of Internal Medicine and Research Institute, National and Kapodistrian University of Athens, Attikon University General Hospital, Athens 12462, Greece
| | - Thomas Thomopoulos
- Second Department of Internal Medicine and Research Institute, National and Kapodistrian University of Athens, Attikon University General Hospital, Athens 12462, Greece
| | - Periklis Foukas
- Second Department of Pathology, National and Kapodistrian University of Athens, Attikon University General Hospital, Athens 12462, Greece
| | - Vasiliki Pappa
- Second Department of Internal Medicine and Research Institute, National and Kapodistrian University of Athens, Attikon University General Hospital, Athens 12462, Greece
| |
Collapse
|
9
|
Epigenetic changes in FOXO3 and CHEK2 genes and their correlation with clinicopathological findings in myelodysplastic syndromes. Hematol Oncol Stem Cell Ther 2020; 13:214-219. [PMID: 32217071 DOI: 10.1016/j.hemonc.2019.11.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 11/07/2019] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVES/BACKGROUND Myelodysplastic syndromes (MDSs) are a heterogeneous disease in terms of clinical course and response to therapy. Epigenetic changes are the primary mechanism of MDS pathogenesis. FOXO3 and CHEK2 genes play significant roles in normal cellular mechanisms and are also known as tumor suppressor genes. We aimed to clarify the correlation of epigenetic changes in these genes with clinicopathologic findings in MDS. METHODS A total of 54 newly diagnosed MDS patients referred to Shariati and Firouzgar Hospitals (Tehran, Iran) were included in the study from 2013 to 2015, comprising the following cases: 26 with refractory cytopenia with unilineage dysplasia, 10 with refractory cytopenia with multilineage dysplasia, four refractory anemia with excess blasts-1 (RAEB-1), 11 refractory anemia with excess blasts-2 (RAEB-2), and three MDS associated with isolated deletion (5q-). Risk groups were determined according to the Revised International Prognostic Scoring System (IPSS-R). The methylation status of CHEK2 and FOXO3 promoters were determined by methylation-sensitive high-resolution melting analysis of sodium bisulfite-converted DNA. Expressions of CHEK2, FOXO3, and GAPDH were measured by quantitative real-time polymerase chain reaction and fold changes were calculated using the ΔΔCT method. RESULTS Statistical analysis revealed no promoter methylation of CHEK2 and FOXO3 in healthy control specimens. FOXO3 promoter methylation was associated with high-risk World Health Organization subgroups (p = .017), high-risk IPSS-R (p = .007), high-risk cytogenetics (p = .045), and more than 5% blasts in bone marrow (p = .001). CHEK2 promoter methylation was correlated with more than 5% blasts in bone marrow (p = .009). CONCLUSIONS Promoter methylation of CHEK2 and especially FOXO3 is associated with adverse clinicopathological findings and disease progression in MDS.
Collapse
|
10
|
The Emerging Role of Sperm-Associated Antigen 6 Gene in the Microtubule Function of Cells and Cancer. MOLECULAR THERAPY-ONCOLYTICS 2019; 15:101-107. [PMID: 31660426 PMCID: PMC6807308 DOI: 10.1016/j.omto.2019.08.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Accumulated evidence shows that sperm-associated antigen 6 (SPAG6) gene has multiple biological functions. It maintains the normal function of a variety of cells including ciliary/flagellar biogenesis and polarization, neurogenesis, and neuronal migration. Moreover, SPAG6 is found to be critically involved in auditory transduction and the fibroblast life cycle. Furthermore, SPAG6 plays an essential role in immuno-regulation. Notably, SPAG6 has been demonstrated to participate in the occurrence and progression of a variety of human cancers. New evidence shows that SPAG6 gene regulates tumor cell proliferation, apoptosis, invasion, and metastasis. Therefore, in this review, we describe the physiological function and mechanism of SPAG6 in human normal cells and cancer cells. We also highlight that SPAG6 gene may be an effective biomarker for the diagnosis of human cancer. Taken together, targeting SPAG6 could be a novel strategy for the treatment of human diseases including cancer.
