1
|
Park MN. The Therapeutic Potential of a Strategy to Prevent Acute Myeloid Leukemia Stem Cell Reprogramming in Older Patients. Int J Mol Sci 2023; 24:12037. [PMID: 37569414 PMCID: PMC10418941 DOI: 10.3390/ijms241512037] [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: 06/26/2023] [Revised: 07/22/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
Acute myeloid leukemia (AML) is the most common and incurable leukemia subtype. Despite extensive research into the disease's intricate molecular mechanisms, effective treatments or expanded diagnostic or prognostic markers for AML have not yet been identified. The morphological, immunophenotypic, cytogenetic, biomolecular, and clinical characteristics of AML patients are extensive and complex. Leukemia stem cells (LSCs) consist of hematopoietic stem cells (HSCs) and cancer cells transformed by a complex, finely-tuned interaction that causes the complexity of AML. Microenvironmental regulation of LSCs dormancy and the diagnostic and therapeutic implications for identifying and targeting LSCs due to their significance in the pathogenesis of AML are discussed in this review. It is essential to perceive the relationship between the niche for LSCs and HSCs, which together cause the progression of AML. Notably, methylation is a well-known epigenetic change that is significant in AML, and our data also reveal that microRNAs are a unique factor for LSCs. Multiple-targeted approaches to reduce the risk of epigenetic factors, such as the administration of natural compounds for the elimination of local LSCs, may prevent potentially fatal relapses. Furthermore, the survival analysis of overlapping genes revealed that specific targets had significant effects on the survival and prognosis of patients. We predict that the multiple-targeted effects of herbal products on epigenetic modification are governed by different mechanisms in AML and could prevent potentially fatal relapses. Thus, these strategies can facilitate the incorporation of herbal medicine and natural compounds into the advanced drug discovery and development processes achievable with Network Pharmacology research.
Collapse
Affiliation(s)
- Moon Nyeo Park
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemungu, Seoul 05253, Republic of Korea
| |
Collapse
|
2
|
Donia HM, Elsweify NM, Farahat NM, Nadwan EA. Wilms tumor 1 gene expression in acute myeloid leukemia: prognostic significance and usefulness in minimal residual disease monitoring—a case–control study. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2022. [DOI: 10.1186/s43042-022-00291-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Minimal residual disease (MRD), which is characterized as leukemic cells at a level below morphologic detection, has been connected to the risk of relapse in acute myeloid leukemia. In 80–90% of acute myeloid leukemia (AML) patients, the Wilms tumor (WT1) gene is overexpressed at the mRNA level. In our prospective study, a total of 55 patients were enrolled in the study. Group I involved 40 AML patients and group II involved 15 patients healthy controls. WT1 gene expression was quantified using quantitative real-time PCR on bone marrow samples from AML patients at initial diagnosis and at day 28 after induction chemotherapy, and compared to 15 healthy controls in group II. Follow up of patients for prognosis evaluation was assessed. IBM SPSS software was used to capture and analyses the data.
Results
At diagnosis, the mean WT1 transcript value in AML patients was substantially higher than the expression observed in control patient’s Bone marrow. There was no statistically relevant relationship between the onset of relapse and WT1 expression. Patients with WT1 overexpression at diagnosis had a shorter overall survival than patients with negative WT1 expression.
Conclusions
Wilms tumor 1 gene expression was found to be significantly higher in AML patients than control cases, overall, our results confirmed the prognostic significance of WT1 overexpression in AML patients. Our findings support the application of MRD in AML patients based on WT1 overexpression.
Collapse
|
3
|
Pishbin F, Ziamajidi N, Abbasalipourkabir R, Najafi R, Farhadian M. Correlation of miR-600 with WT1 expression and its potential clinical significance in breast cancer. Per Med 2021; 18:31-42. [PMID: 33393369 DOI: 10.2217/pme-2020-0010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Aim: The study aimed to explore miR-600 and WT1 expression and its potential clinical significance in breast cancer. Materials & methods: The expression of miR-600 and WT1 in tumor and non-tumor adjacent tissues in 45 breast cancer patients as well as serum level of miR-600 in these patients and 45 healthy group were analyzed. Results: The expression level of miR-600 in tumor tissue and serum of patients was significantly lower than non-tumor adjacent tissues and serum of controls, respectively, while WT1 mRNA and protein levels were higher in tumor tissues compared with non-tumor adjacent tissues. The miR-600 expression was correlated with lymph node metastasis and clinical stage. Conclusion: The miR-600 acts as tumor suppressor and a diagnostic and prognostic biomarker in breast cancer patients.
