1
|
Zhu W, Yang W, Sun G, Huang J. RNA-binding protein quaking: a multifunctional regulator in tumour progression. Ann Med 2025; 57:2443046. [PMID: 39711373 DOI: 10.1080/07853890.2024.2443046] [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: 05/06/2024] [Revised: 10/03/2024] [Accepted: 11/22/2024] [Indexed: 12/24/2024] Open
Abstract
BACKGROUND Quaking (QKI) is a member of the signal transduction and activators of RNA (STAR) family, performing a crucial multifunctional regulatory role in alternative splicing, mRNA precursor processing, mRNA transport and localization, mRNA stabilization, and translation during tumour progression. Abnormal QKI expression or fusion mutations lead to aberrant RNA and protein expression, thereby promoting tumour progression. However, in many types of tumour, QKI played a role as tumour suppressor, the regulatory role of QKI in tumour progression remains ambiguous. OBJECTIVES This review aims to analyze the isoform and function of QKI, the impact of QKI-regulated gene expression or signalling pathway alterations on tumour progression, and its potential clinical applications as a predictive marker or target for tumour therapy. METHODS We reviewed recent studies and summarized the function of QKI alteration in tumour progression. RESULTS QKI mediate post-transcriptional gene regulation including alternative splicing, polyadenylation, mRNA stabilization, mRNA subcellular location, and noncoding RNA by binding to the QRE elements of targeted nucleotide. The dysregulation of QKI is intricately correlated to tumour proliferation, metastasis, angiogenesis, tumor stem cells, the tumour microenvironment, and treatment sensitivity, and represents as a potential biological predictor in tumour diagnosis and prognosis. CONCLUSIONS QKI play a critical role as tumour suppressor or an oncogene in tumour progression due to the different splicing sites and transcripts with various tumour subtype or tumor micorenvironment. Ongoing research about QKI's functions and mechanisms persist is required to conduct for better understanding the role of QKI in tumour regulation.
Collapse
Affiliation(s)
- Wangyu Zhu
- Cell and Molecular Biology Laboratory, Zhoushan Hospital of Wenzhou Medical University, Zhoushan, Zhejiang, China
- Lung Cancer Research Centre, Zhoushan Hospital of Wenzhou Medical, Zhoushan, Zhejiang, China
| | - Weiwei Yang
- Cell and Molecular Biology Laboratory, Zhoushan Hospital of Wenzhou Medical University, Zhoushan, Zhejiang, China
- Lung Cancer Research Centre, Zhoushan Hospital of Wenzhou Medical, Zhoushan, Zhejiang, China
| | - Guoping Sun
- Department of Breast Surgery, Second Affiliated Hospital and Cancer Institute (Provincial Key Laboratory of Tumor Microenvironment and Immunotherapy, Key Laboratory of Cancer Prevention & Intervention, National Ministry of Education), Zhejiang University School of Medicine, Hangzhou, China
| | - Jian Huang
- Department of Breast Surgery, Second Affiliated Hospital and Cancer Institute (Provincial Key Laboratory of Tumor Microenvironment and Immunotherapy, Key Laboratory of Cancer Prevention & Intervention, National Ministry of Education), Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
2
|
Zhao M, Jin Y, Yan Z, He C, You W, Zhu Z, Wang R, Chen Y, Luo J, Zhang Y, Yao Y. The splicing factor QKI inhibits metastasis by modulating alternative splicing of E-Syt2 in papillary thyroid carcinoma. Cancer Lett 2024; 604:217270. [PMID: 39306227 DOI: 10.1016/j.canlet.2024.217270] [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: 03/18/2024] [Revised: 08/27/2024] [Accepted: 09/18/2024] [Indexed: 09/28/2024]
Abstract
Alternative splicing (AS) plays a crucial role in the hallmarks of cancer and can open new avenues for targeted therapies. However, the aberrant AS events and the metastatic cascade in papillary thyroid carcinoma (PTC) remain largely unclear. Here, we identify the splicing factor, quaking protein (QKI), which was significantly downregulated in PTC and correlated with poor survival outcomes in patients with PTC. Functional studies indicated that low expression of QKI promoted the PTC cell growth and metastasis in vitro and in vivo. Mechanistically, low QKI induced exon 14 retention of extended synaptotagmin 2 (E-Syt2) and produced a long isoform transcript (termed E-Syt2L) that acted as an important oncogenic factor of PTC metastasis. Notably, overexpression of long non-coding RNA eosinophil granule ontogeny transcript (EGOT) physically binds to QKI and suppressed its activity by inhibiting ubiquitin specific peptidase 25 (USP25) mediated deubiquitination and subsequent degradation of QKI. Collectively, these data demonstrate the novel mechanistic links between the splicing factor QKI and splicing event in PTC metastasis and support the potential utility of targeting splicing events as a therapeutic strategy for PTC.
Collapse
Affiliation(s)
- Mengya Zhao
- Department of Head and Neck Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University & The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center Nanjing, Nanjing Medical University, Nanjing, China; Wuxi People's Hospital, Wuxi Medical Center Nanjing & Department of Immunology, School of Basic Medical Science & Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China; The Affiliated Huai'an No. 1 People's Hospital, Nanjing Medical University, Nanjing, China
| | - Yu Jin
- Nanjing Red Cross Blood Center, Nanjing, China
| | - Zhongyi Yan
- Department of Oral and Maxillofacial Surgery, Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang 222001, Jiangsu, China
| | - Chunyan He
- Department of Clinical Laboratory, Kunshan Hospital of Chinese Medicine, Affiliated Hospital of Yangzhou University, Kunshan, Jiangsu, China
| | - Wenhua You
- Wuxi People's Hospital, Wuxi Medical Center Nanjing & Department of Immunology, School of Basic Medical Science & Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China; The Affiliated Huai'an No. 1 People's Hospital, Nanjing Medical University, Nanjing, China
| | - Zilong Zhu
- Wuxi People's Hospital, Wuxi Medical Center Nanjing & Department of Immunology, School of Basic Medical Science & Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China; The Affiliated Huai'an No. 1 People's Hospital, Nanjing Medical University, Nanjing, China
| | - Ren Wang
- Wuxi People's Hospital, Wuxi Medical Center Nanjing & Department of Immunology, School of Basic Medical Science & Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China; The Affiliated Huai'an No. 1 People's Hospital, Nanjing Medical University, Nanjing, China
| | - Yun Chen
- Wuxi People's Hospital, Wuxi Medical Center Nanjing & Department of Immunology, School of Basic Medical Science & Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China; The Affiliated Huai'an No. 1 People's Hospital, Nanjing Medical University, Nanjing, China.
| | - Judong Luo
- Department of Radiotherapy, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China.
| | - Yuan Zhang
- Department of Head and Neck Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University & The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center Nanjing, Nanjing Medical University, Nanjing, China.
| | - Yao Yao
- Department of Head and Neck Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University & The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center Nanjing, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
3
|
Pan X, Xu C, Cheng G, Chen Z, Liu M, Mei Y. Transcription factor E2F3 activates CDC25B to regulate DNA damage and promote mitoxantrone resistance in stomach adenocarcinoma. Mol Biol Rep 2024; 51:90. [PMID: 38194158 DOI: 10.1007/s11033-023-08933-0] [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: 09/05/2023] [Accepted: 10/10/2023] [Indexed: 01/10/2024]
Abstract
BACKGROUND CDC25B, as a member of the cell cycle regulating protein family, is located in the cytoplasm and is involved in the transition of the cell cycle and mitosis. CDC25B is highly expressed in various tumors and is a newly discovered oncogene. This study aimed to investigate the impact of CDC25B on mitoxantrone resistance in stomach adenocarcinoma (STAD) and its possible mechanisms. METHODS This study analyzed the expression of CDC25B and its potential transcription factor E2F3 in STAD, as well as the IC50 values of tumor tissues by bioinformatics analysis. Expression levels of CDC25B and E2F3 in STAD cells were measured by qRT-PCR. MTT was utilized to evaluate cell viability and IC50 values of STAD cells, and comet assay was utilized to analyze the level of DNA damage in STAD cells. Western blot was used to analyze the expression of DNA damage-related proteins. The targeting relationship between E2F3 and CDC25B was validated by dual-luciferase and ChIP assays. RESULTS Bioinformatics analysis and molecular experiments showed that CDC25B and E2F3 were highly expressed in STAD, and CDC25B was enriched in the mismatch repair and nucleotide excision repair pathways. The IC50 values of tumor tissues with high expression of CDC25B were relatively high. Dual-luciferase and ChIP assays confirmed that CDC25B could be transcriptionally activated by E2F3. Cell experiments revealed that CDC25B promoted mitoxantrone resistance in STAD cells by regulating DNA damage. Further research found that low expression of E2F3 inhibited mitoxantrone resistance in STAD cells by DNA damage, but overexpression of CDC25B reversed the impact of E2F3 knockdown on mitoxantrone resistance in STAD cells. CONCLUSION This study confirmed a novel mechanism by which E2F3/CDC25B mediated DNA damage to promote mitoxantrone resistance in STAD cells, providing a new therapeutic target for STAD treatment.
