1
|
Wang J, Zhang Y, Li Z. Advancements in Understanding the Role of Circular RNA in Osteosarcoma. Mol Biotechnol 2024; 66:2157-2167. [PMID: 37661210 DOI: 10.1007/s12033-023-00838-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 07/24/2023] [Indexed: 09/05/2023]
Abstract
Osteosarcoma, the most prevalent primary malignant bone tumor and the third most frequent cancer in children and adolescents worldwide, still poses a significant therapeutic challenge. Even though combined chemotherapy and surgical resection have improved survival rates up to 60%, the prognosis for most patients with metastatic osteosarcoma continues to be dismal. The specific pathogenesis and key regulators of tumor invasion and metastasis remain largely elusive. Circular RNAs (circRNAs), novel endogenous non-coding RNA molecules that form covalently closed continuous loops through splicing, play a crucial role in the development, progression, clinical diagnosis, and treatment of various diseases. Recently, an escalating number of circular structures have been identified in osteosarcoma. Understanding their role in osteosarcoma is advantageous for early detection, diagnosis, and treatment of this disease. The primary function of circRNA involves its unique ability to bind specifically to miRNA, although their biological functions also extend to interacting with proteins, regulating gene transcription, and serving as translation templates. In this review, we explore the mechanisms and clinical applications of circRNAs in the pathogenesis and progression of osteosarcoma, with a particular emphasis on the regulatory mechanisms and functions of circRNAs as miRNA sponges in osteosarcoma development.
Collapse
Affiliation(s)
- Jin Wang
- Department of Orthopedics, Wuwei People's Hospital, Xuanwu Street, Liangzhou District, Wuwei, Gansu, 730030, People's Republic of China
| | - Yan Zhang
- Department of Outpatient, Liangzhou District Huangyang Hospital, Wuwei, Gansu, 733000, People's Republic of China
| | - Zicai Li
- Department of Orthopedics, Wuwei People's Hospital, Xuanwu Street, Liangzhou District, Wuwei, Gansu, 730030, People's Republic of China.
| |
Collapse
|
2
|
Wang M, Jin F, Tong X. From bench to bedside: The promising value of exosomes in precision medicine for CNS tumors. Heliyon 2024; 10:e32376. [PMID: 38961907 PMCID: PMC11219334 DOI: 10.1016/j.heliyon.2024.e32376] [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: 01/11/2024] [Revised: 05/31/2024] [Accepted: 06/03/2024] [Indexed: 07/05/2024] Open
Abstract
Exosomes are naturally present extracellular vesicles (EVs) released into the surrounding body fluids upon the fusion of polycystic and plasma membranes. They facilitate intercellular communication by transporting DNA, mRNA, microRNA, long non-coding RNA, circular RNA, proteins, lipids, and nucleic acids. They contribute to the onset and progression of Central Nervous System (CNS) tumors. In addition, they can be used as biomarkers of tumor proliferation, migration, and blood vessel formation, thereby affecting the Tumor Microenvironment (TME). This paper reviews the recent advancements in the diagnosis and treatment of exosomes in various CNS tumors, the promise and challenges of exosomes as natural carriers of CNS tumors, and the therapeutic prospects of exosomes in CNS tumors. Furthermore, we hope this research can contribute to the development of more targeted and effective treatments for central nervous system tumors.
Collapse
Affiliation(s)
- Mengjie Wang
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, 300070, China
| | - Feng Jin
- Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Hospital).266042, Qingdao, Shandong, China
| | - Xiaoguang Tong
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, 300070, China
| |
Collapse
|
3
|
Lin S, Chen Q, Tan C, Su M, Min L, Ling L, Zhou J, Zhu T. ZEB family is a prognostic biomarker and correlates with anoikis and immune infiltration in kidney renal clear cell carcinoma. BMC Med Genomics 2024; 17:153. [PMID: 38840097 PMCID: PMC11151722 DOI: 10.1186/s12920-024-01895-7] [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: 12/27/2023] [Accepted: 04/28/2024] [Indexed: 06/07/2024] Open
Abstract
BACKGROUND Zinc finger E-box binding homEeobox 1 (ZEB1) and ZEB2 are two anoikis-related transcription factors. The mRNA expressions of these two genes are significantly increased in kidney renal clear cell carcinoma (KIRC), which are associated with poor survival. Meanwhile, the mechanisms and clinical significance of ZEB1 and ZEB2 upregulation in KIRC remain unknown. METHODS Through the Cancer Genome Atlas (TCGA) database and Gene Expression Omnibus (GEO) database, expression profiles, prognostic value and receiver operating characteristic curves (ROCs) of ZEB1 and ZEB2 were evaluated. The correlations of ZEB1 and ZEB2 with anoikis were further assessed in TCGA-KIRC database. Next, miRTarBase, miRDB, and TargetScan were used to predict microRNAs targeting ZEB1 and ZEB2, and TCGA-KIRC database was utilized to discern differences in microRNAs and establish the association between microRNAs and ZEBs. TCGA, TIMER, TISIDB, and TISCH were used to analyze tumor immune infiltration. RESULTS It was found that ZEB1 and ZEB2 expression were related with histologic grade in KIRC patient. Kaplan-Meier survival analyses showed that KIRC patients with low ZEB1 or ZEB2 levels had a significantly lower survival rate. Meanwhile, ZEB1 and ZEB2 are closely related to anoikis and are regulated by microRNAs. We constructed a risk model using univariate Cox and LASSO regression analyses to identify two microRNAs (hsa-miR-130b-3p and hsa-miR-138-5p). Furthermore, ZEB1 and ZEB2 regulate immune cell invasion in KIRC tumor microenvironments. CONCLUSIONS Anoikis, cytotoxic immune cell infiltration, and patient survival outcomes were correlated with ZEB1 and ZEB2 mRNA upregulation in KIRC. ZEB1 and ZEB2 are regulated by microRNAs.
Collapse
Affiliation(s)
- Sheng Lin
- Department of Laboratory Medicine, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Qi Chen
- Department of Urology, Foshan First People's Hospital, Foshan City, Guangdong Province, China
| | - Canliang Tan
- Department of general surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong Province, China
| | - Manyi Su
- Department of Laboratory Medicine, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Ling Min
- Department of Laboratory Medicine, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Lv Ling
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Junhao Zhou
- Department of general surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong Province, China.
- KingMed school of Laboratory Medicine, Guangzhou Medical University, Guangzhou, Guangdong Province, China.
| | - Ting Zhu
- Department of Laboratory Medicine, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong Province, China.