Collapse
Affiliation(s)
- Fariba Pishbin
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Nasrin Ziamajidi
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.,Molecular Medicine Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Roghayeh Abbasalipourkabir
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Rezvan Najafi
- Department of Molecular Medicine & Genetics, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Maryam Farhadian
- Department of Biostatistics, School of Health, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
4
|
Zhou B, Jin X, Jin W, Huang X, Wu Y, Li H, Zhu W, Qin X, Ye H, Gao S. WT1 facilitates the self-renewal of leukemia-initiating cells through the upregulation of BCL2L2: WT1-BCL2L2 axis as a new acute myeloid leukemia therapy target. J Transl Med 2020; 18:254. [PMID: 32580769 PMCID: PMC7313134 DOI: 10.1186/s12967-020-02384-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 05/21/2020] [Indexed: 12/24/2022] Open
Abstract
Background Overexpression of Wilms’ tumor-1 (WT1) transcription factor facilitates proliferation in acute myeloid leukemia (AML). However, whether WT1 is enriched in the leukemia-initiating cells (LICs) and leukemia stem cells (LSCs) and facilitates the self-renewal of LSCs remains poorly understood. Methods MLL-AF9-induced murine leukemia model was used to evaluate the effect of knockdown of wt1 on the self-renewal ability of LSC. RNA sequencing was performed on WT1-overexpressing cells to select WT1 targets. Apoptosis and colony formation assays were used to assess the anti-leukemic potential of a deubiquitinase inhibitor WP1130. Furthermore, NOD/SCID-IL2Rγ (NSG) AML xenotransplantation and MLL-AF9-induced murine leukemia models were used to evaluate the anti-leukemogenic potential of WP1130 in vivo. Results We found that wt1 is highly expressed in LICs and LSCs and facilitates the maintenance of leukemia in a murine MLL-AF9-induced model of AML. WT1 enhanced the self-renewal of LSC by increasing the expression of BCL2L2, a member of B cell lymphoma 2 (BCL2) family, by direct binding to its promoter region. Loss of WT1 impaired self-renewal ability in LSC and delayed the progression of leukemia. WP1130 was found to modify the WT1-BCL2L2 axis, and WP1130-induced anti-leukemic activity was mediated by ubiquitin proteasome-mediated destruction of WT1 protein. WP1130 induced apoptosis and decreased colony formation abilities of leukemia cells and prolonged the overall survival in the THP1-based xenograft NSG mouse model. WP1130 also decreased the frequency of LSC and prolonged the overall survival in MLL-AF9-induced murine leukemia model. Mechanistically, WP1130 induced the degradation of WT1 by positively affecting the ubiquitination of WT1 protein. Conclusions Our results indicate that WT1 is required for the development of AML. WP1130 exhibits anti-leukemic activity by inhibiting the WT1-BCL2L2 axis, which may represent a new acute myeloid leukemia therapy target.