Collapse
Affiliation(s)
- Xiaoming Pan
- Department of Gastrointestinal Surgery, Lishui People's Hospital, No.15 Dazhong Street, Liandu District, Lishui, Zhejiang Province, 323000, China
| | - Chaobo Xu
- Department of Gastrointestinal Surgery, Lishui People's Hospital, No.15 Dazhong Street, Liandu District, Lishui, Zhejiang Province, 323000, China
| | - Guoxiong Cheng
- Department of Gastrointestinal Surgery, Lishui People's Hospital, No.15 Dazhong Street, Liandu District, Lishui, Zhejiang Province, 323000, China
| | - Zhengwei Chen
- Department of Gastrointestinal Surgery, Lishui People's Hospital, No.15 Dazhong Street, Liandu District, Lishui, Zhejiang Province, 323000, China
| | - Ming Liu
- Department of Gastrointestinal Surgery, Lishui People's Hospital, No.15 Dazhong Street, Liandu District, Lishui, Zhejiang Province, 323000, China
| | - Yijun Mei
- Department of Gastrointestinal Surgery, Lishui People's Hospital, No.15 Dazhong Street, Liandu District, Lishui, Zhejiang Province, 323000, China.
| |
Collapse
|
4
|
Huang H, Liang X, Wu W, Yuan T, Chen Z, Wang L, Wu Z, Zhang T, Yang K, Wen K. FOXP3-regulated lncRNA NONHSAT136151 promotes colorectal cancer progression by disrupting QKI interaction with target mRNAs. J Cell Mol Med 2024; 28:e18068. [PMID: 38041531 PMCID: PMC10826441 DOI: 10.1111/jcmm.18068] [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: 05/18/2023] [Revised: 11/16/2023] [Accepted: 11/22/2023] [Indexed: 12/03/2023] Open
Abstract
The role of lncRNAs in the pathogenesis of cancer, including colorectal cancer (CRC), has repeatedly been demonstrated. However, very few lncRNAs have been well annotated functionally. Our study identified a novel lncRNA upregulated in CRC, NONHSAT136151, which was correlated with clinical progression. In functional assays, NONHSAT136151 significantly enhanced CRC cell proliferation, migration and invasion. Mechanistically, NONHSAT136151 interacted with RNA-binding protein (RBP) QKI (Quaking) to interfere with QKI binding to target mRNAs and regulate their expression. As well, FOXP3 may be causally related to the dysregulation of NONHSAT136151 in CRC cells through its transcriptional activity. In conclusion, our findings identified a novel lncRNA regulated by FOXP3 participates in CRC progression through interacting with QKI, indicating a novel lncRNA-RBP interaction mechanism is involved in CRC pathogenesis.
Collapse
Affiliation(s)
- Handong Huang
- Soochow University Medical CollegeSuzhouJiangsuChina
- Department of General SurgeryAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| | - Xiaoxiang Liang
- Department of General SurgeryAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| | - Weizheng Wu
- Department of General SurgeryAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| | - Tao Yuan
- Department of General SurgeryAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| | - Zhengquan Chen
- Department of General SurgeryAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| | - Lin Wang
- Department of General SurgeryAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| | - Zhenyu Wu
- Department of General SurgeryAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| | - Tao Zhang
- Department of General SurgeryAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| | - Kai Yang
- Department of General SurgeryAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| | - Kunming Wen
- Soochow University Medical CollegeSuzhouJiangsuChina
- Department of General SurgeryAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| |
Collapse
|
5
|
Mohan S, Hakami MA, Dailah HG, Khalid A, Najmi A, Zoghebi K, Halawi MA, Alotaibi TM. From inflammation to metastasis: The central role of miR-155 in modulating NF-κB in cancer. Pathol Res Pract 2024; 253:154962. [PMID: 38006837 DOI: 10.1016/j.prp.2023.154962] [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: 10/24/2023] [Revised: 11/16/2023] [Accepted: 11/16/2023] [Indexed: 11/27/2023]
Abstract
Cancer is a multifaceted, complex disease characterized by unchecked cell growth, genetic mutations, and dysregulated signalling pathways. These factors eventually cause evasion of apoptosis, sustained angiogenesis, tissue invasion, and metastasis, which makes it difficult for targeted therapeutic interventions to be effective. MicroRNAs (miRNAs) are essential gene expression regulators linked to several biological processes, including cancer and inflammation. The NF-κB signalling pathway, a critical regulator of inflammatory reactions and oncogenesis, has identified miR-155 as a significant participant in its modulation. An intricate network of transcription factors known as the NF-κB pathway regulates the expression of genes related to inflammation, cell survival, and immunological responses. The NF-κB pathway's dysregulation contributes to many cancer types' development, progression, and therapeutic resistance. In numerous cancer models, the well-studied miRNA miR-155 has been identified as a crucial regulator of NF-κB signalling. The p65 subunit and regulatory molecules like IκB are among the primary targets that miR-155 directly targets to alter NF-κB activity. The molecular processes by which miR-155 affects the NF-κB pathway are discussed in this paper. It also emphasizes the miR-155's direct and indirect interactions with important NF-κB cascade elements to control the expression of NF-κB subunits. We also investigate how miR-155 affects NF-κB downstream effectors in cancer, including inflammatory cytokines and anti-apoptotic proteins.
Collapse
Affiliation(s)
- Syam Mohan
- Substance Abuse and Toxicology Research Centre, Jazan University, Jazan 45142, Saudi Arabia; School of Health Sciences, University of Petroleum and Energy Studies, Dehradun, Uttarakhand, India; Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India.
| | - Mohammed Ageeli Hakami
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Al, Quwayiyah, Shaqra University, Riyadh, Saudi Arabia.
| | - Hamad Ghaleb Dailah
- Research and Scientific Studies Unit, College of Nursing, Jazan University, Jazan 45142, Saudi Arabia
| | - Asaad Khalid
- Substance Abuse and Toxicology Research Centre, Jazan University, Jazan 45142, Saudi Arabia
| | - Asim Najmi
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Khalid Zoghebi
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Maryam A Halawi
- Department of Clinical Pharmacy, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | | |
Collapse
|
6
|
Hsu FT, Liu WL, Lee SR, Jeng LB, Chen JH. Unveiling nature's potential weapon: Magnolol's role in combating bladder cancer by upregulating the miR-124 and inactivating PKC-δ/ERK axis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 119:154947. [PMID: 37549536 DOI: 10.1016/j.phymed.2023.154947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/07/2023] [Accepted: 06/28/2023] [Indexed: 08/09/2023]
Abstract
BACKGROUND Bladder cancer (BC) is a challenging disease to manage. Researchers have been investigating the potential of magnolol, a compound derived from Magnolia officinalis, as an anti-cancer agent. However, the exact regulatory mechanism of magnolol and its impact on the NF-κB signaling pathway in BC remain unclear. MATERIALS To comprehensively evaluate its therapeutic potential, the researchers conducted a series of experiments using BC cell lines (TSGH8301, T24, and MB49) and in vivo animal models. RESULTS The results of the study demonstrated that magnolol exhibits cytotoxic effects on BC cells by activating both the extrinsic and intrinsic apoptosis signaling pathways. Additionally, the expression of anti-apoptotic genes was downregulated by magnolol treatment. The researchers also uncovered the regulatory role of PKCδ/ERK and miR-124-3p in the NF-κB pathway, which may be influenced by magnolol. Treatment with magnolol led to the inactivation of PKCδ/ERK and an increase in miR-124-3p expression, effectively inhibiting NF-κB-mediated progression of BC. Importantly, the administration of magnolol did not result in significant toxicity in normal tissues, highlighting its potential as a safe adjunctive therapy with minimal adverse effects. CONCLUSION These findings position magnolol as a promising therapeutic agent for the treatment of BC. By activating apoptosis signaling pathways and inhibiting NF-κB pathway through the upregulation of miR-124-3p and downregulation of PKCδ/ERK activation, magnolol holds promise for suppressing tumor progression and improving patient outcomes in BC. Further research and clinical trials are warranted to explore the full potential of magnolol in the future.