- KingMed school of Laboratory Medicine, Guangzhou Medical University, Guangzhou, Guangdong Province, China.
| |
Collapse
|
4
|
Wang C, Fu R, Wang Y, Wei J, Yu Y, Hu L, Zhang C. miR-124-3p and miR-194-5p regulation of the PI3K/AKT pathway via ROR2 in medulloblastoma progression. Cancer Gene Ther 2024; 31:941-954. [PMID: 38632356 PMCID: PMC11192632 DOI: 10.1038/s41417-024-00762-y] [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: 12/25/2023] [Revised: 03/01/2024] [Accepted: 03/07/2024] [Indexed: 04/19/2024]
Abstract
Medulloblastoma (MB), a prevalent pediatric central nervous system tumor, is influenced by microRNAs (miRNAs) that impact tumor initiation and progression. However, the specific involvement of miRNAs in MB tumorigenesis remains unclear. Using single-cell RNA sequencing, we identified ROR2 expression in normal human fetal cerebellum. Subsequent analyses, including immunofluorescence, quantitative real-time PCR (qRT-PCR), and Western blot, assessed ROR2 expression in MB tissues and cell lines. We investigated miR-124-3p and miR-194-5p and their regulatory role in ROR2 expression through the dual-luciferase reporter, qRT-PCR, and western blot assays. Mechanistic insights were gained through functional assays exploring the impact of miR-124-3p, miR-194-5p, and ROR2 on MB growth in vitro and in vivo. We observed significantly reduced miR-124-3p and miR-194-5p expression and elevated ROR2 expression in MB tissues and cell lines. High ROR2 expression inversely correlated with overall survival in WNT and SHH subgroups of MB patients. Functionally, overexpressing miR-124-3p and miR-194-5p and inhibiting ROR2 suppressed in vitro malignant transformation and in vivo tumorigenicity. Mechanistically, miR-124-3p and miR-194-5p synergistically regulated the ROR2/PI3K/Akt pathway, influencing MB progression. Our findings indicate that miR-124-3p and miR-194-5p function as tumor suppressors, inhibiting MB progression via the ROR2/PI3K/Akt axis, suggesting a key mechanism and therapeutic targets for MB patients.
Collapse
Affiliation(s)
- Chen Wang
- Department of Pediatric Neurosurgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Runxi Fu
- Department of Pediatric Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute for Pediatric Research, Shanghai, China
| | - Yunkun Wang
- Department of Pediatric Neurosurgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jia Wei
- Department of Pediatric Neurosurgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Yu
- Department of Pediatric Neurosurgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liuhua Hu
- Department of Cardiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Chenran Zhang
- Department of Pediatric Neurosurgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
5
|
Wu Z, Zhang H, Chen X, Zhang P, Fang J, Yang S, Chen H, Ji J, Chen L, Zheng Y, Yu D, Zhao Y. miR-145a-5p/SIK1/cAMP-dependent alteration of synaptic structural plasticity drives cognitive impairment induced by coke oven emissions. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 277:116401. [PMID: 38677069 DOI: 10.1016/j.ecoenv.2024.116401] [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: 01/17/2024] [Revised: 04/22/2024] [Accepted: 04/24/2024] [Indexed: 04/29/2024]
Abstract
Exposure to fine particulate matter (PM) is associated with the neurodegenerative diseases. Coke oven emissions (COEs) in occupational environment are important sources of PM. However, its neurotoxicity is still unclear. Therefore, evaluating the toxicological effects of COE on the nervous system is necessary. In the present study, we constructed mouse models of COE exposure by tracheal instillation. Mice exposed to COE showed signs of cognitive impairment. This was accompanied by a decrease in miR-145a-5p and an increase in SIK1 expression in the hippocampus, along with synaptic structural damage. Our results demonstrated that COE-induced miR-145a-5p downregulation could increase the expression of SIK1 and phosphorylated SIK1, inhibiting the cAMP/PKA/CREB pathway by activating PDE4D, which was associated with reduced synaptic structural plasticity. Furthermore, restoring of miR-145a-5p expression based on COE exposure in HT22 cells could partially reversed the negative effects of COE exposure through the SIK1/PDE4D/cAMP axis. Collectively, our findings link epigenetic regulation with COE-induced neurotoxicity and imply that miR-145a-5p could be an early diagnostic marker for neurological diseases in patients with COE occupational exposure.
Collapse
Affiliation(s)
- Zhaoxu Wu
- Department of Toxicology, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Heng Zhang
- Department of Toxicology, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Xian Chen
- Department of Toxicology, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Pimei Zhang
- Department of Toxicology, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Jiacheng Fang
- Department of Nutrition, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Shuaishuai Yang
- Department of Toxicology, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Hongguang Chen
- Department of Toxicology, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Jing Ji
- Department of Toxicology, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Lei Chen
- Department of Nutrition, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Yuxin Zheng
- Department of Toxicology, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Dianke Yu
- Department of Toxicology, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Yanjie Zhao
- Department of Toxicology, School of Public Health, Qingdao University, Qingdao 266071, China.
| |
Collapse
|
6
|
Singhto N, Pongphitcha P, Jinawath N, Hongeng S, Chutipongtanate S. Extracellular Vesicles for Childhood Cancer Liquid Biopsy. Cancers (Basel) 2024; 16:1681. [PMID: 38730633 PMCID: PMC11083250 DOI: 10.3390/cancers16091681] [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: 04/09/2024] [Revised: 04/22/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
Liquid biopsy involves the utilization of minimally invasive or noninvasive techniques to detect biomarkers in biofluids for disease diagnosis, monitoring, or guiding treatments. This approach is promising for the early diagnosis of childhood cancer, especially for brain tumors, where tissue biopsies are more challenging and cause late detection. Extracellular vesicles offer several characteristics that make them ideal resources for childhood cancer liquid biopsy. Extracellular vesicles are nanosized particles, primarily secreted by all cell types into body fluids such as blood and urine, and contain molecular cargos, i.e., lipids, proteins, and nucleic acids of original cells. Notably, the lipid bilayer-enclosed structure of extracellular vesicles protects their cargos from enzymatic degradation in the extracellular milieu. Proteins and nucleic acids of extracellular vesicles represent genetic alterations and molecular profiles of childhood cancer, thus serving as promising resources for precision medicine in cancer diagnosis, treatment monitoring, and prognosis prediction. This review evaluates the recent progress of extracellular vesicles as a liquid biopsy platform for various types of childhood cancer, discusses the mechanistic roles of molecular cargos in carcinogenesis and metastasis, and provides perspectives on extracellular vesicle-guided therapeutic intervention. Extracellular vesicle-based liquid biopsy for childhood cancer may ultimately contribute to improving patient outcomes.
