1
|
Zhu J, Yu Z, Wang X, Zhang J, Chen Y, Chen K, Zhang B, Sun J, Jiang J, Zheng S. LncRNA MACC1-AS1 induces gemcitabine resistance in pancreatic cancer cells through suppressing ferroptosis. Cell Death Discov 2024; 10:101. [PMID: 38413579 PMCID: PMC10899202 DOI: 10.1038/s41420-024-01866-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 01/28/2024] [Accepted: 02/13/2024] [Indexed: 02/29/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDA) mortality is primarily attributed to metastasis and chemotherapy resistance. In this research, the long non-coding RNA MACC1-AS1 was studied, playing a significant role in regulating lipid oxidation processes. This regulation could further lead to the inhibition of ferroptosis induced by chemotherapeutic drugs, making it a contributing factor to gemcitabine resistance in PDA. In both gemcitabine-resistant PDA patients and mouse models, the elevated expression level of MACC1-AS1 in the tumors was noted. Additionally, overexpression of MACC1-AS1 in pancreatic cancer cells was found to enhance tolerance to gemcitabine and suppress ferroptosis. Proteomic analysis of drug-resistant pancreatic cells revealed that overexpressed MACC1-AS1 inhibited the ubiquitination degradation of residues in the protein kinase STK33 by MDM4. Furthermore, its accumulation in the cytoplasm activated STK33, further activating the ferroptosis-suppressing proteins GPX4, thereby counteracting gemcitabine-induced cellular oxidative damage. These findings suggested that the long non-coding RNA MACC1-AS1 could play a significant role in the ability of pancreatic cancer cells to evade iron-mediated ferroptosis induced by gemcitabine. This discovery holds promise for developing clinical therapeutic strategies to combat chemotherapy resistance in pancreatic cancer.
Collapse
Affiliation(s)
- Jiyun Zhu
- Hepatopancreatobiliary Surgery Department, The First Affiliated Hospital of Ningbo University, Ningbo, China
- FUJIAN Medical University, Fuzhou, China
| | - Zehao Yu
- Health Science Center, Ningbo University, Ningbo, China
| | - Xuguang Wang
- Emergency Department, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Jinghui Zhang
- Health Science Center, Ningbo University, Ningbo, China
| | - Yi Chen
- Emergency Department, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Kaibo Chen
- Department of Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Bin Zhang
- Hepatopancreatobiliary Surgery Department, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Jianhong Sun
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Ningbo University, Ningbo, China.
| | - Jianshuai Jiang
- Hepatopancreatobiliary Surgery Department, The First Affiliated Hospital of Ningbo University, Ningbo, China.
| | - Siming Zheng
- Hepatopancreatobiliary Surgery Department, The First Affiliated Hospital of Ningbo University, Ningbo, China.
- FUJIAN Medical University, Fuzhou, China.
| |
Collapse
|
2
|
Jiang H, Li L, Ma T, Wang R, Chen X, Xu K, Chen C, Liu Z, Wang H, Huang L. Serine/Threonine Kinase (STK) 33 promotes the proliferation and metastasis of human esophageal squamous cell carcinoma via inflammation-related pathway. Pathol Res Pract 2024; 254:155154. [PMID: 38286054 DOI: 10.1016/j.prp.2024.155154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 11/16/2023] [Accepted: 01/18/2024] [Indexed: 01/31/2024]
Abstract
The serine/threonine kinase (STK) 33 plays a key role in cancer cell proliferation and metastasis. Abnormal STK33 expression is closely related to malignancy of numerous cancers. This study suggests the important role of STK33 in the pathogenesis and metastatic progression of esophageal squamous cell carcinoma (ESCC). STK33 expression in human ESCC tissues was detected by immunohistochemical technique. Further, we analyzed the relationship between STK33 and clinical and pathological factors as well as the prognosis of patients. ECa109 cell line was cultured and transfected with STK33-RNAi lentiviral vector to perform Hochest33342 & PI and metastasis experiments. The TCGA database was used to analyze the STK33 expression level in ESCC. All statistical analyses were performed in SPSS 23.0 software. Differences with P < 0.05 were considered statistically significant. In human ESCC specimens, STK33 was overexpressed and associated with poor prognosis. Silencing STK33 expression suppressed ESCC proliferation, migration, invasion, and tumor growth. STK33 also mediated angiogenesis, TGFβ, and inflammatory response in ESCC. Mechanistic investigations revealed that STK33 regulates ESCC through multiple complex pathways. Dysregulated STK33 signaling promotes ESCC growth and progression. Thus, our findings identified STK33 as a candidate treatment target that improves ESCC therapy.