Collapse
Affiliation(s)
- Fei-Ting Hsu
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan, R.O.C
| | - Wei-Lin Liu
- Department of Radiation Oncology, Show Chwan Memorial Hospital, Changhua, Taiwan, R.O.C
| | - Sin-Rong Lee
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan, R.O.C; Institute of Biochemical Sciences, College of Life Science, National Taiwan University, Taipei, Taiwan, R.O.C
| | - Long-Bin Jeng
- Organ Transplantation Center, China Medical University Hospital, Taichung, Taiwan, R.O.C; Cell Therapy Center, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Jiann-Hwa Chen
- Department of Emergency Medicine, Cathay General Hospital, Taipei, Taiwan, R.O.C; School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan, R.O.C.
| |
Collapse
|
7
|
Gao Q, An K, Lv Z, Wang Y, Ding C, Huang W. E2F3 accelerates the stemness of colon cancer cells by activating the STAT3 pathway. Front Oncol 2023; 13:1203712. [PMID: 37456248 PMCID: PMC10346838 DOI: 10.3389/fonc.2023.1203712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 06/07/2023] [Indexed: 07/18/2023] Open
Abstract
Introduction Colon cancer is one of the most prevalent malignancies and causes of cancer-related deaths worldwide. Thus, further research is required to explicate the latent molecular mechanisms and look for novel biomarkers. E2F3 has been confirmed to be an oncogene in a variety of cancers. However, the particular regulation of E2F3 in colon cancer needs further investigation. Methods The self-renewal ability was detected through a sphere formation assay. The tumorigenic ability was measured through nude mice in vivo assay. The protein expression of genes was examined through a Western blot. The expression of E2F3 in tumor tissues was detected through an IHC assay. The resistance to cisplatin was assessed through the CCK-8 assay. The cell migration and invasion abilities were measured after upregulating or suppressing E2F3 through the Transwell assay. Results Results uncovered that E2F3 was upregulated in spheroid cells. In addition, E2F3 facilitates stemness in colon cancer. Moreover, E2F3 facilitated colon cancer cell migration and invasion. Finally, it was revealed that E2F3 affected the STAT3 pathway to modulate stemness in colon cancer. E2F3 served as a promoter regulator in colon cancer, aggravating tumorigenesis and stemness in colon cancer progression through the STAT3 pathway. Conclusion E2F3 may be a useful biomarker for anticancer treatment in colon cancer.
Collapse
|
8
|
Sun M, Shen Y, Jia G, Deng Z, Shi F, Jing Y, Xia S. Activation of the HNRNPA2B1/ miR-93-5p/FRMD6 axis facilitates prostate cancer progression in an m6A-dependent manner. J Cancer 2023; 14:1242-1256. [PMID: 37215455 PMCID: PMC10197942 DOI: 10.7150/jca.83863] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 04/16/2023] [Indexed: 05/24/2023] Open
Abstract
It is becoming increasingly clear that N6-methyladenosine (m6A) plays a key role in post-transcriptional modification of eukaryotic RNAs in cancer. The regulatory mechanism of m6A modifications in prostate cancer is still not completely elucidated. Heterogeneous nuclear ribonucleoprotein A2/B1 (HNRNPA2B1), an m6A reader, has been revealed to function as an oncogenic RNA-binding protein. However, its contribution to prostate cancer progression remains poorly understood. Here, we found that HNRNPA2B1 was highly overexpressed and correlated with a poor prognosis in prostate cancer. In vitro and in vivo functional experiments demonstrated that HNRNPA2B1 knockout impaired proliferation and metastasis of prostate cancer. Mechanistic studies indicated that HNRNPA2B1 interacted with primary miRNA-93 and promoted its processing by recruiting DiGeorge syndrome critical region gene 8 (DGCR8), a key subunit of the Microprocessor complex, in an METTL3-dependent mechanism, while HNRNPA2B1 knockout significantly restored miR-93-5p levels. HNRNPA2B1/miR-93-5p downregulated FERM domain-containing protein 6 (FRMD6), a cancer suppressor, and enhanced proliferation and metastasis in prostate cancer. In conclusion, our findings identified a novel oncogenic axis, HNRNPA2B1/miR-93-5p/FRMD6, that stimulates prostate cancer progression via an m6A-dependent manner.
Collapse
Affiliation(s)
- Menghao Sun
- Clinical Medical Center of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Urology, Shanghai Jiao Tong University, Shanghai, China
| | - Yuanhao Shen
- Clinical Medical Center of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Urology, Shanghai Jiao Tong University, Shanghai, China
| | - Gaozhen Jia
- Clinical Medical Center of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Urology, Shanghai Jiao Tong University, Shanghai, China
| | - Zheng Deng
- Cancer Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fei Shi
- Clinical Medical Center of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Urology, Shanghai Jiao Tong University, Shanghai, China
| | - Yifeng Jing
- Clinical Medical Center of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Urology, Shanghai Jiao Tong University, Shanghai, China
| | - Shujie Xia
- Clinical Medical Center of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Urology, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
9
|
Gao Y, Cao H, Huang D, Zheng L, Nie Z, Zhang S. RNA-Binding Proteins in Bladder Cancer. Cancers (Basel) 2023; 15:cancers15041150. [PMID: 36831493 PMCID: PMC9953953 DOI: 10.3390/cancers15041150] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 02/09/2023] [Accepted: 02/09/2023] [Indexed: 02/15/2023] Open
Abstract
RNA-binding proteins (RBPs) are key regulators of transcription and translation, with highly dynamic spatio-temporal regulation. They are usually involved in the regulation of RNA splicing, polyadenylation, and mRNA stability and mediate processes such as mRNA localization and translation, thereby affecting the RNA life cycle and causing the production of abnormal protein phenotypes that lead to tumorigenesis and development. Accumulating evidence supports that RBPs play critical roles in vital life processes, such as bladder cancer initiation, progression, metastasis, and drug resistance. Uncovering the regulatory mechanisms of RBPs in bladder cancer is aimed at addressing the occurrence and progression of bladder cancer and finding new therapies for cancer treatment. This article reviews the effects and mechanisms of several RBPs on bladder cancer and summarizes the different types of RBPs involved in the progression of bladder cancer and the potential molecular mechanisms by which they are regulated, with a view to providing information for basic and clinical researchers.
Collapse
|
10
|
Zhang H, Li J, Tian F, Su X, Wang X, Tang D, Zhang L, Zhang T, Ni Y. QKI-6 Suppresses Cell Proliferation, Migration, and EMT in Non-Small Cell Lung Cancer. Front Oncol 2022; 12:897553. [PMID: 35600368 PMCID: PMC9117621 DOI: 10.3389/fonc.2022.897553] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 04/07/2022] [Indexed: 11/13/2022] Open
Abstract
The RNA-binding protein quaking homolog 6 (QKI-6) is a tumor-suppressor gene in several cancers. However, its role in non-small cell lung cancer (NSCLC) is unclear. In this study, we aimed to determine the association between QKI-6 expression and survival and clinicopathological features in patients with NSCLC and identify the related mechanisms. Western blot and immunohistochemistry (IHC) were used to detect QKI-6 expression in NSCLC. The effect of QKI-6 on NSCLC cells was determined by overexpression and knockdown assays, and label-free quantitative proteomics and Western blot were used to identify the underlying mechanisms. Low QKI-6 expression level was positively correlated with poor overall survival in patients with NSCLC. Furthermore, QKI-6 overexpression inhibited NSCLC cell proliferation and migration and induced a block in the G0/G1 phase, and QKI-6 downregulation increased proliferation and migration. QKI-6 inhibited EMT processes via EGFR/SRC/STAT3 signaling by upregulating AGR2. In conclusion, QKI-6 could be used to develop novel strategies for the treatment of NSCLC.