Collapse
Affiliation(s)
- Nilubon Singhto
- Ramathibodi Comprehensive Cancer Center, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand;
| | - Pongpak Pongphitcha
- Bangkok Child Health Center, Bangkok Hospital Headquarters, Bangkok 10130, Thailand;
- Division of Hematology and Oncology, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand;
| | - Natini Jinawath
- Program in Translational Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand;
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan 10540, Thailand
- Integrative Computational Biosciences Center, Mahidol University, Nakon Pathom 73170, Thailand
| | - Suradej Hongeng
- Division of Hematology and Oncology, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand;
| | - Somchai Chutipongtanate
- MILCH and Novel Therapeutics Laboratory, Division of Epidemiology, Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
- Extracellular Vesicle Working Group, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| |
Collapse
|
7
|
Yang Z, Tang Y, Wu X, Wang J, Yao W. MicroRNA-130b Suppresses Malignant Behaviours and Inhibits the Activation of the PI3K/Akt Signaling Pathway by Targeting MET in Pancreatic Cancer. Biochem Genet 2024:10.1007/s10528-024-10696-7. [PMID: 38607540 DOI: 10.1007/s10528-024-10696-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 01/10/2024] [Indexed: 04/13/2024]
Abstract
There has been interested in the microRNAs' roles in pancreatic cancer (PC) cell biology, particularly in regulating pathways related to tumorigenesis. The study aimed to explore the hub miRNAs in PC and underlying mechanisms by bioinformatics and fundamental experiments. RNA datasets collected from the Gene Expression Omnibus were analysed to find out differentially expressed RNAs (DERNAs). The miRNA-mRNA and protein-protein interaction (PPI) networks were built. The clinicopathological features and expressions of hub miRNAs and hub mRNAs were explored. Dual-luciferase reporter gene assay was performed to assess the interaction between microRNA and target gene. RT-qPCR and western blot were employed to explore RNA expression. The roles of RNA were detected by CCK-8 test, wound healing, transwell, and flow cytometry experiment. We verified 40 DEmiRNAs and 1613 DEmRNAs, then detected a total of 69 final functional mRNAs (FmRNAs) and 23 DEmiRNAs. In the miRNA-mRNA networks, microRNA-130b (miR-130b) was the hub RNA with highest degrees. Clinical analysis revealed that miR-130b was considerably lower expressed in cancerous tissues than in healthy ones, and patients with higher-expressed miR-130b had a better prognosis. Mechanically, miR-130b directly targeted MET in PC cells. Cell functional experiments verified that miR-130b suppressed cell proliferation, migration, promoted apoptosis, and inhibited the PI3K/Akt pathway by targeting MET in PC cells. Our findings illustrated the specific molecular mechanism of miR-130b regulating PC progress. The miR-130b/MET axis may be an alternative target in the therapeutic intervention of PC and provide an opportunity to deepen our understanding of the pathogenesis of PC.
Collapse
Affiliation(s)
- Zilin Yang
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yuming Tang
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xuejiao Wu
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jiancheng Wang
- Department of General Surgery, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Weiyan Yao
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
8
|
Verma P, Joshi H, Singh T, Sharma B, Sharma U, Ramniwas S, Rana R, Gupta M, Kaur G, Tuli HS. Temozolomide and flavonoids against glioma: from absorption and metabolism to exosomal delivery. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:41-57. [PMID: 37566307 DOI: 10.1007/s00210-023-02660-w] [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: 02/12/2023] [Accepted: 08/01/2023] [Indexed: 08/12/2023]
Abstract
Patients with glioblastoma multiforme and anaplastic astrocytoma are treated with temozolomide. Although it has been demonstrated that temozolomide increases GBM patient survival, it has also been connected to negative immune-related adverse effects. Numerous research investigations have shown that flavonoids have strong antioxidant and chemo-preventive effects. Consequently, it might lessen chemotherapeutic medicines' side effects while also increasing therapeutic effectiveness. The need for creating innovative, secure, and efficient drug carriers for cancer therapy has increased over time. Recent research indicates that exosomes have enormous potential to serve as carriers and cutting-edge drug delivery systems to the target cell. In recent years, researchers have been paying considerable attention to exosomes because of their favorable biodistribution, biocompatibility, and low immunogenicity. In the present review, the mechanistic information of the anti-glioblastoma effects of temozolomide and flavonoids coupled with their exosomal delivery to the targeted cell has been discussed. In addition, we discuss the safety aspects of temozolomide and flavonoids against glioma. The in-depth information of temozolomide and flavonoids action via exosomal delivery can unravel novel strategies to target Glioma.
Collapse
Affiliation(s)
- Priyanka Verma
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala, 133207, India
| | - Hemant Joshi
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Tejveer Singh
- Translational Oncology Laboratory, Department of Zoology, Hansraj College, Delhi University, New Delhi, 110007, India
| | - Bunty Sharma
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala, 133207, India
| | - Ujjawal Sharma
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bhatinda, 151001, India
| | - Seema Ramniwas
- University Centre for Research and Development, University Institute of Pharmaceutical Sciences, Chandigarh University, Gharuan, Mohali, 140413, India
| | - Rashmi Rana
- Department of Research, Sir Ganga Ram Hospital, New Delhi, 122016, India.
| | - Madhu Gupta
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, 110017, India
| | - Ginpreet Kaur
- Department of Pharmacology, Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, Vile Parle-West, Mumbai, 400056, India
| | - Hardeep Singh Tuli
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala, 133207, India.
| |
Collapse
|
9
|
Liang H, Lu Y, Huang X, Ye T. LncRNA BRCAT54 is downregulated and inhibits cancer cell proliferation by downregulating miR-130b-3p through methylation in prostate cancer. J Biochem Mol Toxicol 2024; 38:e23552. [PMID: 37888834 DOI: 10.1002/jbt.23552] [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: 06/02/2022] [Revised: 09/08/2023] [Accepted: 10/02/2023] [Indexed: 10/28/2023]
Abstract
BRCAT54 and miR-130b-3p are two recently characterized critical players in cancer biology, while their functions in prostate cancer (PC) are unknown. From preliminary sequencing analysis, we observed altered expression of BRCAT54 and miR-130b-3p in PC and an inverse correlation between them. This study was conducted to explore their involvement in PC. A total of 64 PC patients were enrolled to collect paired PC and nontumor tissues. The expression of BRCAT54 and miR-130b-3p were determined by RT-qPCR. Overexpression of BRCAT54 and miR-130b-3p was achieved in PC cells to explore their roles in regulating the expression of each other. Methylation-specific PCR (MSP) was conducted to explore the role of BRCAT54 in regulating promoter methylation of miR-130b-3p. BrdU assay was used to evaluate the role of BRCAT54 and miR-130b-3p in regulating PC cell proliferation. The results showed that PC tissues exhibited downregulation of BRCAT54 and upreglation of miR-130b-3p compared to that in nontumor tissues. They were inversely correlated across PC tissue samples. Overexpression of BRCAT54 decreased RNA accumulation of miR-130b-3p in PC cells. In addition, overexpression of BRCAT54 increased promoter methylation of miR-130b-3p. Moreover, BRCAT54 suppressed the role of miR-130b-3p in promoting PC cell proliferation. In conclusion, BRCAT54 is downregulated in PC and it may inhibit cancer cell proliferation by downregulating miR-130b-3p through methylation.