Collapse
Affiliation(s)
- Haifeng Jiang
- Department of Pathology, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Liping Li
- Public Health and Management College in Ningxia Medical University, Yinchuan 750004, China
| | - Tao Ma
- Department of Pathology, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Ruixiao Wang
- Department of Pathology, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Xiaozhen Chen
- Department of Pathology, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Ke Xu
- Department of Pathology, General Hospital of Ningxia Medical University, Yinchuan 750004, China; Ningxia Armed Police Corps, Yinchuan 750004, China
| | - Chen Chen
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, China; Department of Pathology and Pathophysiology, Shandong University, Cheeloo Healthy Science Center, Jinan 250012, China
| | - Zijin Liu
- Clinical Medical College in Ningxia Medical University, Yinchuan 750004, China
| | - Hongmei Wang
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China; School of Medicine, Shanxi University of Chinese Medicine, Xi'an 712046, China.
| | - Lingyan Huang
- Department of Pathology, General Hospital of Ningxia Medical University, Yinchuan 750004, China.
| |
Collapse
|
3
|
Le MT, Nguyen HT, Nguyen XH, Do XH, Mai BT, Ngoc Nguyen HT, Trang Than UT, Nguyen TH. Regulation and therapeutic potentials of microRNAs to non-small cell lung cancer. Heliyon 2023; 9:e22080. [PMID: 38058618 PMCID: PMC10696070 DOI: 10.1016/j.heliyon.2023.e22080] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 11/02/2023] [Accepted: 11/03/2023] [Indexed: 12/08/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) is the most common type of lung cancer, accounting for 80%-85% of total cases and leading to millions of deaths worldwide. Drug resistance is the primary cause of treatment failure in NSCLC, which urges scientists to develop advanced approaches for NSCLC treatment. Among novel approaches, the miRNA-based method has emerged as a potential approach as it allows researchers to modulate target gene expression. Subsequently, cell behaviors are altered, which leads to the death and the depletion of cancer cells. It has been reported that miRNAs possess the capacity to regulate multiple genes that are involved in various signaling pathways, including the phosphoinositide 3-kinase, receptor tyrosine kinase/rat sarcoma virus/mitogen-activated protein kinase, wingless/integrated, retinoblastoma, p53, transforming growth factor β, and nuclear factor-kappa B pathways. Dysregulation of these signaling pathways in NSCLC results in abnormal cell proliferation, tissue invasion, and drug resistance while inhibiting apoptosis. Thus, understanding the roles of miRNAs in regulating these signaling pathways may enable the development of novel NSCLC treatment therapies. However, a comprehensive review of potential miRNAs in NSCLC treatment has been lacking. Therefore, this review aims to fill the gap by summarizing the up-to-date information on miRNAs regarding their targets, impact on cancer-associated pathways, and prospective outcomes in treating NSCLC. We also discuss current technologies for delivering miRNAs to the target cells, including virus-based, non-viral, and emerging extracellular vesicle-based delivery systems. This knowledge will support future studies to develop an innovative miRNA-based therapy and select a suitable carrier to treat NSCLC effectively.