Collapse
Affiliation(s)
- Haihua Zhang
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi’an, China
| | - Junqiang Li
- Department of Oncology, Tangdu Hospital, Fourth Military Medical University, Xi’an, China
| | - Feng Tian
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi’an, China
| | - Xuan Su
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi’an, China
| | - Xinxin Wang
- Department of Pulmonary and Critical Care Medicine, Tangdu Hospital, Fourth Military Medical University, Xi’an, China
| | - Di Tang
- Seventh Battalion, Second Cadet Regiment, Fourth Military Medical University, Xi’an, China
| | - Lei Zhang
- Department of Oncology, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Tao Zhang
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi’an, China
| | - Yunfeng Ni
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi’an, China
| |
Collapse
|
11
|
Long Non-coding RNA FOXD2-AS1 Promotes Proliferation, Migration, and Invasion in Cholangiocarcinoma Through Regulating miR-760/E2F3 Axis. Dig Dis Sci 2022; 67:546-558. [PMID: 33570683 DOI: 10.1007/s10620-021-06876-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 01/23/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND Long non-coding RNA (lncRNA) has been testified to influence the initiation and evolution of sundry carcinomas. Recently, lncRNA FOXD2 adjacent opposite strand RNA 1 (FOXD2-AS1) has been found to display vital regulating functions in various cancers. METHODS qRT-PCR was used to verify the dysregulation of FOXD2-AS1 expression in CCA cells and tissues, and the correlation of FOXD2-AS1 expression with clinicopathological characteristics was investigated. The viability, migration, and invasion of CCA cells were verified through CCK-8 assay, colony formation experiment, wound healing assay, and transwell assay. The regulatory networks of FOXD2-AS1 were analyzed by Bioinformatic prediction and dual-luciferase reporter assay. RESULTS We discovered that FOXD2-AS1 was significantly upregulated in CCA and its up-regulation was closely correlated with terminal TNM stage, lymph node metastasis and poor survival in the current research. In addition, it was revealed that FOXD2-AS1 was an independent prognostic factor. Functional tests uncovered that the cell viability, migration, and invasion could be restrained through downregulating the expression of FOXD2-AS1, while FOXD2-AS1 overexpression could facilitate the cell viability, migration, and invasion. Mechanistically, FOXD2-AS1 was founded to interact directly with miR-760 and the oncogene E2F3 was the downstream target of miR-760 through bioinformatic prediction and dual-luciferase reporter assays. Finally, we testified that FOXD2-AS1 could competitively sponge miR-760 and further upregulated the E2F3 expression to play a vital part in cholangiocarcinoma. CONCLUSIONS This research revealed that lncRNA FOXD2-AS1 could enhance CCA malignant progression through regulating the miR-760/E2F3 axis and was expected to be a prognostic biomarker and therapeutic target for cholangiocarcinoma.
Collapse
|
12
|
Nie C, Han J, Bi W, Qiu Z, Chen L, Yu J, Pang R, Liu B, Sheng R, Zhang J. Circular RNA circ_0000644 promotes papillary thyroid cancer progression via sponging miR-1205 and regulating E2F3 expression. Cell Cycle 2021; 21:126-139. [PMID: 34919034 DOI: 10.1080/15384101.2021.2012334] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
The dysregulation of circular RNAs (circRNAs) facilitates the tumorigenesis of papillary thyroid carcinoma (PTC). This study was targeted at determining the functions and mechanism of circ_0000644 in regulating PTC development. Circ_0000644, microRNA-1205 (miR-1205) and E2F transcription factor 3 (E2F3) expressions were detected by quantitative real-time polymerase chain reaction (qRT-PCR). Actinomycin D (ActD) and Ribonuclease R (RNase R) assays were used to verify the circular characteristic of circ_0000644. After circ_0000644 was knocked down, PTC cell growth, migration, invasion and apoptosis were assessed by cell counting kit-8 (CCK-8) assay, Transwell assay and flow cytometry analysis, respectively. The regulating relationships among circ_0000644, E2F3 and miR-1205 were confirmed through RNA immunoprecipitation (RIP) assay and dual-luciferase reporter assay. Besides, the regulatory effects of circ_0000644 on the protein level of E2F3 was analyzed via Western blot. In PTC, circ_0000644 was highly expressed, and it was located mainly in the cytoplasm, and it had stable structure. The knockdown of circ_0000644 repressed PTC cell growth, migration, and invasion, and facilitated apoptosis. Circ_0000644 could directly interact with miR-1205 to repress the expression of miR-1205, and it served as a miR-1205 sponge to modulate E2F3 expression in PTC cells. Circ_0000644 up-regulates E2F3 expression via sponging miR-1205 to promote PTC progression.
Collapse
Affiliation(s)
- Chunlei Nie
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Jihua Han
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Wen Bi
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Zhilin Qiu
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Lili Chen
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Jiawei Yu
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Rui Pang
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Bo Liu
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Ruinan Sheng
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Jiewu Zhang
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| |
Collapse
|
13
|
Luo Y, Li Y, Ge P, Zhang K, Liu H, Jiang N. QKI-Regulated Alternative Splicing Events in Cervical Cancer: Pivotal Mechanism and Potential Therapeutic Strategy. DNA Cell Biol 2021; 40:1261-1277. [PMID: 34551268 DOI: 10.1089/dna.2021.0069] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
QKI is a vital regulator in RNA splicing and maturation, but its role in cervical cancer (CC) is little known. In this study, we found that QKI is decreased in human CC, and overexpression of QKI inhibits HeLa cell proliferation and promotes the apoptosis of cancer cells. We identified hundreds of endogenous QKI-regulated alternative splicing events (ASEs) and differentially expressed genes (DEGs) in QKI-overexpressed HeLa cells by RNA-seq and selectively validated their expression by quantitative reverse-transcription polymerase chain reaction. The gene ontology and Kyoto encyclopedia of genes and genomes (KEGG) enrichment analysis showed that QKI-regulated ASEs and DEGs were closely related to cancer, apoptosis, and transcriptional regulatory functions. In short, QKI may affect the occurrence and development of CC by regulating gene expression through AS.
Collapse
Affiliation(s)
- Yalan Luo
- Laboratory of Integrative Medicine, The First Affiliated Hospital, Dalian Medical University, Dalian, China.,Department of General Surgery, The First Affiliated Hospital, Dalian Medical University, Dalian, China.,Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Yuyuan Li
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Peng Ge
- Laboratory of Integrative Medicine, The First Affiliated Hospital, Dalian Medical University, Dalian, China.,Department of General Surgery, The First Affiliated Hospital, Dalian Medical University, Dalian, China.,Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Kaina Zhang
- Department of Gynecology and Obstetrics, Central Hospital of Zhuanghe City, Zhuanghe, China
| | - Huanhuan Liu
- Laboratory of Integrative Medicine, The First Affiliated Hospital, Dalian Medical University, Dalian, China.,Department of General Surgery, The First Affiliated Hospital, Dalian Medical University, Dalian, China.,Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Nan Jiang
- Department of Gynecology and Obstetrics, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| |
Collapse
|
14
|
The Indication of Poor Prognosis by High Expression of ENO1 in Squamous Cell Carcinoma of the Lung. JOURNAL OF ONCOLOGY 2021; 2021:9910962. [PMID: 34504528 PMCID: PMC8423576 DOI: 10.1155/2021/9910962] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 06/27/2021] [Accepted: 08/19/2021] [Indexed: 12/21/2022]
Abstract
The purpose of this study is to investigate the significance of alpha-enolase (ENO1) expression in squamous cell carcinoma of the lung (LUSC), its prognostic value, and prospective molecular mechanism. Using multiplatforms data, including in-house immunohistochemistry, in-house real-time fluorescence quantitative polymerase chain reaction (RT-qPCR), in-house microarray, and public high-throughput data, the expression significance and prognostic role of ENO1 in LUSC tissues were analyzed comprehensively. With the combination of all eligible cases, compared with 941 non-LUSC lung tissues, ENO1 was significantly overexpressed in 1163 cases of LUSC (standardized mean difference (SMD) = 1.23, 95% confidence interval (CI) = 0.76–1.70, P < 0.001). ENO1 also displayed a great ability to differentiate LUSC tissues from non-LUSC lung tissues (AUC = 0.8705) with the comprehensive sensitivity being 0.88 [0.83–0.92], and comprehensive specificity being 0.89 [0.84–0.94]). Moreover, in 1860 cases of LUSC with survival information, patients with higher expression of ENO1 had poorer prognosis (hazard ratio (HR) = 1.20, 95% CI = 1.01–1.43, P = 0.043). ENO1 and its related genes mainly participated in the pathways of cell division and proliferation. In conclusion, the upregulation of ENO1 could affect the carcinogenesis and unfavorable outcome of LUSC.