Collapse
Affiliation(s)
- Haote Liang
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yongyong Lu
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xixi Huang
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Tingyu Ye
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
10
|
Feng S, Wei F, Shi H, Chen S, Wang B, Huang D, Luo L. Roles of salt‑inducible kinases in cancer (Review). Int J Oncol 2023; 63:118. [PMID: 37654200 PMCID: PMC10546379 DOI: 10.3892/ijo.2023.5566] [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: 04/13/2023] [Accepted: 08/01/2023] [Indexed: 09/02/2023] Open
Abstract
Salt inducible kinases (SIKs) with three subtypes SIK1, SIK2 and SIK3, belong to the AMP‑activated protein kinase family. They are expressed ubiquitously in humans. Under normal circumstances, SIK1 regulates adrenocortical function in response to high salt or adrenocorticotropic hormone stimulation, SIK2 is involved in cell metabolism, controlling insulin signaling and gluconeogenesis and SIK3 coordinates with the mTOR complex, promoting cancer. The dysregulation of SIKs has been widely detected in various types of cancers. Based on most of the existing studies, SIK1 is mostly considered a tumor inhibitor, SIK2 and SIK3 are usually associated with tumor promotion. However, the functions of SIKs have shown contradictory in certain tumors, suggesting that SIKs cannot be simply classified as oncogenes or tumor suppressor genes. The present review provided a comprehensive summary of the roles of SIKs in the initiation and progression of different cancers, aiming to elucidate their clinical value and discuss potential strategies for targeting SIKs in cancer therapy.
Collapse
Affiliation(s)
- Shenghui Feng
- Department of Gastroenterology, Research Institute of Digestive Diseases, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Queen Mary School, Medical Department, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Fangyi Wei
- Department of Gastroenterology, Research Institute of Digestive Diseases, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Queen Mary School, Medical Department, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Haoran Shi
- Department of Gastroenterology, Research Institute of Digestive Diseases, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Shen Chen
- Department of Gastroenterology, Research Institute of Digestive Diseases, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Queen Mary School, Medical Department, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Bangqi Wang
- Department of Gastroenterology, Research Institute of Digestive Diseases, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Queen Mary School, Medical Department, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Deqiang Huang
- Department of Gastroenterology, Research Institute of Digestive Diseases, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Lingyu Luo
- Department of Gastroenterology, Research Institute of Digestive Diseases, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
11
|
Mu C, Dang X, Luo XJ. Mendelian randomization reveals the causal links between microRNA and schizophrenia. J Psychiatr Res 2023; 163:372-377. [PMID: 37267734 DOI: 10.1016/j.jpsychires.2023.05.071] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 04/12/2023] [Accepted: 05/25/2023] [Indexed: 06/04/2023]
Abstract
MicroRNAs have pivotal roles in gene regulation. However, microRNAs that have causal effects on schizophrenia remain largely unknown. To investigate the causal relationships between microRNAs and schizophrenia, here we conduct a Mendelian randomization (MR) study. The genome-wide association study (GWAS) of schizophrenia (67,390 cases and 94,015 controls) from PGC3 were used as the outcome. Genetic variants associated with microRNAs were used as exposure in MR analysis. We identified 6 microRNAs that showed causality on schizophrenia. These microRNAs include hsa-miR-570-3p (OR = 1.03, 95% confidence interval (CI): 1.02 to 1.05, P = 5.45 × 10-5), hsa-miR-550a-3p (OR = 1.12, 95% CI: 1.06 to 1.18, P = 5.99 × 10-5), hsa-miR-130a-3p (OR = 1.10, 95% CI: 1.05 to 1.15, P = 1.58 × 10-4), hsa-miR-210 (OR = 0.87, 95% CI: 0.82 to 0.93, P = 3.09 × 10-5), hsa-miR-337-3p (OR = 1.01, 95% CI: 1.01 to 1.02, P = 3.39 × 10-4), and hsa-miR-130b-3p (OR = 0.89, 95% CI: 0.84 to 0.94, P = 1.50 × 10-5). Differential expression analysis showed dysregulation of hsa-miR-130b-3p in schizophrenia cases compared with controls. Gene Ontology (GO) analysis showed that the targets of these causal microRNAs were significantly enriched in RNA splicing pathways. This MR study identified six microRNAs whose genetically regulated expression might have a causal role in schizophrenia, indicating the causality of these microRNAs in schizophrenia. Our findings also indicate that these microRNAs may be used as potential biomarkers for schizophrenia.
Collapse
Affiliation(s)
- Changgai Mu
- Zhongda Hospital, School of Medicine, Advanced Institute for Life and Health, Southeast University, Nanjing, Jiangsu, 210096, China; Department of Neurology, Affiliated Zhongda Hospital, Southeast University, Nanjing, Jiangsu, 210096, China
| | - Xinglun Dang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650204, China
| | - Xiong-Jian Luo
- Zhongda Hospital, School of Medicine, Advanced Institute for Life and Health, Southeast University, Nanjing, Jiangsu, 210096, China; Department of Neurology, Affiliated Zhongda Hospital, Southeast University, Nanjing, Jiangsu, 210096, China.