Collapse
Affiliation(s)
- Mai Thi Le
- Vinmec Hi-tech Center, Vinmec Healthcare System, Hanoi, 100000, Viet Nam
- Faculty of Biology, VNU University of Science, Vietnam National University, Hanoi, 100000, Viet Nam
| | - Huyen-Thu Nguyen
- Vinmec Hi-tech Center, Vinmec Healthcare System, Hanoi, 100000, Viet Nam
| | - Xuan-Hung Nguyen
- Vinmec Hi-tech Center, Vinmec Healthcare System, Hanoi, 100000, Viet Nam
- College of Health Sciences, Vin University, Hanoi, 100000, Viet Nam
- Vinmec-VinUni Institute of Immunology, Vinmec Healthcare System, Hanoi, 100000, Viet Nam
| | - Xuan-Hai Do
- Department of Gastroenterology, 108 Military Central Hospital, Hanoi, Viet Nam
| | - Binh Thanh Mai
- Department of Practical and Experimental Surgery, Vietnam Military Medical University, 160 Phung Hung Street, Phuc La, Ha Dong, Hanoi, Viet Nam
| | - Ha Thi Ngoc Nguyen
- Vinmec Hi-tech Center, Vinmec Healthcare System, Hanoi, 100000, Viet Nam
| | - Uyen Thi Trang Than
- Vinmec Hi-tech Center, Vinmec Healthcare System, Hanoi, 100000, Viet Nam
- Vinmec-VinUni Institute of Immunology, Vinmec Healthcare System, Hanoi, 100000, Viet Nam
| | - Thanh-Hong Nguyen
- Vinmec Hi-tech Center, Vinmec Healthcare System, Hanoi, 100000, Viet Nam
| |
Collapse
|
4
|
Brown JS. Comparison of Oncogenes, Tumor Suppressors, and MicroRNAs Between Schizophrenia and Glioma: The Balance of Power. Neurosci Biobehav Rev 2023; 151:105206. [PMID: 37178944 DOI: 10.1016/j.neubiorev.2023.105206] [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: 11/29/2022] [Revised: 04/25/2023] [Accepted: 04/30/2023] [Indexed: 05/15/2023]
Abstract
The risk of cancer in schizophrenia has been controversial. Confounders of the issue are cigarette smoking in schizophrenia, and antiproliferative effects of antipsychotic medications. The author has previously suggested comparison of a specific cancer like glioma to schizophrenia might help determine a more accurate relationship between cancer and schizophrenia. To accomplish this goal, the author performed three comparisons of data; the first a comparison of conventional tumor suppressors and oncogenes between schizophrenia and cancer including glioma. This comparison determined schizophrenia has both tumor-suppressive and tumor-promoting characteristics. A second, larger comparison between brain-expressed microRNAs in schizophrenia with their expression in glioma was then performed. This identified a core carcinogenic group of miRNAs in schizophrenia offset by a larger group of tumor-suppressive miRNAs. This proposed "balance of power" between oncogenes and tumor suppressors could cause neuroinflammation. This was assessed by a third comparison between schizophrenia, glioma and inflammation in asbestos-related lung cancer and mesothelioma (ALRCM). This revealed that schizophrenia shares more oncogenic similarity to ALRCM than glioma.
Collapse
|
5
|
Li X, Lin M, Liu M, Ye H, Qin S. Interaction between STK33 and autophagy promoted renal cell carcinoma metastasis by regulating mTOR/ULK1 signaling pathway. Mol Biol Rep 2023; 50:5059-5067. [PMID: 37101009 DOI: 10.1007/s11033-023-08396-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 03/17/2023] [Indexed: 04/28/2023]
Abstract
BACKGROUND The roles of STK33 in renal cell carcinoma (RCC) remain unclear. This study was designed to investigate the interaction between STK33 and the autophagy in the RCC. METHODS AND RESULTS STK33 was knocked down in 786-O and CAKI-1 cells. Then CCK8, clony formation assay, wound healing assay and Transwell assay were performed to analyze the proliferation, migration and invasion of the cancer cells. In addition, the activation of autophagy was determined using fluorescence, followed by investigating the potential signaling pathways in this process. After STK33 knockdown, the proliferation and migration of cell lines were inhibited, and the apoptosis of renal cancer cells was promoted. Autophagy fluorescence experiment showed that after STK33 knockdown, green LC3 protein fluorescence particles could be seen in the cells. Western blot analysis showed that after STK33 knockdown, there was significant down-regulation in P62 and p-mTOR, as well as significant up-regulation of Beclin1, LC3 and p-ULK1. CONCLUSIONS STK33 affected autophagy in RCC cells by activating mTOR/ ULK1pathway.