Collapse
|
15
|
Liao Y, Zou X, Wang K, Wang Y, Wang M, Guo T, Zhong B, Jiang N. Comprehensive analysis of Transcription Factors identified novel prognostic biomarker in human bladder cancer. J Cancer 2021; 12:5605-5621. [PMID: 34405021 PMCID: PMC8364643 DOI: 10.7150/jca.58484] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 07/20/2021] [Indexed: 12/13/2022] Open
Abstract
Background: Transcriptional factors (TFs) are responsible for regulating the transcription of pro-oncogenes and tumor suppressor genes in the process of tumor development. However, the role of these transcription factors in Bladder cancer (BCa) remains unclear. And the main purpose of this research is to explore the possibility of these TFs serving as biomarkers for BCa. Methods: We analyzed the differential expression of TFs in BCa from The Cancer Genome Atlas (TCGA) online database, identified 408 up-regulated TFs and 751down-regulated TFs. We obtained some hub genes via WGCNA model and detected the RNAs level in BCa cells and tissues. Then, the relationship between the expression and clinicopathological parameters was further investigated. Kaplan-Meier curves and the log-rank test were carried out to analyze the relationship between NFATC1, AKNA and five-TFs combination and overall survival (OS). And RT-PCR assay was conducted to further consolidate and verify these results. Results: There were significant differences in the expression of five TFs (CBX7, AKNA, HDAC4, EBF2 and NFATC1) between bladder cancer and normal bladder tissue. In BCa tissue and cell lines, the five TFs were frequently down-regulated, and closely related to poor prognosis. Moreover, the RT-PCR results of five TFs in bladder cancer and normal bladder tissue were consistent with the database results, and reduced TFs could significantly induce or restrain the transcription of many critical factors. The expression level of AKNA and NFATC1 could serve as independent biomarker to predict the overall survival (P<0.05). And the above five TFs combined detection of bladder cancer has higher sensitivity and specificity. Furthermore, differential neutrophils expression between high-risk and low-risk were found, which consolidated the role and function of the five TFs combination model in the progression of BCa. Conclusions: Our analysis effectively provides a newly TFs-associated prognostic model for bladder cancer. The combination of five identified-TFs is an independent prognostic biomarker, which could serve as a more effective therapeutic target for BCa patients.
Collapse
Affiliation(s)
- Yihao Liao
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Xuanxuan Zou
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Keke Wang
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Youzhi Wang
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Miaomiao Wang
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Tao Guo
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Boqiang Zhong
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Ning Jiang
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| |
Collapse
|
16
|
Chen X, Yin J, Cao D, Xiao D, Zhou Z, Liu Y, Shou W. The Emerging Roles of the RNA Binding Protein QKI in Cardiovascular Development and Function. Front Cell Dev Biol 2021; 9:668659. [PMID: 34222237 PMCID: PMC8242579 DOI: 10.3389/fcell.2021.668659] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 05/10/2021] [Indexed: 12/30/2022] Open
Abstract
RNA binding proteins (RBPs) have a broad biological and physiological function and are critical in regulating pre-mRNA posttranscriptional processing, intracellular migration, and mRNA stability. QKI, also known as Quaking, is a member of the signal transduction and activation of RNA (STAR) family, which also belongs to the heterogeneous nuclear ribonucleoprotein K- (hnRNP K-) homology domain protein family. There are three major alternatively spliced isoforms, QKI-5, QKI-6, and QKI-7, differing in carboxy-terminal domains. They share a common RNA binding property, but each isoform can regulate pre-mRNA splicing, transportation or stability differently in a unique cell type-specific manner. Previously, QKI has been known for its important role in contributing to neurological disorders. A series of recent work has further demonstrated that QKI has important roles in much broader biological systems, such as cardiovascular development, monocyte to macrophage differentiation, bone metabolism, and cancer progression. In this mini-review, we will focus on discussing the emerging roles of QKI in regulating cardiac and vascular development and function and its potential link to cardiovascular pathophysiology.
Collapse
Affiliation(s)
- Xinyun Chen
- Department of Pediatrics, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
- Guangdong Key Laboratory for Genome Stability and Human Disease Prevention, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shenzhen University, Shenzhen, China
| | - Jianwen Yin
- Department of Foot, Ankle and Hand Surgery, Shenzhen Second People’s Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Dayan Cao
- Department of Pediatrics, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Deyong Xiao
- Department of Pediatrics, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Zhongjun Zhou
- Faculty of Medicine, School of Biomedical Sciences, The University of Hong Kong, Hong Kong
| | - Ying Liu
- Department of Pediatrics, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Weinian Shou
- Department of Pediatrics, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
17
|
Pengcheng Z, Peng G, Haowen F, Xida L, Yuhua L, Yao W, Mingyan Z, Xiangjun F, Zhiwei W, Yewei Z, Lei W. MiR-573 suppresses cell proliferation, migration and invasion via regulation of E2F3 in pancreatic cancer. J Cancer 2021; 12:3033-3044. [PMID: 33854603 PMCID: PMC8040892 DOI: 10.7150/jca.51147] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 02/15/2021] [Indexed: 01/06/2023] Open
Abstract
Background: Pancreatic cancer is among the most lethal malignancies worldwide. In this study, we aimed to determine whether miR-573 could suppress pancreatic cancer cell proliferation, migration, and invasion by targeting E2F3. Materials and Methods: MiR-573 expression in pancreatic cancer tissues and cell lines was measured using real-time PCR. Target genes of miR-573 were screened using bioinformatics tools and confirmed using dual-luciferase reporter assay and real-time PCR. Pancreatic cancer cells were transfected using an miR-573 mimic or siRNA E2F3. Furthermore, cell proliferation, migration, and invasion were assessed using CCK-8, Edu staining, colony-forming assay, wound healing assay, and transwell assay in vitro. The in vivo effects of miR-573 were verified using tumor xenografts. Differential expression and prognostic analyses of miR-573 and E2F3 were visualized using the Kaplan‑Meier plotter and GEPIA. Results: We found that the expression of miR-573 was significantly reduced in pancreatic cancer tissues and cell lines. Overexpression of miR-573 obviously suppressed the proliferation, migration, and invasion of pancreatic cancer cells. The Dual-luciferase assay showed that miR-573 could specifically target E2F3. Furthermore, E2F3 was up-regulated in pancreatic cancer tissues and cell lines and E2F3 down-regulation inhibited the proliferation, migration, and invasion of pancreatic cancer cells. The ectopic expression of miR-573 inhibited xenograft tumor growth in vivo. Results from the Kaplan-Meier analysis and GEPIA showed that patients with a high level of miR-573 had a significantly reduced risk of death while those with a high level of E2F3 displayed significant correlation with the tumor stage and suffered worse prognosis. Conclusions: MiR-573 could suppress the proliferation, migration, and invasion of pancreatic cancer cells by targeting E2F3, thereby establishing miR-573 as a novel regulator of E2F3 and indicating its critical role in tumorigenesis, especially in pancreatic cancer.