| |
Collapse
|
12
|
Jackson HK, Mitoko C, Linke F, Macarthur D, Kerr ID, Coyle B. Extracellular Vesicles Potentiate Medulloblastoma Metastasis in an EMMPRIN and MMP-2 Dependent Manner. Cancers (Basel) 2023; 15:cancers15092601. [PMID: 37174066 PMCID: PMC10177484 DOI: 10.3390/cancers15092601] [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: 03/27/2023] [Revised: 04/19/2023] [Accepted: 04/23/2023] [Indexed: 05/15/2023] Open
Abstract
Extracellular vesicles (EVs) have emerged as pivotal mediators of communication in the tumour microenvironment. More specifically, nanosized extracellular vesicles termed exosomes have been shown to contribute to the establishment of a premetastatic niche. Here, we sought to determine what role exosomes play in medulloblastoma (MB) progression and elucidate the underlying mechanisms. Metastatic MB cells (D458 and CHLA-01R) were found to secrete markedly more exosomes compared to their nonmetastatic, primary counterparts (D425 and CHLA-01). In addition, metastatic cell-derived exosomes significantly enhanced the migration and invasiveness of primary MB cells in transwell migration assays. Protease microarray analysis identified that matrix metalloproteinase-2 (MMP-2) was enriched in metastatic cells, and zymography and flow cytometry assays of metastatic exosomes demonstrated higher levels of functionally active MMP-2 on their external surface. Stable genetic knockdown of MMP-2 or extracellular matrix metalloproteinase inducer (EMMPRIN) in metastatic MB cells resulted in the loss of this promigratory effect. Analysis of serial patient cerebrospinal fluid (CSF) samples showed an increase in MMP-2 activity in three out of four patients as the tumour progressed. This study demonstrates the importance of EMMPRIN and MMP-2-associated exosomes in creating a favourable environment to drive medulloblastoma metastasis via extracellular matrix signalling.
Collapse
Affiliation(s)
- Hannah K Jackson
- Children's Brain Tumour Research Centre, School of Medicine, Biodiscovery Institute, University Park, University of Nottingham, Nottingham NG7 2RD, UK
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK
| | - Christine Mitoko
- Children's Brain Tumour Research Centre, School of Medicine, Biodiscovery Institute, University Park, University of Nottingham, Nottingham NG7 2RD, UK
| | - Franziska Linke
- Children's Brain Tumour Research Centre, School of Medicine, Biodiscovery Institute, University Park, University of Nottingham, Nottingham NG7 2RD, UK
- Department of Experimental Urology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Donald Macarthur
- Department of Neurosurgery, Nottingham University Hospital, Nottingham NG7 2UH, UK
| | - Ian D Kerr
- School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK
| | - Beth Coyle
- Children's Brain Tumour Research Centre, School of Medicine, Biodiscovery Institute, University Park, University of Nottingham, Nottingham NG7 2RD, UK
| |
Collapse
|
13
|
Ding N, Song X, Yu H, Wang J, Huang L, Zhou Y, He X. Mechanism of Exosomal LncRNA PART1 in Esophageal Cancer Angiogenesis by Targeting miR-302a-3p/CDC25A Axis. Technol Cancer Res Treat 2023; 22:15330338231184327. [PMID: 37386808 PMCID: PMC10333641 DOI: 10.1177/15330338231184327] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 05/16/2023] [Accepted: 06/05/2023] [Indexed: 07/01/2023] Open
Abstract
OBJECTIVE LncRNA PART1 has been confirmed related to multiple cancer bioactivities mediated with vascular endothelial growth factor signaling. Nevertheless, the role of LncRNA PART1 in esophageal cancer induced angiogenesis remains unclear. The present work focused on assessing LncRNA PART1 effects on esophageal cancer-induced angiogenesis and exploring possible mechanisms. METHODS Western blot and immunofluorescence were conducted for identifying EC9706 exosomes. MiR-302a-3p and LncRNA PART1 levels were assessed by real-time quantitative polymerase chain reaction. Cell Counting Kit-8, EdU, wound healing, transwell, and tubule information were adopted for detecting human umbilical vein endothelial cell viability, proliferation, migration, invasion, and tubule information, respectively. Starbase software and dual-luciferase reporter were conducted for predicting and judging the expression interrelation of LncRNA PART1 and its potential target-miR-302a-3p. The same methods were carried out for verifying the inhibiting influences of miR-302a-3p upregulation and its potential target-cell division cycle 25 A. RESULTS LncRNA PART1 levels were upregulated and related to the overall survival of patients in esophageal cancer. EC9706-Exos accelerated human umbilical vein endothelial cell proliferation, migration, invasion, and tubule formation via LncRNA PART1. LncRNA PART1 served as a sponge of miR-302a-3p, then miR-302a-3p targeted cell division cycle 25 A, and EC9706-Exos accelerated human umbilical vein endothelial cell angiogenesis via LncRNA PART1/ miR-302a-3p/cell division cycle 25 A axis. CONCLUSION EC9706-Exos accelerates human umbilical vein endothelial cell angiogenesis via LncRNA PART1/miR-302a-3p/ cell division cycle 25 A axis, indicating EC9706-Exos may act as a promoter of angiogenesis. Our research will contribute to clarify the mechanism of tumor angiogenesis.
Collapse
Affiliation(s)
- Naixin Ding
- Department of Radiotherapy, Jiangsu
Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated
Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Xue Song
- Department of Radiotherapy, Jiangsu
Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated
Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Hongliang Yu
- Department of Radiotherapy, Jiangsu
Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated
Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Jie Wang
- Department of Tumor Biobank, Jiangsu
Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated
Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Lei Huang
- Department of Radiotherapy, Jiangsu
Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated
Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Yiqin Zhou
- Department of Radiotherapy, Jiangsu
Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated
Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Xia He
- Department of Radiotherapy, Jiangsu
Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated
Cancer Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
14
|
Qian C, Wang Y, Ji Y, Chen D, Wang C, Zhang G, Wang Y. Neural stem cell‑derived exosomes transfer miR‑124‑3p into cells to inhibit glioma growth by targeting FLOT2. Int J Oncol 2022; 61:115. [PMID: 35929514 PMCID: PMC9387557 DOI: 10.3892/ijo.2022.5405] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 06/08/2022] [Indexed: 11/24/2022] Open
Abstract
Currently, exosomes (EXOs) are being explored as novel drug delivery carriers with greater advantages, including crossing the blood-brain-barrier and loading drugs. The present study utilized EXOs derived from neural stem cells (NSCs) for the delivery of molecular drugs to treat gliomas. miR-124-3p was selected according to previous studies by the authors, and the effects of the delivery of miR-124-3p to glioma cells by NSC-EXOs in vitro and in vivo were evaluated. It was found that NSC-EXOs successfully delivered miR-124-3p into glioma cells, and NSC-EXOs loaded with miR-124-3p significantly inhibited glioma cell proliferation, invasion and migration. Furthermore, the delivery of miR-124-3p by NSC-EXOs suppressed flotillin 2 (FLOT2) expression by specifically binding to the 3' untranslated region of the FLOT2 gene in gliomas; subsequently, AKT1 was found to be associated with the EXO-miR-124-3p/FLOT2 pathway. Moreover, the therapeutic effects of the delivery of miR-124-3p by NSC-EXOs were confirmed in a mouse tumor xenograft model of glioma. Thus, bio-carrier NSC-EXOs loaded with miR-124-3p suppressed glioma growth via the EXO-miR-124-3p/FLOT2/AKT1 pathway. On the whole, the present study provides insight into stem cell-free molecular-targeted therapy based on bio-carrier NSC-EXOs and provides a potential strategy for the treatment of glioma.