Collapse
Affiliation(s)
- Xiaomei Li
- Department of Pathology, The Affiliated Taian City Central Hospital of Qingdao University, No. 29, Longtan Road, Taishan District, Taian, 271000, China.
| | - Min Lin
- Department of Pathology, The Affiliated Taian City Central Hospital of Qingdao University, No. 29, Longtan Road, Taishan District, Taian, 271000, China
| | - Min Liu
- Department of Pathology, The Affiliated Taian City Central Hospital of Qingdao University, No. 29, Longtan Road, Taishan District, Taian, 271000, China
| | - Hong Ye
- Department of Pathology, The Affiliated Taian City Central Hospital of Qingdao University, No. 29, Longtan Road, Taishan District, Taian, 271000, China
| | - Shuming Qin
- Department of Pathology, The Affiliated Taian City Central Hospital of Qingdao University, No. 29, Longtan Road, Taishan District, Taian, 271000, China
| |
Collapse
|
6
|
Effects of miR-107 on Breast Cancer Cell Growth and Death via Regulation of the PTEN/AKT Signaling Pathway. JOURNAL OF ONCOLOGY 2023; 2023:1244067. [PMID: 36816358 PMCID: PMC9931464 DOI: 10.1155/2023/1244067] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/19/2022] [Accepted: 08/26/2022] [Indexed: 02/10/2023]
Abstract
Objective Investigate the influence of miR-107 on breast cancer cell growth and death through the PTEN/AKT signaling pathway. Method As study subjects, the human breast cancer cell line MCF-7 and the normal breast cell line Hs 578Bst were chosen, and MCF-7 cells were, respectively, transfected with control miRNA and miR-107 inhibitor. CCK-8, flow cytometry, scratch assay, and Transwell assay were used to analyze the proliferation, apoptosis, and invasion, and in order to identify the proteins associated with apoptosis in each of the three categories, we used western blot analysis. Bcl-2, cleaved caspase-3, and cleaved caspase-9 expression, as well as PTEN/AKT signaling pathway-associated protein expression, are correlated. Result The expression of miR-107 in MCF-7 cells was significantly greater than that in Hs 578Bst cells, with a P < 0.05 difference; compared to the blank and miRNA control groups, the miR-107 inhibitor group had a P < 0.05 difference. P < 0.05 showed a decrease in proliferation (42.52) but no difference in proliferation between the blank and miRNA control groups (P > 0.05); the miR-107 inhibitor group had higher apoptosis (38.96) with P < 0.05 than the blank group (4.85) and the miRNA control group (5.89); there was no difference in apoptosis between the blank and miRNA groups (P > 0.05). There was no significant difference between the blank group and the miRNA control group with P > 0.05; compared with the blank group, the miR-107 inhibitor group had a lower expression of Bcl-2 protein (0.18), in addition to the degraded paradigms (0.73) and caspase-9 protein concentrations (0.79), respectively. Conclusion The PTEN/AKT signaling pathway may be regulated by miR-107 to limit breast cancer cell growth and increase apoptosis, which suggests that miR-107 may be exploited as a tumor marker for therapeutic therapy.