Collapse
Affiliation(s)
- Zhou Pengcheng
- Medical school of Southeast University, Nanjing, Jiangsu, China.,Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Gao Peng
- Nantong Traditional Chinese Medicine Hospital, Nantong, Jiangsu, China
| | - Fan Haowen
- Nantong University, Nantong, Jiangsu, China
| | - Lin Xida
- Nantong University, Nantong, Jiangsu, China
| | - Lu Yuhua
- Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Wang Yao
- Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Zhu Mingyan
- Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Fan Xiangjun
- Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Wang Zhiwei
- Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Zhang Yewei
- Zhongda Hospital affiliated to Southeast University, Nanjing, Jiangsu, China
| | - Wang Lei
- Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
18
|
Chen X, Liu Y, Xu C, Ba L, Liu Z, Li X, Huang J, Simpson E, Gao H, Cao D, Sheng W, Qi H, Ji H, Sanderson M, Cai CL, Li X, Yang L, Na J, Yamamura K, Liu Y, Huang G, Shou W, Sun N. QKI is a critical pre-mRNA alternative splicing regulator of cardiac myofibrillogenesis and contractile function. Nat Commun 2021; 12:89. [PMID: 33397958 PMCID: PMC7782589 DOI: 10.1038/s41467-020-20327-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Accepted: 11/27/2020] [Indexed: 01/29/2023] Open
Abstract
The RNA-binding protein QKI belongs to the hnRNP K-homology domain protein family, a well-known regulator of pre-mRNA alternative splicing and is associated with several neurodevelopmental disorders. Qki is found highly expressed in developing and adult hearts. By employing the human embryonic stem cell (hESC) to cardiomyocyte differentiation system and generating QKI-deficient hESCs (hESCs-QKIdel) using CRISPR/Cas9 gene editing technology, we analyze the physiological role of QKI in cardiomyocyte differentiation, maturation, and contractile function. hESCs-QKIdel largely maintain normal pluripotency and normal differentiation potential for the generation of early cardiogenic progenitors, but they fail to transition into functional cardiomyocytes. In this work, by using a series of transcriptomic, cell and biochemical analyses, and the Qki-deficient mouse model, we demonstrate that QKI is indispensable to cardiac sarcomerogenesis and cardiac function through its regulation of alternative splicing in genes involved in Z-disc formation and contractile physiology, suggesting that QKI is associated with the pathogenesis of certain forms of cardiomyopathies.
Collapse
Affiliation(s)
- Xinyun Chen
- grid.8547.e0000 0001 0125 2443Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China ,grid.411333.70000 0004 0407 2968Shanghai Key Laboratory of Birth Defects, Children’s Hospital of Fudan University, Shanghai, China ,grid.257413.60000 0001 2287 3919Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN USA
| | - Ying Liu
- grid.257413.60000 0001 2287 3919Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN USA
| | - Chen Xu
- grid.8547.e0000 0001 0125 2443Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China ,grid.257413.60000 0001 2287 3919Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN USA
| | - Lina Ba
- grid.257413.60000 0001 2287 3919Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN USA
| | - Zhuo Liu
- grid.257413.60000 0001 2287 3919Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN USA
| | - Xiuya Li
- grid.8547.e0000 0001 0125 2443Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jie Huang
- grid.8547.e0000 0001 0125 2443Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ed Simpson
- grid.257413.60000 0001 2287 3919Department of Bioinformatics, Indiana University School of Medicine, Indianapolis, IN USA
| | - Hongyu Gao
- grid.257413.60000 0001 2287 3919Department of Bioinformatics, Indiana University School of Medicine, Indianapolis, IN USA
| | - Dayan Cao
- Institute of Materia Medica and Center of Translational Medicine, College of Pharmacy, Army Medical University, Chongqing, China
| | - Wei Sheng
- grid.411333.70000 0004 0407 2968Shanghai Key Laboratory of Birth Defects, Children’s Hospital of Fudan University, Shanghai, China ,grid.257413.60000 0001 2287 3919Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN USA
| | - Hanping Qi
- grid.257413.60000 0001 2287 3919Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN USA
| | - Hongrui Ji
- grid.257413.60000 0001 2287 3919Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN USA
| | - Maria Sanderson
- grid.257413.60000 0001 2287 3919Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN USA
| | - Chen-Leng Cai
- grid.257413.60000 0001 2287 3919Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN USA
| | - Xiaohui Li
- Institute of Materia Medica and Center of Translational Medicine, College of Pharmacy, Army Medical University, Chongqing, China
| | - Lei Yang
- grid.257413.60000 0001 2287 3919Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN USA
| | - Jie Na
- grid.12527.330000 0001 0662 3178Center for Stem Cell Biology and Regenerative Medicine, School of Medicine, Tsinghua University, Beijing, China
| | - Kenichi Yamamura
- Institute of Resource Development and Analysis, Kumanoto University, Kumanoto, Japan
| | - Yunlong Liu
- grid.257413.60000 0001 2287 3919Department of Bioinformatics, Indiana University School of Medicine, Indianapolis, IN USA
| | - Guoying Huang
- grid.411333.70000 0004 0407 2968Shanghai Key Laboratory of Birth Defects, Children’s Hospital of Fudan University, Shanghai, China
| | - Weinian Shou
- grid.257413.60000 0001 2287 3919Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN USA
| | - Ning Sun
- grid.8547.e0000 0001 0125 2443Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China ,grid.411333.70000 0004 0407 2968Shanghai Key Laboratory of Birth Defects, Children’s Hospital of Fudan University, Shanghai, China
| |
Collapse
|
19
|
Hu C, Fang K, Zhang X, Guo Z, Li L. Dyregulation of the lncRNA TPT1-AS1 positively regulates QKI expression and predicts a poor prognosis for patients with breast cancer. Pathol Res Pract 2020; 216:153216. [PMID: 32961484 DOI: 10.1016/j.prp.2020.153216] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/31/2020] [Accepted: 09/08/2020] [Indexed: 12/17/2022]
Abstract
The long noncoding RNA (lncRNA) TPT1-AS1 has been reported to be involved in the development of multiple cancers. However, its clinical value, biological function, and underlying molecular mechanism in breast cancer (BC) remain unclear. In the present study, TPT1-AS1 expression was decreased in BC tissues, based on RNA-seq data download from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases and the qRT-PCR results confirmed the above findings. Otherwise, low TPT1-AS1 expression was significantly associated with some clinical features of malignancy, such as high TNM stage, lymph node metastasis, a Her-2-negative status, and shorter overall survival. More importantly, univariate and multivariate Cox regression analyses indicated that TPT1-AS1 is an independent prognostic factor for patients with BC. Overexpression and knockdown of TPT1-AS1 in BC cell lines altered their proliferation, metastasis and invasion, as measured using the cell counting kit-8 (CCK-8) assay, wound-healing assay and transwell assay, respectively. In addition, a dual luciferase activity reporter assay validated that TPT1-AS1 and QKI shared a binding site in miR-330-3p. Based on these findings, TPT1-AS1 potentially represents a prognostic biomarker for patients with BC and participates in the development of BC through the TPT1-AS1/miR-330-3p/QKI axis.