Collapse
Affiliation(s)
- Bin Zhou
- Laboratory of Internal Medicine, The First Affiliated Hospital of Wenzhou Medical University, 1 Xuefubei Street, Ouhai District, Wenzhou, 325000, Zhejiang, China
| | - Xianghong Jin
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, 1 Xuefubei Street, Ouhai District, Wenzhou, 325000, Zhejiang, China
| | - Weiwei Jin
- Department of Obstetrics and Gynecology, Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine, Wenzhou, 325000, Zhejiang, China
| | - Xingzhou Huang
- Laboratory of Internal Medicine, The First Affiliated Hospital of Wenzhou Medical University, 1 Xuefubei Street, Ouhai District, Wenzhou, 325000, Zhejiang, China
| | - Yanfei Wu
- Laboratory of Internal Medicine, The First Affiliated Hospital of Wenzhou Medical University, 1 Xuefubei Street, Ouhai District, Wenzhou, 325000, Zhejiang, China
| | - Haiying Li
- Laboratory of Internal Medicine, The First Affiliated Hospital of Wenzhou Medical University, 1 Xuefubei Street, Ouhai District, Wenzhou, 325000, Zhejiang, China
| | - Weijian Zhu
- Laboratory of Internal Medicine, The First Affiliated Hospital of Wenzhou Medical University, 1 Xuefubei Street, Ouhai District, Wenzhou, 325000, Zhejiang, China
| | - Xiaoyi Qin
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, 1 Xuefubei Street, Ouhai District, Wenzhou, 325000, Zhejiang, China
| | - Haige Ye
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, 1 Xuefubei Street, Ouhai District, Wenzhou, 325000, Zhejiang, China.
| | - Shenmeng Gao
- Laboratory of Internal Medicine, The First Affiliated Hospital of Wenzhou Medical University, 1 Xuefubei Street, Ouhai District, Wenzhou, 325000, Zhejiang, China.
| |
Collapse
|
5
|
Li Q, Wang J. LncRNA TUG1 Regulates Cell Viability and Death by Regulating miR-193a-5p/Rab10 Axis in Acute Myeloid Leukemia. Onco Targets Ther 2020; 13:1289-1301. [PMID: 32103996 PMCID: PMC7025684 DOI: 10.2147/ott.s234935] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 12/20/2019] [Indexed: 12/13/2022] Open
Abstract
Background Acute myeloid leukemia (AML) is a serious threat to human health. Long non-coding RNA (lncRNA) Taurine-Upregulated Gene1 (TUG1) has been reported to participate in the development and progression of several cancers, including AML. Herein, we aimed to investigate the pathognomonic role of TUG1 in AML cells and its potential mechanistic pathway. Methods Quantitative real-time PCR (qRT-PCR) assay was applied to detect the expression levels of lncRNA TUG1, miR-193a-5p and Rab10 in AML bone marrow and cell lines. The CCK-8 assay was conducted to assess the cell viability of AML HL-60 and NB4 cells and cell apoptotic assay was performed to assess the cell death. Dual-luciferase reporter assay was carried out to clarify the relationships among TUG1, miR-193a-5p and Rab10. Also, the protein level of Rab10 was examined by Western blot assay. Results LncRNA TUG1 was up-regulated in AML bone marrow and cells. Functional analysis showed that the silencing of TUG1 suppressed cell viability, while promoted cell death in AML HL-60 and NB4 cells. TUG1 targeted miR-193a-5p and negatively regulated miR-193a-5p expression. Overexpressed miR-193a-5p resulted in the decrease of cell viability and the increase in the cell death in AML cells. Restoration experiments proved that TUG1 regulated the cell viability and death of AML cells through regulating the miR-193a-5p/Rab10 axis. Rab10 was a direct target of miR-193a-5p and was inversely regulated by miR-193a-5p. TUG1 regulated the cell viability and death of AML cells through upregulating Rab10. Conclusion Silencing of lncRNA TUG1 induces a cytotoxic effect on AML cell lines through sponging miR-193a-5p and the suppression of Rab10.