Collapse
|
11
|
Therapeutic Modulation of Autophagy in Leukaemia and Lymphoma. Cells 2019; 8:cells8020103. [PMID: 30704144 PMCID: PMC6406467 DOI: 10.3390/cells8020103] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 01/26/2019] [Accepted: 01/28/2019] [Indexed: 02/07/2023] Open
Abstract
Haematopoiesis is a tightly orchestrated process where a pool of hematopoietic stem and progenitor cells (HSPCs) with high self-renewal potential can give rise to both lymphoid and myeloid lineages. The HSPCs pool is reduced with ageing resulting in few HSPC clones maintaining haematopoiesis thereby reducing blood cell diversity, a phenomenon called clonal haematopoiesis. Clonal expansion of HSPCs carrying specific genetic mutations leads to increased risk for haematological malignancies. Therefore, it comes as no surprise that hematopoietic tumours develop in higher frequency in elderly people. Unfortunately, elderly patients with leukaemia or lymphoma still have an unsatisfactory prognosis compared to younger ones highlighting the need to develop more efficient therapies for this group of patients. Growing evidence indicates that macroautophagy (hereafter referred to as autophagy) is essential for health and longevity. This review is focusing on the role of autophagy in normal haematopoiesis as well as in leukaemia and lymphoma development. Attenuated autophagy may support early hematopoietic neoplasia whereas activation of autophagy in later stages of tumour development and in response to a variety of therapies rather triggers a pro-tumoral response. Novel insights into the role of autophagy in haematopoiesis will be discussed in light of designing new autophagy modulating therapies in hematopoietic cancers.
Collapse
|
12
|
Targeting Cancer through the Epigenetic Features of Telomeric Regions. Trends Cell Biol 2019; 29:281-290. [PMID: 30660503 DOI: 10.1016/j.tcb.2018.12.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 12/18/2018] [Accepted: 12/20/2018] [Indexed: 12/16/2022]
Abstract
The integrity of the chromatin associated with telomeric regions, which include telomeres and subtelomeres, is essential for telomeres function and cell viability. Whereas human subtelomeres are heterochromatic, telomeres are labeled with euchromatic marks like H4K20me1 and H3K27ac in most commonly studied human cell lines. The epigenetic marks of human telomeric regions influence oncogenic processes. Indeed, different drugs that decrease their genome-wide levels are currently being used or tested in specific cancer therapies. These drugs can challenge cancer by altering the function of key cellular proteins. However, they should also compromise oncogenic processes by modifying the epigenetic landscape of telomeric regions. We believe that studies of telomeric chromatin structure and telomeres dysfunction should help to design epigenetic therapies for cancer treatment.
Collapse
|
13
|
A novel all-trans retinoic acid derivative inhibits proliferation and induces apoptosis of myelodysplastic syndromes cell line SKM-1 cells via up-regulating p53. Int Immunopharmacol 2018; 65:561-570. [DOI: 10.1016/j.intimp.2018.10.041] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 10/19/2018] [Accepted: 10/30/2018] [Indexed: 12/31/2022]
|
14
|
Li G, Song Y, Li G, Ren J, Xie J, Zhang Y, Gao F, Mu J, Dai J. Downregulation of microRNA‑21 expression inhibits proliferation, and induces G1 arrest and apoptosis via the PTEN/AKT pathway in SKM‑1 cells. Mol Med Rep 2018; 18:2771-2779. [PMID: 30015844 PMCID: PMC6102657 DOI: 10.3892/mmr.2018.9255] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 04/19/2018] [Indexed: 12/27/2022] Open
Abstract
Myelodysplastic syndromes (MDS) are characterized by ineffective hematopoiesis and may progress to acute myeloid leukemia (AML). MicroRNAs (miRNA/miRs) as oncogenes or tumor suppressors regulate a number of biological processes including cell proliferation, cell cycle and apoptosis in different types of cancer cells. Recently, it has been reported that miR-21 as an oncogene is overexpressed and directly targets SMAD-7 in MDS. However, little is known about the mechanism of miR-21 in the progression of MDS. In the present study, the role of miR-21 in the proliferation and apoptosis of SKM-1 cells, an acute myeloid leukemia cell line established in the AML/MDS leukemic phase was investigated. The present results demonstrated that downregulation of miR-21 inhibited proliferation, induced apoptosis and caused G1 phase cell cycle arrest of SKM-1 cells. In addition, the expression levels of apoptosis regulator Bcl-2 (bcl2), cyclinD1 and phosphorylated-protein kinase B (AKT) were significantly decreased in SKM-1 cells transfected with the miR-21 inhibitor, whilst the expression levels of phosphatase and tensin homolog (PTEN), bcl-associated protein X (bax) and cleaved caspase 3 were significantly elevated. Furthermore, knockdown of Akt by small interfering (si)RNA significantly increased the expression of bax, cleaved caspase 3 and reduced the expression of bcl2 and cyclinD1 in SKM-1 cells. Taken together, these data indicate that miR-21 targets the PTEN/AKT pathway in the pathogenesis of MDS and could be a potential target for MDS therapy.