Collapse
Affiliation(s)
- Caixia Hu
- Oncology Institute, The Affiliated Hospital of Jiangnan University, Wuxi, 214062, China
| | - Kai Fang
- Oncology Institute, The Affiliated Hospital of Jiangnan University, Wuxi, 214062, China
| | - Xiufen Zhang
- Oncology Institute, The Affiliated Hospital of Jiangnan University, Wuxi, 214062, China
| | - Zijian Guo
- Department of Oncological Surgery, The Affiliated Hospital of Jiangnan University, Wuxi, 214062, China.
| | - Lihua Li
- Oncology Institute, The Affiliated Hospital of Jiangnan University, Wuxi, 214062, China.
| |
Collapse
|
20
|
Xue C, Cheng Y, Wu J, Ke K, Miao C, Chen E, Zhang L. Circular RNA CircPRMT5 Accelerates Proliferation and Invasion of Papillary Thyroid Cancer Through Regulation of miR-30c/E2F3 Axis. Cancer Manag Res 2020; 12:3285-3291. [PMID: 32494192 PMCID: PMC7231777 DOI: 10.2147/cmar.s249237] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 04/21/2020] [Indexed: 12/11/2022] Open
Abstract
Background The role of circular RNA (circRNA) in papillary thyroid cancer (PTC) is largely unknown. This study aims to determine the function and mechanism of circPRMT5 in the regulation of PTC development. Methods PTC tissues and cell lines were used to determine circPRMT5 expression via quantitative real-time polymerase chain reaction. Small interfering RNA (siRNA) was utilized to knock down circPRMT5. Proliferation was analyzed through CCK8 and colony formation assays. Transwell assay was performed to determine cell migration and invasion. Luciferase assay and RIP assay were carried out to analyze the interaction between circPRMT5 and miR-30c. Results CircPRMT5 expression was upregulated in PTC tissues and cell lines. And circPRMT5 level was positively linked with advanced stage and lymph node metastasis. CircPRMT5 knockdown inhibited proliferation, migration and invasion while inducing apoptosis. CircPRMT5 worked as a competing endogenous RNA for miR-30c. By inhibiting miR-30c, circPRMT5 promoted the expression of E2F3. Conclusion Our findings demonstrate that circPRMT5 acts as an oncogenic circRNA to promote PTC progression via regulating miR-30c/E2F3 axis.
Collapse
Affiliation(s)
- Cheng Xue
- Department of Endocrinology, Wenling First People's Hospital, Wenling 317500, People's Republic of China
| | - Yi Cheng
- Department of Endocrinology, Wenling First People's Hospital, Wenling 317500, People's Republic of China
| | - Jinyou Wu
- Department of Endocrinology, Wenling First People's Hospital, Wenling 317500, People's Republic of China
| | - Kongliang Ke
- Department of Anorectal Surgery, Ningbo Hangzhou Bay Hospital, Ningbo 315000, People's Republic of China
| | - Chundi Miao
- Department of Anorectal Surgery, Ningbo Hangzhou Bay Hospital, Ningbo 315000, People's Republic of China
| | - Enfu Chen
- Department of Endocrinology, Wenling First People's Hospital, Wenling 317500, People's Republic of China
| | - Luqing Zhang
- Department of Anorectal Surgery, Ningbo Hangzhou Bay Hospital, Ningbo 315000, People's Republic of China
| |
Collapse
|
21
|
Fei Y, Shan W, Chen X. MiR-503-5p functions as an oncogene in oral squamous cell carcinoma by targeting Smad7. Histol Histopathol 2020; 35:893-901. [PMID: 32319077 DOI: 10.14670/hh-18-220] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROND Oral squamous cell carcinoma (OSCC) is a common oral malignancy. Previous studies indicated that the level of miR-503-5p was upregulated in OSCC tissues. However, the mechanism by which miR-503-5p regulates the proliferation and invasion of OSCC cells remains unclear. Therefore, this study aimed to investigate the role of miR-503-5p during the progression of OSCC. METHODS The level of miR-503-5p in Tca8113 cells was detected using RT-qPCR assay. In addition, CCK-8, transwell assays and flow cytometry assays were conducted to detect cell viability, migration, invasion and apoptosis, respectively. Meanwhile, the dual luciferase reporter assay was applied to explore the interaction between miR-503-5p and Smad7 in Tca8113 cells. RESULTS Overexpression of miR-503-5p significantly promoted the proliferation, migration and invasion of Tca8113 cells, while downregulation of miR-503-5p markedly inhibited proliferation, migration and invasion of cells. In addition, knockdown of miR-503-5p obviously induced the apoptosis of Tca8113 cells via increasing the levels of Bax and cleaved caspase 3, and decreased the expression of Bcl-2. Moreover, SMAD family member 7 (Smad7) was identified as a direct binding target of miR-503-5p in Tca8113 cells. Overexpression of miR-503-5p significantly downregulated the levels of Smad7 and E-cadherin, but upregulated the levels of N-cadherin and MMP-9 in Tca8113 cells. CONCLUSION These results indicated that miR-503-5p might act as an oncogene in OSCC cells by targeting Smad7. Therefore, miR-503-5p might act as a novel and potential therapeutic target for the treatment of OSCC.
Collapse
Affiliation(s)
- Yifan Fei
- Department of Stomatology, Shanghai Changzheng Hospital, the Second Military Medical University, Shanghai, PR China
| | - Weilan Shan
- Department of Stomatology, Shanghai Changzheng Hospital, the Second Military Medical University, Shanghai, PR China
| | - Xiaoqing Chen
- Department of Stomatology, Shanghai Changzheng Hospital, the Second Military Medical University, Shanghai, PR China.
| |
Collapse
|
22
|
Zhou Z, Cui D, Sun MH, Huang JL, Deng Z, Han BM, Sun XW, Xia SJ, Sun F, Shi F. CAFs-derived MFAP5 promotes bladder cancer malignant behavior through NOTCH2/HEY1 signaling. FASEB J 2020; 34:7970-7988. [PMID: 32293074 DOI: 10.1096/fj.201902659r] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 03/10/2020] [Accepted: 03/30/2020] [Indexed: 12/19/2022]
Abstract
Cancer-associated fibroblasts (CAFs) are an important component of the tumor microenvironment and contribute to tumor cell proliferation and metastasis. Microfibrillar-associated protein 5 (MFAP5), a component of elastic microfibers and an oncogenic protein in several types of tumors, is secreted by CAFs. However, the role of MFAP5 in the bladder cancer remains unclear. Here, we report that MFAP5 is upregulated in bladder cancer and is associated with poor patient survival. Downregulation of MFAP5 in CAFs led to an impairment in proliferation and invasion of bladder cancer cells. Luciferase reporter assays and electrophoretic mobility shift assays (EMSA) showed QKI directly downregulates MFAP5 in CAFs. In addition, CAFs-derived MFAP5 led to an activation of the NOTCH2/HEY1 signaling pathway through direct interaction with the NOTCH2 receptor, thereby stimulating the N2ICD release. RNA-sequencing revealed that MFAP5-mediated PI3K-AKT signaling activated the DLL4/NOTCH2 pathway axis in bladder cancer. Moreover, downregulation of NOTCH2 by short hairpin RNA or the inactivating anti-body NRR2Mab was able to reverse the adverse effects of MFAP5 stimulation in vitro and in vivo. Together, these results demonstrate CAFs-derived MFAP5 promotes the bladder cancer proliferation and metastasis and provides new insight for targeting CAFs as novel diagnostic and therapeutic strategy.
Collapse
Affiliation(s)
- Zheng Zhou
- Department of Urology, Shanghai General Hospital, Nanjing Medical University, Shanghai, China
| | - Di Cui
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Institute of Urology, Shanghai Jiao Tong University, Shanghai, China
| | - Meng-Hao Sun
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing-Lang Huang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zheng Deng
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bang-Min Han
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Institute of Urology, Shanghai Jiao Tong University, Shanghai, China
| | - Xiao-Wen Sun
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Institute of Urology, Shanghai Jiao Tong University, Shanghai, China
| | - Shu-Jie Xia
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Institute of Urology, Shanghai Jiao Tong University, Shanghai, China
| | - Feng Sun
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Institute of Urology, Shanghai Jiao Tong University, Shanghai, China
| | - Fei Shi
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Institute of Urology, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
23
|
Gu S, Dai H, Zhao X, Gui C, Gui J. AKT3 deficiency in M2 macrophages impairs cutaneous wound healing by disrupting tissue remodeling. Aging (Albany NY) 2020; 12:6928-6946. [PMID: 32291381 PMCID: PMC7202485 DOI: 10.18632/aging.103051] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 03/04/2020] [Indexed: 12/14/2022]
Abstract
AKT signaling and M2 macrophage-guided tissue repair are key factors in cutaneous wound healing. A delay in this process threatens human health worldwide. However, the role of AKT3 in delayed cutaneous wound healing is largely unknown. In this study, histological staining and transcriptomics demonstrated that prolonged tissue remodeling delayed wound healing. This delay was accompanied by defects in AKT3, collagen alpha-1(I) chain (COL1A1), and collagen alpha-1(XI) chain (COL11A1) expression and AKT signaling. The defect in AKT3 expression was M2 macrophage-specific, and decreased AKT3 protein levels were observed in CD68/CD206-positive macrophages from delayed wound tissue. Downregulation of AKT3 in M2 macrophages did not influence cell polarization but impaired collagen organization by inhibiting COL1A1 and COL11A1 expression in human skin fibroblasts (HSFs). Moreover, a co-culture model revealed that the downregulation of AKT3 in the human monocytic cell line (THP-1)-derived M2 macrophages impaired HSF proliferation and migration. Finally, cutaneous wound healing in AKT3-/- mice was much slower than that of AKT3+/+ mice, and F4/80 macrophages from the AKT3-/- mice had an impaired ability to promote wound healing. Thus, the downregulation of AKT3 in M2 macrophages prolonged tissue remodeling and delayed cutaneous wound healing.