Collapse
Affiliation(s)
- Qun Li
- Department of PICU, First People's Hospital of Shangqiu City, Shangqiu, Henan Province, People's Republic of China
| | - Jianmin Wang
- Department of PICU, First People's Hospital of Shangqiu City, Shangqiu, Henan Province, People's Republic of China
| |
Collapse
|
6
|
Li J, Li H, Zhu W, Zhou B, Ying J, Wu J, Zhang H, Sun H, Gao S. Deubiquitinase inhibitor degrasyn suppresses metastasis by targeting USP5-WT1-E-cadherin signalling pathway in pancreatic ductal adenocarcinoma. J Cell Mol Med 2020; 24:1370-1382. [PMID: 31845546 PMCID: PMC6991651 DOI: 10.1111/jcmm.14813] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 10/05/2019] [Accepted: 10/26/2019] [Indexed: 12/18/2022] Open
Abstract
Wilm's tumour-1 (WT1) is overexpressed in pancreatic ductal adenocarcinoma (PDAC) and enhances metastasis. Deubiquitination stabilizes target proteins, and inhibiting deubiquitination facilitates the degradation of target proteins. However, whether inhibiting deubiquitination of WT1 facilitates its degradation and presents anti-cancer ability in PDAC is unknown. Here, we found that deubiquitinase inhibitor degrasyn rapidly induced the degradation of endogenous and exogenous WT1 through enhancing ubiquitination of WT1 followed by the up-regulation of E-cadherin. Knockdown of WT1 by short hairpin RNAs (shRNAs) inhibited metastasis and overexpression of WT1 partially prevented degrasyn-induced anti-metastasis activity, suggesting that degrasyn presents anti-metastasis activity partially through degrading WT1 protein. We further identified that USP5 deubiquitinated WT1 and stabilized its expression. The higher expressions of USP5 and WT1 are associated with tumour metastasis. More importantly, degrasyn inhibited the activity of USP5 and overexpression of USP5 partially prevented degrasyn-induced degradation of WT1 protein, suggesting that degrasyn degraded WT1 protein through inhibiting the activity of USP5. Finally, degrasyn reduced the tumorigenicity in a xenograft mouse model and reduced the metastasis in vivo. Our results indicate that degrasyn presents strong anti-cancer activity through USP5-WT1-E-cadherin signalling in PDAC. Therefore, degrasyn holds promise as cancer therapeutic agent in PDAC with high expressions of USP5 and WT1.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Apoptosis
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Cadherins/antagonists & inhibitors
- Cadherins/genetics
- Cadherins/metabolism
- Carcinoma, Pancreatic Ductal/drug therapy
- Carcinoma, Pancreatic Ductal/metabolism
- Carcinoma, Pancreatic Ductal/secondary
- Cell Proliferation
- Cyanoacrylates/pharmacology
- Deubiquitinating Enzymes/antagonists & inhibitors
- Endopeptidases/chemistry
- Endopeptidases/genetics
- Endopeptidases/metabolism
- Gene Expression Regulation, Neoplastic
- Humans
- Mice
- Mice, Nude
- Neoplasm Invasiveness
- Pancreatic Neoplasms/drug therapy
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/pathology
- Prognosis
- Pyridines/pharmacology
- Tumor Cells, Cultured
- WT1 Proteins/antagonists & inhibitors
- WT1 Proteins/genetics
- WT1 Proteins/metabolism
- Xenograft Model Antitumor Assays
- Pancreatic Neoplasms
Collapse
Affiliation(s)
- Jiajia Li
- Department of Gastroenterologythe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouZhejiang ProvinceChina
| | - Haiying Li
- Laboratory of Internal Medicinethe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouZhejiang ProvinceChina
| | - Weijian Zhu
- Laboratory of Internal Medicinethe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouZhejiang ProvinceChina
| | - Bin Zhou
- Laboratory of Internal Medicinethe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouZhejiang ProvinceChina
| | - Jianchao Ying
- Laboratory of Internal Medicinethe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouZhejiang ProvinceChina
| | - Jiansheng Wu
- Department of Gastroenterologythe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouZhejiang ProvinceChina
| | - Huxiang Zhang
- Pathology Departmentthe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouZhejiang ProvinceChina
| | - Hongwei Sun
- Department of Hepatobiliary and Pancreatic Surgerythe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouZhejiang ProvinceChina
| | - Shenmeng Gao