Collapse
Affiliation(s)
- Cheng Qian
- Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510260, P.R. China
| | - You Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510260, P.R. China
| | - Yunxiang Ji
- Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510260, P.R. China
| | - Danmin Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510260, P.R. China
| | - Chuanfang Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510260, P.R. China
| | - Guilong Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510260, P.R. China
| | - Yezhong Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510260, P.R. China
| |
Collapse
|
15
|
Exosomal microRNA-25 released from cancer cells targets SIK1 to promote hepatocellular carcinoma tumorigenesis. Dig Liver Dis 2022; 54:954-963. [PMID: 34384713 DOI: 10.1016/j.dld.2021.07.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 07/05/2021] [Accepted: 07/19/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is recognized as a leading cause of cancer-associated fatality worldwide. Our study here aimed to probe the mechanism by which exosomes secreted by CSQT-2, an HCC cell line, affected the progression of HCC. METHODS Exosomes were extracted from CSQT-2 cells. Colony formation, Transwell, sphere formation and flow cytometric analyses were applied to assess cell biological activities. Microarray analysis detected the change of microRNA (miRNA) expression after exosome treatment, followed by RT-qPCR validation. Luciferase reporter was applied to detect the binding between SIK1 and miR-25. Xenograft studies in nude mice manifested tumor growth and metastatic ability of miR-25 and SIK1. RESULTS The exosome treatment enhanced cell malignant phenotype in vitro and tumor growth and liver and lung metastases in vivo. The exosomes elevated miR-25 expression in HCC cells. miR-25 targeted SIK1 which was decreased in the exosomes-treated cells. miR-25 inhibitor reduced cell malignant phenotype and attenuated tumorigenesis and metastasis in vivo. SIK1 silencing reversed the effect of miR-25 inhibitor. The exosome treatment potentiated the Wnt/β-catenin pathway in cells, whereas miR-25 inhibitor blunted the pathway activity. CONCLUSION MiR-25 shuttled through CSQT-2-derived exosomes promoted the development of HCC by reducing SIK1 expression and potentiating the Wnt/β-catenin pathway.
Collapse
|
16
|
Hartono AB, Kang HJ, Shi L, Phipps W, Ungerleider N, Giardina A, Chen W, Spraggon L, Somwar R, Moroz K, Drewry DH, Burow ME, Flemington E, Ladanyi M, Lee SB. Salt-Inducible Kinase 1 is a potential therapeutic target in Desmoplastic Small Round Cell Tumor. Oncogenesis 2022; 11:18. [PMID: 35443736 PMCID: PMC9021191 DOI: 10.1038/s41389-022-00395-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 03/28/2022] [Accepted: 04/01/2022] [Indexed: 11/10/2022] Open
Abstract
Desmoplastic Small Round Cell Tumor (DSRCT) is a rare and aggressive malignant cancer caused by a chromosomal translocation t(11;22)(p13;q12) that produces an oncogenic transcription factor, EWSR1-WT1. EWSR1-WT1 is essential for the initiation and progression of DSRCT. However, the precise mechanism by which EWSR1-WT1 drives DSRCT oncogenesis remains unresolved. Through our integrative gene expression analysis, we identified Salt Inducible Kinase 1 (SIK1) as a direct target of EWSR1-WT1. SIK1 as a member of the AMPK related kinase is involved in many biological processes. We showed that depletion of SIK1 causes inhibition of tumor cell growth, similar to the growth inhibition observed when EWSR1-WT1 is depleted. We further showed that silencing SIK1 leads to cessation of DNA replication in DSRCT cells and inhibition of tumor growth in vivo. Lastly, combined inhibition of SIK1 and CHEK1with small molecule inhibitors, YKL-05-099 and prexasertib, respectively, showed enhanced cytotoxicity in DSRCT cells compared to inhibition of either kinases alone. This work identified SIK1 as a new potential therapeutic target in DSRCT and the efficacy of SIK1 inhibition may be improved when combined with other intervention strategies.
Collapse
Affiliation(s)
- Alifiani Bonita Hartono
- Tulane University School of Medicine, Department of Pathology and Laboratory Medicine, New Orleans, LA, USA
| | - Hong-Jun Kang
- Tulane University School of Medicine, Department of Pathology and Laboratory Medicine, New Orleans, LA, USA
| | - Lawrence Shi
- Tulane University School of Medicine, Department of Pathology and Laboratory Medicine, New Orleans, LA, USA
| | - Whitney Phipps
- Tulane University School of Medicine, Department of Pathology and Laboratory Medicine, New Orleans, LA, USA
| | - Nathan Ungerleider
- Tulane University School of Medicine, Department of Pathology and Laboratory Medicine, New Orleans, LA, USA
| | - Alexandra Giardina
- Tulane University School of Medicine, Department of Pathology and Laboratory Medicine, New Orleans, LA, USA
| | - WeiPing Chen
- Genomics Core, National Institute of Diabetes and Digestive and Kidney Diseases, Maryland, USA
| | - Lee Spraggon
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Romel Somwar
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Krzysztof Moroz
- Tulane University School of Medicine, Department of Pathology and Laboratory Medicine, New Orleans, LA, USA
| | - David H Drewry
- University of North Carolina, Eshelman School of Pharmacy, Chapel Hill, NC, USA
| | | | - Erik Flemington
- Tulane University School of Medicine, Department of Pathology and Laboratory Medicine, New Orleans, LA, USA
| | - Marc Ladanyi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sean Bong Lee
- Tulane University School of Medicine, Department of Pathology and Laboratory Medicine, New Orleans, LA, USA.