Collapse
|
7
|
circCCT3 Enhances Invasion and Epithelial-Mesenchymal Transition (EMT) of Non-Small-Cell Lung Cancer (NSCLC) via the miR-107/Wnt/FGF7 Axis. JOURNAL OF ONCOLOGY 2022; 2022:7020774. [PMID: 35783154 PMCID: PMC9242791 DOI: 10.1155/2022/7020774] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 05/31/2022] [Accepted: 06/02/2022] [Indexed: 11/17/2022]
Abstract
Background. CircRNAs play a role in a variety of biological processes, including tumorigenesis. circCCT3 has been shown to regulate cancer initiation and progression. Unfortunately, whether circCCT3 is involved in non-small-cell lung cancer (NSCLC) metastasis remains unclear. Methods. Our study utilized RT-qPCR to examine gene expression levels. A transwell assay was used to measure invasion ability of cells. Starbase software and TargetScan software were used to predict target genes. Results. circCCT3 knockdown attenuated invasion and epithelial-mesenchymal transition (EMT) of A549 and Calu-1 cells. miR-107 mimics could rescue circCCT3-induced invasion and EMT. Next, miR-107 mimics and circCCT3 knockdown suppressed Wnt3a and FGF7 expression. An miR-107 inhibitor promoted Wnt3a and FGF7 expressions. Finally, FGF7 greatly restored miR-107-inhibited invasion and EMT of A549 cells. Conclusion. Here, we reveal a molecular mechanism circCCT3/miR-107/Wnt/FGF7 responsible for NSCLC metastasis.
Collapse
|
8
|
Yan J, Dai L, Yuan J, Pang M, Wang Y, Lin L, Shi Y, Wu F, Nie R, Chen Q, Wang L. miR-107 Inhibits the Proliferation of Gastric Cancer Cells In vivo and In vitro by Targeting TRIAP1. Front Genet 2022; 13:855355. [PMID: 35480301 PMCID: PMC9035523 DOI: 10.3389/fgene.2022.855355] [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: 01/15/2022] [Accepted: 03/03/2022] [Indexed: 11/13/2022] Open
Abstract
Gastric cancer is a kind of gastrointestinal tumor with high morbidity and mortality. Finding effective methods for early diagnosis and treatment of gastric cancer has important significance and application prospects. MicroRNAs without protein coding potential affect the occurrence and development of gastric cancer. This study aims to explore the biological function and mechanism of microRNA-107 (miR-107) in gastric cancer. The results show that miR-107 is low expressed in gastric cancer, while TRIAP1 is highly expressed; the overexpression of miR-107 can inhibit the progression of gastric cancer in vivo and in vitro, while the overexpression plasmid of TRIAP1 can restore the miR-107 mimic-induced cell proliferation and metastasis inhibition, and the small interfering RNA of TRIAP1 can inhibit the cell proliferation and invasion induced by miR-107 inhibitor. In conclusion, the results of this study show that miR-107 can inhibit the proliferation of gastric cancer in vivo and in vitro by targeting TRIAP1.
Collapse
Affiliation(s)
- Jiexin Yan
- Emergency of Department, Changhai Hospital, Shanghai, China
| | - Lu Dai
- The Fourth Outpatient Department, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
| | - Jun Yuan
- The Fourth Outpatient Department, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
| | - Min Pang
- The Fourth Outpatient Department, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
| | - Yueqiu Wang
- The Fourth Outpatient Department, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
| | - Lang Lin
- Emergency of Department, Changhai Hospital, Shanghai, China
| | - Yawei Shi
- Department of Oral and Maxilliofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
| | - Fuli Wu
- Department of Oral and Maxilliofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
| | - Rongping Nie
- The Seventh Outpatient Department, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
| | - Qiuling Chen
- Cangnan Hospital Affiliated to Wenzhou Medical University, Wenzhou, China
- *Correspondence: Qiuling Chen, ; Lei Wang,
| | - Lei Wang
- Department of Gastroenterology, Changhai Hospital, Shanghai, China
- *Correspondence: Qiuling Chen, ; Lei Wang,
| |
Collapse
|
9
|