- Laboratory of Internal Medicinethe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouZhejiang ProvinceChina
| |
Collapse
|
7
|
To KKW, Fong W, Tong CWS, Wu M, Yan W, Cho WCS. Advances in the discovery of microRNA-based anticancer therapeutics: latest tools and developments. Expert Opin Drug Discov 2019; 15:63-83. [PMID: 31739699 DOI: 10.1080/17460441.2020.1690449] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Introduction: MicroRNAs (miRNAs) are small endogenous non-coding RNAs that repress the expression of their target genes by reducing mRNA stability and/or inhibiting translation. miRNAs are known to be aberrantly regulated in cancers. Modulators of miRNA (mimics and antagonists) have emerged as novel therapeutic tools for cancer treatment.Areas covered: This review summarizes the various strategies that have been applied to correct the dysregulated miRNA in cancer cells. The authors also discuss the recent advances in the technical development and preclinical/clinical evaluation of miRNA-based therapeutic agents.Expert opinion: Application of miRNA-based therapeutics for cancer treatment is appealing because they are able to modulate multiple dysregulated genes and/or signaling pathways in cancer cells. Major obstacles hindering their clinical development include drug delivery, off-target effects, efficacious dose determination, and safety. Tumor site-specific delivery of novel miRNA therapeutics may help to minimize off-target effects and toxicity. Combination of miRNA therapeutics with other anticancer treatment modalities could provide a synergistic effect, thus allowing the use of lower dose, minimizing off-target effects, and improving the overall safety profile in cancer patients. It is critical to identify individual miRNAs with cancer type-specific and context-specific regulation of oncogenes and tumor-suppressor genes in order to facilitate the precise use of miRNA anticancer therapeutics.
Collapse
Affiliation(s)
- Kenneth K W To
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Winnie Fong
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Christy W S Tong
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Mingxia Wu
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Wei Yan
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - William C S Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong SAR, China
| |
Collapse
|
8
|
Liu H, Wang X, Zhang H, Wang J, Chen Y, Ma T, Shi J, Kang Y, Xi J, Wang M, Zhang M. Dynamic changes in the level of WT1 as an MRD marker to predict the therapeutic outcome of patients with AML with and without allogeneic stem cell transplantation. Mol Med Rep 2019; 20:2426-2432. [PMID: 31257540 DOI: 10.3892/mmr.2019.10440] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 05/14/2019] [Indexed: 11/06/2022] Open
Abstract
Monitoring minimal residue disease (MRD) is an effective approach to evaluate the response to chemotherapy, and it is used to select the ideal therapeutic strategy and to predict the recurrence during therapy for hematological disorders. The Wilm's tumor 1 (WT1) gene, which is highly expressed in >80% of patients with acute myeloid leukemia (AML) and its increased expression level may cause poor clinical outcomes, is a potential MRD marker of hematological neoplasms. In the present study, the expression levels of WT1 and other molecular markers were retrospectively analyzed by reverse transcription‑quantitative PCR in 195 patients with AML. The expression level of WT1 was significantly lower in patients with remission compared with patients with early‑stage and recurrent AML. Moreover, WT1 expression was significantly decreased in patients with RUNX family transcription factor 1‑RUNX1 translocation partner 1 fusion, but higher in patients with promyelocytic leukemia‑retinoic acid receptor α fusion. WT1 expression was significantly reduced during remission. In patients with AML who underwent allogeneic hematopoietic stem cell transplantation (allo‑HSCT), the mortality rate 2 years after allo‑HSCT was significantly lower in patients with low expression level of WT1 compared with subjects presenting high expression level of WT1. Collectively, the upregulation of the expression level of WT1 in combination with the identification of other genetic abnormalities may be used as MRD markers of hematological neoplasms.