| |
Collapse
|
17
|
Wang H, Wang Y, Wang Y. MiR-222-3p inhibits formation of medulloblastoma stem-like cells by targeting Notch2/c-myc signaling pathway. Immunopharmacol Immunotoxicol 2022; 44:548-555. [PMID: 35379056 DOI: 10.1080/08923973.2022.2062381] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND Medulloblastoma (MB) is an embryonal tumor of the cerebellum, which commonly occurs in childhood. Herein, we investigated the effects of miR-222-3p on the formation of MB stem-like cells via the Notch2/c-myc pathway. METHODS Quantitative real-time PCR (qRT-PCR) or western blotting was performed to determine the expression of miR-222-3p and Notch2, c-myc, proliferating cell nuclear antigen (PCNA), and caspase-3. Luciferase reporter gene, RNA immunoprecipitation (RIP), and RNA pull-down assay were applied to confirm the interaction between miR-222-3p and Notch2. Cell growth was examined by Cell Counting Kit-8. Cell cycle distribution and the number of stem cell marker CD133+ cells were examined using flow cytometry. The sphere formation assay was performed. RESULTS miR-222-3p expression was decreased and Notch2 expression was increased in human medulloblastoma cells. miR-222-3p overexpression inhibited cell viability and the sphere formation, induced cell cycle arrest, decreased the number of CD133+ cells, and up-regulated caspase-3 expression and down-regulated PCNA, Notch2, and c-myc expression. However, Notch2 overexpression counteracted these effects of miR-222-3p overexpression. Simultaneous overexpression of Notch2 and miR-222-3p increased the c-myc promoter luciferase activity which was decreased by miR-222-3p overexpression. Luciferase reporter gene, RIP, and RNA pull-down assay revealed that miR-222-3p targeted Notch2. CONCLUSION MiR-222-3p suppressed cell viability, altered cell cycle distribution, and inhibited the formation of MB stem-like cells via the Notch2/c-myc pathway.
Collapse
Affiliation(s)
- Hongxin Wang
- Department of Clinical Laboratory, Henan Provincial People' s Hospital, Zhengzhou University People' s Hospital, Henan University People' s Hospital, Zhengzhou; 450003, China
| | - Yushe Wang
- Department of Neurosurgery, Henan Provincial People' s Hospital, Zhengzhou University People' s Hospital, Henan University People' s Hospital, Zhengzhou, China
| | - Yong Wang
- Department of Neurosurgery, Henan Provincial People' s Hospital, Zhengzhou University People' s Hospital, Henan University People' s Hospital, Zhengzhou, China
| |
Collapse
|
18
|
Wang Q, Wu Y, Lin M, Wang G, Liu J, Xie M, Zheng B, Shen C, Shen J. BMI1 promotes osteosarcoma proliferation and metastasis by repressing the transcription of SIK1. Cancer Cell Int 2022; 22:136. [PMID: 35346195 PMCID: PMC8961961 DOI: 10.1186/s12935-022-02552-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 03/14/2022] [Indexed: 01/09/2023] Open
Abstract
Abstract
Background
Osteosarcoma (OS) is the most common malignant tumor of bone, and the clinical efficacy of current treatments and associated survival rates need to be further improved by employing novel therapeutic strategies. Although various studies have shown that BMI1 protein is universally upregulated in OS cells and tissues, its specific role and underlying mechanism have not yet been fully explored.
Methods
Expression of BMI1 protein in OS cells was detected by western blot. The effect of BMI1 on proliferation and migration of OS cells (143B and U-2OS cell lines) was investigated in vitro using CCK-8, colony formation and transwell assays, and in vivo using subcutaneous tumorigenesis and lung metastasis assays in xenograft nude mice. Expression of epithelial–mesenchymal transition (EMT)-associated proteins was detected by immunofluorescence imaging. Bioinformatic analysis was performed using ENCODE databases to predict downstream targets of BMI1. SIK1 mRNA expression in osteosarcoma cells was detected by quantitative real-time reverse transcription PCR (qPCR). Chromatin immunoprecipitation-qPCR (ChIP-qPCR) was used to investigate expression of BMI1-associated, RING1B-associated, H2AK119ub-associated and H3K4me3-associated DNA at the putative binding region of BMI1 on the SIK1 promoter in OS cells.
Results
Using both in vitro and in vivo experimental approaches, we found that BMI1 promotes OS cell proliferation and metastasis. The tumor suppressor SIK1 was identified as the direct target gene of BMI1 in OS cells. In vitro experiments demonstrated that SIK1 could inhibit proliferation and migration of OS cells. Inhibition of SIK1 largely rescued the altered phenotypes of BMI1-deficient OS cells. Mechanistically, we demonstrated that BMI1 directly binds to the promoter region of SIK1 in a complex with RING1B to promote monoubiquitination of histone H2A at lysine 119 (H2AK119ub) and inhibit H3K4 trimethylation (H3K4me3), resulting in inhibition of SIK1 transcription. We therefore suggest that BMI1 promotes OS cell proliferation and metastasis by inhibiting SIK1.
Conclusions
Our results reveal a novel molecular mechanism of OS development promoted by BMI1 and provides a new potential target for OS treatment.
Collapse
|
19
|
Huang H, Zhong P, Zhang J, Chen X, Chen J, Lin T, Wu Q. Human umbilical cord-mesenchymal stem cells-derived exosomes carrying microRNA-15a-5p possess therapeutic effects on Wilms tumor via regulating septin 2. Bioengineered 2022; 13:6136-6149. [PMID: 35200105 PMCID: PMC8973990 DOI: 10.1080/21655979.2022.2037379] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The exact mechanism of miR-15a-5p shuttled by human umbilical cord-mesenchymal stem cell-derived exosomes (hUC-MSCs-Exo) in Wilms tumor (WT) was estimated. WT tissues were collected clinically. miR-15a-5p and septin 2 (SEPT2) expression levels were examined in tissues . hUC-MSCs-Exo were transfected with miR-15a-5p-related oligonucleotides and co-cultured with WT cells (G-401). In addition, SEPT2 loss-of-function was performed in G-401 cells. The biological functions of G-401 cells after treatments were evaluated. Moreover, tumor formation tests further assessed the role of exosomal miR-15a-5p in WT. The miR-15a-5p level was lower and the SEPT2 level was higher in WT. hUC-MSCs-Exo impaired the biological functions of G-401 cells. hUC-MSCs-Exo carried upregulated miR-15a-5p into G-401 cells, thereby lessening the tumorigenic properties of G-401 cells. Inhibition of SEPT2 suppressed the biological function of WT cells and upregulated SEPT2 reversed hUC-MSCs-Exo-mediated inhibition of G-401 cell growth. The tumorigenicity of G-401 cells in mice was impaired by hUC-MSCs-Exo overexpressing miR-15a-5p. The data prove that miR-15a-5p shuttled by hUC-MSCs-Exo negatively regulates SEPT2 expression, and disrupts WT cell growth in vivo and in vitro.