Collapse
Affiliation(s)
- Song Gu
- Department of Sports Medicine and Joint Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, P.R. China.,Trauma Center, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, P.R. China
| | - Hanhao Dai
- Department of Sports Medicine and Joint Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, P.R. China
| | - Xilian Zhao
- Department of Sports Medicine and Joint Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, P.R. China
| | - Chang Gui
- Department of Biomedical Engineering, Washington University in St. Louis, MO 63130, USA
| | - Jianchao Gui
- Department of Sports Medicine and Joint Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, P.R. China
| |
Collapse
|
24
|
Wei X, Wang B, Wang Q, Yang X, Yang Y, Fang Z, Yi C, Shi L, Fan X, Tao J, Guo Y, Song D. MiR-362-5p, Which Is Regulated by Long Non-Coding RNA MBNL1-AS1, Promotes the Cell Proliferation and Tumor Growth of Bladder Cancer by Targeting QKI. Front Pharmacol 2020; 11:164. [PMID: 32194406 PMCID: PMC7063466 DOI: 10.3389/fphar.2020.00164] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 02/07/2020] [Indexed: 11/13/2022] Open
Abstract
In this study, we found miR-362-5p was upregulated in bladder cancer tissues and we predicted that QKI is potential a target of miR-362-5p and MBNL1-AS1 might be able to directly target to miR-362-5p. We attempted to evaluate whether miR-362-5p could play its roles in bladder cancer through regulating QKI (quaking) and whether the expression and function of miR-362-5p could be mediated by lncRNA MBNL1-AS1. We performed the gain- and loss-function experiments to explore the association between miR-362-5p expression and bladder cancer proliferation. In vivo, the nude mice were injected with miR-362-5p knockdown SW780 cells to assess the effects of miR-362-5p on tumor growth. The results showed upregulation of miR-362-5p promoted cell proliferation of bladder cancer cells. MBNL1-AS1 and QKI could directly bind with miR-362-5p, and knockdown of MBNL1-AS1 or QKI could abrogate the regulatory effects of miR-362-5p on bladder cancer cell proliferation. Furthermore, downregulation of miR-362-5p inhibited bladder tumor growth and increased QKI expression. Our data unveiled that miR-362-5p may play an oncogenic role in bladder cancer through QKI and MBNL1-AS1 might function as a sponge to mediate the miR-362-5p expression and function.
Collapse
Affiliation(s)
- Xiaosong Wei
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Beibei Wang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qi Wang
- College of Science, The Australian National University, Canberra, ACT, Australia
| | - Xiaoming Yang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yang Yang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhiwei Fang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chengzhi Yi
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lei Shi
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xin Fan
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jin Tao
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yufeng Guo
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Dongkui Song
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
25
|
Shi F, Deng Z, Zhou Z, Jiang C, Zhao R, Sun F, Cui D, Bei X, Yang B, Sun Q, Wang X, Wu Q, Xia S, Han B. QKI-6 inhibits bladder cancer malignant behaviours through down-regulating E2F3 and NF-κB signalling. J Cell Mol Med 2019; 23:6578-6594. [PMID: 31449345 PMCID: PMC6787450 DOI: 10.1111/jcmm.14481] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 04/13/2019] [Accepted: 05/15/2019] [Indexed: 02/06/2023] Open
Abstract
Quaking homolog (QKI) is a member of the RNA-binding signal transduction and activator of proteins family. Previous studies showed that QKI possesses the tumour suppressor activity in human cancers by interacting with the 3'-untraslated region (3'-UTR) of various gene transcripts via the STAR domain. This study first assessed the association of QKI-6 expression with clinicopathological and survival data from bladder cancer patients and then investigated the underlying molecular mechanisms. Bladder cancer tissues (n = 223) were subjected to immunohistochemistry, and tumour cell lines and nude mice were used for different in vitro and in vivo assays following QKI-6 overexpression or knockdown. QKI-6 down-regulation was associated with advanced tumour TNM stages and poor patient overall survival. QKI-6 overexpression inhibited bladder cancer cell growth and invasion capacity, but induced tumour cell apoptosis and cell cycle arrest. Furthermore, ectopic expression of QKI-6 reduced tumour xenograft growth and expression of proliferation markers, Ki67 and PCNA. However, knockdown of QKI-6 expression had opposite effects in vitro and in vivo. QKI-6 inhibited expression of E2 transcription factor 3 (E2F3) by directly binding to the E2F3 3'-UTR, whereas E2F3 induced QKI-6 transcription by binding to the QKI-6 promoter in negative feedback mechanism. QKI-6 expression also suppressed activity and expression of nuclear factor-κB (NF-κB) signalling proteins in vitro, implying a novel multilevel regulatory network downstream of QKI-6. In conclusion, QKI-6 down-regulation contributes to bladder cancer development and progression.
Collapse
Affiliation(s)
- Fei Shi
- Department of Urology, School of MedicineShanghai General Hospital, Shanghai Jiao Tong UniversityShanghaiChina
| | - Zheng Deng
- Department of Urology, School of MedicineShanghai General Hospital, Shanghai Jiao Tong UniversityShanghaiChina
| | - Zheng Zhou
- Department of UrologyShanghai General Hospital Affiliated to Nanjing Medical UniversityShanghaiChina
| | - Chen‐Yi Jiang
- Department of Urology, School of MedicineShanghai General Hospital, Shanghai Jiao Tong UniversityShanghaiChina
| | - Rui‐Zhe Zhao
- Department of Urology, School of MedicineShanghai General Hospital, Shanghai Jiao Tong UniversityShanghaiChina
| | - Feng Sun
- Department of Urology, School of MedicineShanghai General Hospital, Shanghai Jiao Tong UniversityShanghaiChina
- Institute of UrologyShanghai Jiao Tong UniversityShanghaiChina
| | - Di Cui
- Department of Urology, School of MedicineShanghai General Hospital, Shanghai Jiao Tong UniversityShanghaiChina
- Institute of UrologyShanghai Jiao Tong UniversityShanghaiChina
| | - Xiao‐Yu Bei
- Department of Urology, School of MedicineShanghai General Hospital, Shanghai Jiao Tong UniversityShanghaiChina
- Institute of UrologyShanghai Jiao Tong UniversityShanghaiChina
| | - Bo‐Yu Yang
- Department of Urology, School of MedicineShanghai General Hospital, Shanghai Jiao Tong UniversityShanghaiChina
| | - Qian Sun
- Department of UrologyShanghai General Hospital Affiliated to Nanjing Medical UniversityShanghaiChina
| | - Xing‐Jie Wang
- Department of Urology, School of MedicineShanghai General Hospital, Shanghai Jiao Tong UniversityShanghaiChina
- Institute of UrologyShanghai Jiao Tong UniversityShanghaiChina
| | - Qi Wu
- Department of UrologyShanghai General Hospital Affiliated to Nanjing Medical UniversityShanghaiChina
| | - Shu‐Jie Xia
- Department of Urology, School of MedicineShanghai General Hospital, Shanghai Jiao Tong UniversityShanghaiChina
- Institute of UrologyShanghai Jiao Tong UniversityShanghaiChina
| | - Bang‐Min Han
- Department of Urology, School of MedicineShanghai General Hospital, Shanghai Jiao Tong UniversityShanghaiChina
- Institute of UrologyShanghai Jiao Tong UniversityShanghaiChina
| |
Collapse
|