Feng L, Xu X, Zhao K. NFYB potentiates STK33 activation to promote cisplatin resistance in diffuse large B-cell lymphoma. Leuk Res 2021; 111:106708. [PMID: 34536775 DOI: 10.1016/j.leukres.2021.106708] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 08/30/2021] [Accepted: 09/08/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND The aberrant expression of serine/threonine kinase 33 (STK33) has been implicated in cancers. However, its clinical significance and biological functions in diffuse large B cell lymphoma (DLBCL) remain largely unknown. In the present investigation, we delineated the expression of STK33 in DLBCL and its function in cisplatin resistance. METHODS First, genes associated with drug resistance as well as occurrence in DLBCL were analyzed by bioinformatics, followed by correlation analysis between STK33 expression and clinical baseline information of patients with DLBCL. Further, cisplatin-resistant DLBCL cell lines were constructed, and changes in cell sensitivity to cisplatin treatment were examined after interfering the expression of STK33 in parental cells as well as in drug-resistant cells, respectively. Subsequently, the downstream signaling pathways of STK33 were analyzed. Finally, the upstream regulatory mechanism of STK33 was predicted by bioinformatics as well as experimentally validated. RESULTS STK33 was overexpressed in the patients with DLBCL as well as in cisplatin-resistant DLBCL cells, and knockdown of STK33 significantly promoted sensitivity of resistant cells to cisplatin. Moreover, our further analysis revealed that STK33 promoted cisplatin resistance in DLBCL by activating the Hedgehog signaling pathway. We found in subsequent experiments that nuclear transcription factor Y subunit beta (NFYB) can bind to the STK33 promoter and thus promote STK33 expression. CONCLUSIONS The transcription factor NFYB expedites the transcription of SYK33 by binding to the STK33 promoter, thereby activating the Hedgehog signaling pathway in DLBCL cells, which in turn promotes the resistance of DLBCL cells to cisplatin.
Collapse
Affiliation(s)
- Lili Feng
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, PR China
| | - Xiaofang Xu
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, PR China.
| | - Keke Zhao
- Department of Ophthalmology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, PR China.
| |
Collapse
|
10
|
Huang S, Luo S, Gong C, Liang L, Xiao Y, Li M, He J. MTTL3 upregulates microRNA-1246 to promote occurrence and progression of NSCLC via targeting paternally expressed gene 3. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 24:542-553. [PMID: 33898106 PMCID: PMC8059086 DOI: 10.1016/j.omtn.2021.02.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 02/19/2021] [Indexed: 12/11/2022]
Abstract
Non-small cell lung cancer (NSCLC) is one of the major causes of morbidity and mortality worldwide. We aimed to investigate the role of N6-methyladenosine (m6A) methyltransferase-like 3 (METTL3) regulating microRNA-1246 (miR-1246) in the progression of NSCLC by targeting paternally expressed gene 3 (PEG3). METTL3, miR-1246, and PEG3 expression in tissues was assessed, and the predictive role of METTL3 in prognosis of patients with NSCLC was detected. NSCLC cells were relatively treated with altered expression of METTL3, miR-1246, or PEG3 to measure their roles in the proliferation, migration, invasion, apoptosis, and in vivo growth of the NSCLC cells. The RNA m6A level was determined, and the targeting relationship between miR-1246 and PEG3 was confirmed. Our results revealed that METTL3 and miR-1246 were upregulated, whereas PEG3 was downregulated in NSCLC tissues. METTL3 knockdown or PEG3 overexpression in NSCLC cells suppressed malignant behaviors of NSCLC cells. METTL3 affected the m6A modification of miR-1246, thus upregulating miR-1246 and miR-1246-targeted PEG3. The elevation of PEG3 reversed the effects of miR-1246 upregulation on NSCLC cells. This study revealed that m6A methyltransferase METTL3 affects the m6A modification of miR-1246, thus upregulating miR-1246 to promote NSCLC progression by inhibiting PEG3.
Collapse
Affiliation(s)
- Shaohong Huang
- Department of Thoracocardiac Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, Guangdong, China
| | - Shaoning Luo
- Department of Emergency Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, Guangdong, China
| | - Chulian Gong
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, Guangdong, China
| | - Limin Liang
- Department of Thoracocardiac Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, Guangdong, China
| | - Yi Xiao
- Department of Thoracocardiac Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, Guangdong, China
| | - Mingan Li
- Department of Interventional Radiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, Guangdong, China
| | - Jinyuan He
- Department of Thoracocardiac Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, Guangdong, China
| |
Collapse
|