Collapse
Affiliation(s)
- Huasheng Liu
- Department of Hematology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Xiaoning Wang
- Department of Hematology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Hailing Zhang
- Department of Hematology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jincheng Wang
- Department of Hematology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Ying Chen
- Department of Hematology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Tiantian Ma
- Department of Hematology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jing Shi
- Department of Hematology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Ya Kang
- Department of Hematology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jieying Xi
- Department of Hematology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Mengchang Wang
- Department of Hematology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Mei Zhang
- Department of Hematology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
9
|
Chi Y, Luo Q, Song Y, Yang F, Wang Y, Jin M, Zhang D. Circular RNA circPIP5K1A promotes non‐small cell lung cancer proliferation and metastasis through miR‐600/HIF‐1α regulation. J Cell Biochem 2019; 120:19019-19030. [PMID: 31241217 DOI: 10.1002/jcb.29225] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 06/06/2019] [Indexed: 12/18/2022]
Affiliation(s)
- Yongbing Chi
- Clinical LaboratoryShanghai Pudong New Area Gongli Hospital Shanghai China
| | - Qiancheng Luo
- Department of Emergency Medicine, Shanghai Pudong New Area Gongli HospitalThe Second Military Medical University Shanghai China
| | - Yuting Song
- Ningxia Medical University Ningxia China
- Sino‐French Cooperative Central Lab, Shanghai Gongli HospitalSecondary Military Medical University Shanghai China
| | - Fangsong Yang
- Anhui Gaolu Winery Staff Hospital Bozhou Shanghai China
| | - Ying Wang
- Department of Central Laboratory, Shanghai Gongli HospitalThe Second Military Medical University Shanghai China
| | - Mingming Jin
- Department of Central Laboratory, Shanghai Gongli HospitalThe Second Military Medical University Shanghai China
| | - Denghai Zhang
- Department of Central Laboratory, Shanghai Gongli HospitalThe Second Military Medical University Shanghai China
| |
Collapse
|
10
|
Zhou B, Ye H, Xing C, Liang B, Li H, Chen L, Huang X, Wu Y, Gao S. Targeting miR-193a-AML1-ETO-β-catenin axis by melatonin suppresses the self-renewal of leukaemia stem cells in leukaemia with t (8;21) translocation. J Cell Mol Med 2019; 23:5246-5258. [PMID: 31119862 PMCID: PMC6653044 DOI: 10.1111/jcmm.14399] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 04/17/2019] [Accepted: 05/03/2019] [Indexed: 12/12/2022] Open
Abstract
AML1‐ETO, the most common fusion oncoprotein by t (8;21) in acute myeloid leukaemia (AML), enhances hematopoietic self‐renewal and leukemogenesis. However, currently no specific therapies have been reported for t (8;21) AML patients as AML1‐ETO is still intractable as a pharmacological target. For this purpose, leukaemia cells and AML1‐ETO‐induced murine leukaemia model were used to investigate the degradation of AML1‐ETO by melatonin (MLT), synthesized and secreted by the pineal gland. MLT remarkedly decreased AML1‐ETO protein in leukemic cells. Meanwhile, MLT induced apoptosis, decreased proliferation and reduced colony formation. Furthermore, MLT reduced the expansion of human leukemic cells and extended the overall survival in U937T‐AML1‐ETO‐xenografted NSG mice. Most importantly, MLT reduced the infiltration of leukaemia blasts, decreased the frequency of leukaemia stem cells (LSCs) and prolonged the overall survival in AML1‐ETO‐induced murine leukaemia. Mechanistically, MLT increased the expression of miR‐193a, which inhibited AML1‐ETO expression via targeting its putative binding sites. Furthermore, MLT decreased the expression of β‐catenin, which is required for the self‐renewal of LSC and is the downstream of AML1‐ETO. Thus, MLT presents anti‐self‐renewal of LSC through miR‐193a‐AML1‐ETO‐β‐catenin axis. In conclusion, MLT might be a potential treatment for t (8;21) leukaemia by targeting AML1‐ETO oncoprotein.