Collapse
Affiliation(s)
- Guang Li
- Institute of Hematopathy, Xi'an Central Hospital, Xi'an, Shaanxi 710003, P.R. China
| | - Yanping Song
- Institute of Hematopathy, Xi'an Central Hospital, Xi'an, Shaanxi 710003, P.R. China
| | - Gangcan Li
- Institute of Hematopathy, Xi'an Central Hospital, Xi'an, Shaanxi 710003, P.R. China
| | - Jingjing Ren
- Institute of Hematopathy, Xi'an Central Hospital, Xi'an, Shaanxi 710003, P.R. China
| | - Jia Xie
- Institute of Hematopathy, Xi'an Central Hospital, Xi'an, Shaanxi 710003, P.R. China
| | - Yunjie Zhang
- Institute of Hematopathy, Xi'an Central Hospital, Xi'an, Shaanxi 710003, P.R. China
| | - Fei Gao
- Institute of Hematopathy, Xi'an Central Hospital, Xi'an, Shaanxi 710003, P.R. China
| | - Jiao Mu
- Institute of Hematopathy, Xi'an Central Hospital, Xi'an, Shaanxi 710003, P.R. China
| | - Jinqian Dai
- Institute of Hematopathy, Xi'an Central Hospital, Xi'an, Shaanxi 710003, P.R. China
| |
Collapse
|
15
|
Czaja AJ. Under-Evaluated or Unassessed Pathogenic Pathways in Autoimmune Hepatitis and Implications for Future Management. Dig Dis Sci 2018; 63:1706-1725. [PMID: 29671161 DOI: 10.1007/s10620-018-5072-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 04/12/2018] [Indexed: 12/11/2022]
Abstract
Autoimmune hepatitis is a consequence of perturbations in homeostatic mechanisms that maintain self-tolerance but are incompletely understood. The goals of this review are to describe key pathogenic pathways that have been under-evaluated or unassessed in autoimmune hepatitis, describe insights that may shape future therapies, and encourage investigational efforts. The T cell immunoglobulin mucin proteins constitute a family that modulates immune tolerance by limiting the survival of immune effector cells, clearing apoptotic bodies, and expanding the population of granulocytic myeloid-derived suppressor cells. Galectins influence immune cell migration, activation, proliferation, and survival, and T cell exhaustion can be induced and exploited as a possible management strategy. The programmed cell death-1 protein and its ligands comprise an antigen-independent inhibitory axis that can limit the performance of activated T cells by altering their metabolism, and epigenetic changes can silence pro-inflammatory genes or de-repress anti-inflammatory genes that affect disease severity. Changes in the intestinal microbiota and permeability of the intestinal mucosal barrier can be causative or consequential events that affect the occurrence and phenotype of immune-mediated disease, and they may help explain the female propensity for autoimmune hepatitis. Perturbations within these homeostatic mechanisms have been implicated in experimental models and limited clinical experiences, and they have been favorably manipulated by monoclonal antibodies, recombinant molecules, pharmacological agents or dietary supplements. In conclusion, pathogenic mechanisms that have been implicated in other systemic immune-mediated and liver diseases but under-evaluated or unassessed in autoimmune hepatitis warrant consideration and rigorous evaluation.
Collapse
Affiliation(s)
- Albert J Czaja
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Science, 200 First Street S.W., Rochester, MN, 55905, USA.
| |
Collapse
|