Collapse
Affiliation(s)
- Bin Zhou
- Laboratory of Internal Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Haige Ye
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chongyun Xing
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Bin Liang
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Haiying Li
- Laboratory of Internal Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Linling Chen
- Department of Clinical Laboratory, The People's Hospital of Yuhuang County, Taizhou, China
| | - Xingzhou Huang
- Laboratory of Internal Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yanfei Wu
- Laboratory of Internal Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shenmeng Gao
- Laboratory of Internal Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
11
|
Miroshnichenko S, Patutina O. Enhanced Inhibition of Tumorigenesis Using Combinations of miRNA-Targeted Therapeutics. Front Pharmacol 2019; 10:488. [PMID: 31156429 PMCID: PMC6531850 DOI: 10.3389/fphar.2019.00488] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 04/17/2019] [Indexed: 12/18/2022] Open
Abstract
The search for effective strategies to inhibit tumorigenesis remains one of the most relevant scientific challenges. Among the most promising approaches is the direct modulation of the function of short non-coding RNAs, particularly miRNAs. These molecules are propitious targets for anticancer therapy, since they perform key regulatory roles in a variety of signaling cascades related to cell proliferation, apoptosis, migration, and invasion. The development of pathological states is often associated with deregulation of miRNA expression. The present review describes in detail the strategies aimed at modulating miRNA activity that invoke antisense oligonucleotide construction, such as small RNA zippers, miRNases (miRNA-targeted artificial ribonucleases), miRNA sponges, miRNA masks, anti-miRNA oligonucleotides, and synthetic miRNA mimics. The broad impact of developed miRNA-based therapeutics on the various events of tumorigenesis is also discussed. Above all, the focus of this review is to evaluate the results of the combined application of different miRNA-based agents and chemotherapeutic drugs for the inhibition of tumor development. Many studies indicate a considerable increase in the efficacy of anticancer therapy as a result of additive or synergistic effects of simultaneously applied therapies. Different drug combinations, such as a cocktail of antisense oligonucleotides or multipotent miRNA sponges directed at several oncogenic microRNAs belonging to the same/different miRNA families, a mixture of anti-miRNA oligonucleotides and cytostatic drugs, and a combination of synthetic miRNA mimics, have a more complex and profound effect on the various events of tumorigenesis as compared with treatment with a single miRNA-based agent or chemotherapeutic drug. These data provide strong evidence that the simultaneous application of several distinct strategies aimed at suppressing different cellular processes linked to tumorigenesis is a promising approach for cancer therapy.
Collapse
Affiliation(s)
- Svetlana Miroshnichenko
- Laboratory of Nucleic Acids Biochemistry, Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk, Russia
| | - Olga Patutina
- Laboratory of Nucleic Acids Biochemistry, Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk, Russia
| |
Collapse
|
12
|
Wei W, Huo B, Shi X. miR-600 inhibits lung cancer via downregulating the expression of METTL3. Cancer Manag Res 2019; 11:1177-1187. [PMID: 30774445 PMCID: PMC6362936 DOI: 10.2147/cmar.s181058] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background Methyltransferase like 3 (METTL3) is an RNA methyltransferase implicated in mRNA biogenesis, decay, and translation control through N6-methyladenosine (m6A) modification. Methods To find new treatment strategies for lung cancer and to elucidate the mechanism underlying the phenomenon, we treated the human lung cancer cell lines A549 and H1299 to investigate the effect of METTL3 on lung cancer. Results We observed that knockdown of METTL3 inhibited the survival and proliferation of A549 and H1299 cells. The migration and proliferation of both cell lines were significantly decreased, and the apoptosis was induced in comparison with control cells. These results were further confirmed by the transfection of miRNA of METTL3 increased the Bax/Bcl-2 ratio in A549 and H1299 cells, which is a sign that mitochondrial apoptotic pathway was triggered. The PI3K/Akt pathway is implicated in cell growth and survival and we also observed that knockdown of METTL3 changed the expression and phosphorylation of proteins of PI3K signaling pathway members. Further, our results demonstrated that miR-600 inhibited the expression of METTL3 and reversed the positive effect of METTL3 on NSCLC progression, indicating an miR-600/METTL3 pathway in NSCLC. Conclusion These data suggested that miR-600 inhibited lung cancer via down-regulating METTL3 expression, and knockdown of METTL3 might be used as a novel strategy for lung cancer therapy.
Collapse
Affiliation(s)
- Wenwen Wei
- Department of Respiratory Medicine, The Second People's Hospital of Dongying, Guangrao City, Shandong Province 257335, People's Republic of China,
| | - Baosheng Huo
- Department of Thoracic Surgery, The Second People's Hospital of Dongying, Guangrao City, Shandong Province 257335, People's Republic of China,
| | - Xiulan Shi
- Department of Respiratory Medicine, The Second People's Hospital of Dongying, Guangrao City, Shandong Province 257335, People's Republic of China,
| |
Collapse
|
13
|
Szymczyk A, Macheta A, Podhorecka M. Abnormal microRNA expression in the course of hematological malignancies. Cancer Manag Res 2018; 10:4267-4277. [PMID: 30349361 PMCID: PMC6183594 DOI: 10.2147/cmar.s174476] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Research on the carcinogenesis process is currently focused primarily on understanding its genetic basis and molecular abnormalities that may be predictive factors and therapeutic targets. It was clearly confirmed recently that microRNAs are involved in the mechanisms of leukocyte development, differentiation, and apoptosis, as well as in the pathogenesis of proliferative diseases of the hematopoietic system. Currently, research strategies allow determination of the deregulation of microRNA profiles in relation to other cytogenetic aberrations, as well as prognostic factors and primary end points. The problem of the possibility of their use as therapeutic targets is also increasingly discussed. In this article, we analyze literature data on abnormalities in microRNA expression in proliferative diseases of the hematopoietic system in the context of classic cytogenetic and molecular aberrations.
Collapse
Affiliation(s)
- Agnieszka Szymczyk
- Independent Clinical Transplantology Unit, Medical University of Lublin, Lublin, Poland,
| | - Arkadiusz Macheta
- Department of Haematooncology and Bone Marrow Transplantation, Medical University of Lublin, Lublin, Poland
| | - Monika Podhorecka
- Department of Haematooncology and Bone Marrow Transplantation, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
14
|
Sun J, Hu J, Wang G, Yang Z, Zhao C, Zhang X, Wang J. LncRNA TUG1 promoted KIAA1199 expression via miR-600 to accelerate cell metastasis and epithelial-mesenchymal transition in colorectal cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:106. [PMID: 29776371 PMCID: PMC5960095 DOI: 10.1186/s13046-018-0771-x] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 04/25/2018] [Indexed: 12/25/2022]
Abstract
BACKGROUND LncRNA TUG1 has been reported to be highly expressed in CRC samples and cells and promoted metastasis by affecting EMT, indicating a poor prognosis for colorectal cancer (CRC). In this study, we determined the underlying mechanism for tumor oncogenesis of lncRNA TUG1 in CRC metastasis. METHODS The expressions of miR-600 and KIAA1199 in 76 CRC patients and CRC cells and CRC metastatic tissues were determined using qRT-PCR. Epithelial-mesenchymal transition (EMT)-related proteins were determined using western blot. CRC cell metastasis was assessed by colony formation, wound healing and transwell assay. Luciferase reporter gene assay was used to confirm miR-600 binding to KIAA1199 3'UTR. RESULTS Our data showed that lncRNA TUG1 was upregulated in CRC cells, miR-600 was downregulated in CRC tissues, cell lines and CRC metastatic tissues, and low miR-600 expression predicted a poor clinical prognosis. Overexpression of miR-600 suppressed CRC cell migration/invasion and EMT-related proteins in vitro, inhibited tumor volume and weight, and decreased the number of CRC liver metastasis in vivo. KIAA1199 was upregulated in CRC tissues, and was negatively regulated by miR-600. KIAA1199 overexpression promoted CRC cell migration and invasion, which reversed the inhibition effect of miR-600 mimic on migration and invasion of CRC cells. Moreover, TUG1 negatively regulated miR-600, and inhibition of TUG1 suppressed CRC cell migration and invasion and EMT-related proteins via regulating miR-600. CONCLUSION Our study proved that TUG1 promoted KIAA1199 expression to accelerate EMT and metastasis of CRC cell through inhibition of miR-600 expression.
Collapse
Affiliation(s)
- Junfeng Sun
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East, Zhengzhou, 450052, China.
| | - Jiyi Hu
- Colorectal Cancer Center, Fudan University, Shanghai, China
| | - Guojun Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East, Zhengzhou, 450052, China
| | - Zhen Yang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East, Zhengzhou, 450052, China
| | - Chunlin Zhao
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East, Zhengzhou, 450052, China
| | - Xiefu Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East, Zhengzhou, 450052, China
| | - Jiaxiang Wang
- Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|