Collapse
Affiliation(s)
- He Huang
- Department of Pediatric Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou City, Fujian Province, China
| | - Peilin Zhong
- Department of Gynecological, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou City, Fujian Province, China
| | - Jianxing Zhang
- Department of Pediatric Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou City, Fujian Province, China
| | - Xinghe Chen
- Department of Pediatric Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou City, Fujian Province, China
| | - Jinwen Chen
- Department of Pediatric Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou City, Fujian Province, China
| | - Tian Lin
- Department of Pediatric Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou City, Fujian Province, China
| | - Qiang Wu
- Department of Pediatric Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou City, Fujian Province, China
| |
Collapse
|
20
|
Peng J, Liang Q, Xu Z, Cai Y, Peng B, Li J, Zhang W, Kang F, Hong Q, Yan Y, Zhang M. Current Understanding of Exosomal MicroRNAs in Glioma Immune Regulation and Therapeutic Responses. Front Immunol 2022; 12:813747. [PMID: 35095909 PMCID: PMC8796999 DOI: 10.3389/fimmu.2021.813747] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 12/27/2021] [Indexed: 02/05/2023] Open
Abstract
Exosomes, the small extracellular vesicles, are released by multiple cell types, including tumor cells, and represent a novel avenue for intercellular communication via transferring diverse biomolecules. Recently, microRNAs (miRNAs) were demonstrated to be enclosed in exosomes and therefore was protected from degradation. Such exosomal miRNAs can be transmitted to recipient cells where they could regulate multiple cancer-associated biological processes. Accumulative evidence suggests that exosomal miRNAs serve essential roles in modifying the glioma immune microenvironment and potentially affecting the malignant behaviors and therapeutic responses. As exosomal miRNAs are detectable in almost all kinds of biofluids and correlated with clinicopathological characteristics of glioma, they might be served as promising biomarkers for gliomas. We reviewed the novel findings regarding the biological functions of exosomal miRNAs during glioma pathogenesis and immune regulation. Furthermore, we elaborated on their potential clinical applications as biomarkers in glioma diagnosis, prognosis and treatment response prediction. Finally, we summarized the accessible databases that can be employed for exosome-associated miRNAs identification and functional exploration of cancers, including glioma.
Collapse
Affiliation(s)
- Jinwu Peng
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China.,Department of Pathology, Xiangya Changde Hospital, Changde, China
| | - Qiuju Liang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
| | - Zhijie Xu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China.,Department of Pathology, Xiangya Changde Hospital, Changde, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yuan Cai
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Bi Peng
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Jianbo Li
- Department of Pathology, Xiangya Changde Hospital, Changde, China
| | - Wenqin Zhang
- Department of Pathology, Xiangya Changde Hospital, Changde, China
| | - Fanhua Kang
- Department of Pathology, Xiangya Changde Hospital, Changde, China
| | - Qianhui Hong
- Department of Pathology, Xiangya Changde Hospital, Changde, China
| | - Yuanliang Yan
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Mingyu Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
21
|
Wang Y, Yang L, Fan C, Mu H, Han M, Liu T, Xie L, Gao Q. miR-130b Expression Level Changes Promote Cervical Cancer Cell Proliferation But Inhibit its Apoptosis by Targeting CDKN1A Gene. Curr Cancer Drug Targets 2022; 22:153-168. [PMID: 35016595 PMCID: PMC9413419 DOI: 10.2174/1568009622666220111090715] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 10/21/2021] [Accepted: 11/12/2021] [Indexed: 12/05/2022]
Abstract
Background:
Dysregulation of miR-130b expression is associated with the development of different cancers. However, the description of the biological roles of miR-130b in the growth and survival of cervical cancer cells is limited. Methods:
The miR-130b levels in cervical cancer cells during different stages of growth were determined using reverse transcription-quantitative PCR. The methylation level of DNA sequences upstream of the miR-130b gene was measured using an SYBR Green-based quantitative methylation-specific PCR. Reverse transcription-quantitative PCR, Western blotting, and fluorescence report assays were used to identify the miR-130b-targeted gene. Cell counting kit-8 and comet assays were used to determine cell viability and DNA damage levels in cells, respectively. EdU Apopllo488 in vitro Flow Cytometry kit, propidium iodide staining, anti-γ-H2AX antibody staining, and Annexin-V apoptosis kit were subsequently used to determine DNA synthesis rates, cell cycle distribution, count of DNA double-strand breaks, and levels of apoptotic cells. Results:
miR-130b levels increased at exponential phases of the growth of cervical cancer cells but reduced at stationary phases. The methylation of a prominent CpG island near the transcript start site suppressed the miR-130b gene expression. MiR-130b increased cell viability, promoted both DNA synthesis and G1 to S phase transition of the cells at exponential phases, but reduced cell viability accompanied by accumulations of DNA breaks and augmentations in apoptosis rates of the cells in stationary phases by targeting cyclin-dependent kinase inhibitor 1A mRNA. Conclusion:
miR-130b promoted the growth of cervical cancer cells during the exponential phase, whereas it impaired the survival of cells during stationary phases.
Collapse
Affiliation(s)
- Yanli Wang
- Department of Clinical Laboratory, Tianjin Hospital of ITCWM Nankai Hospital, Tianjin, China
| | - Lei Yang
- Department of Clinical Laboratory, Tianjin Hospital of ITCWM Nankai Hospital, Tianjin, China
| | - Caihong Fan
- The First Central Clinical College of Tianjin Medical University, Tianjin Medical University, Tianjin, China
| | - Hong Mu
- Department of Clinical Laboratory, Tianjin First Center Hospital, Tianjin, China
| | - Munan Han
- First Clinical Medical College, Nanjing Medical University, Nanjing, China
| | - Tao Liu
- a; eKey Laboratory for Critical Care Medicine of the Ministry of Health, Tianjin, China
| | - Lili Xie
- The First Central Clinical College of Tianjin Medical University, Tianjin Medical University, Tianjin, China
| | - Qiang Gao
- Department of Clinical Laboratory, Tianjin First Center Hospital, Tianjin, China
| |
Collapse
|
22
|
Xue P, Huang S, Han X, Zhang C, Yang L, Xiao W, Fu J, Li H, Zhou Y. Exosomal miR-101-3p and miR-423-5p inhibit medulloblastoma tumorigenesis through targeting FOXP4 and EZH2. Cell Death Differ 2022; 29:82-95. [PMID: 34294888 PMCID: PMC8738741 DOI: 10.1038/s41418-021-00838-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 07/08/2021] [Accepted: 07/12/2021] [Indexed: 01/03/2023] Open
Abstract
Exosomal microRNAs (miRNAs) have been implicated in the development and progression of a variety of tumors; however, whether they contribute to medulloblastoma (MB) tumorigenesis remains to be elucidated. To address this, we first characterized the miRNA profiles of circulating exosomes by miRNA sequencing to identify miRNAs differentially expressed between children with MB and healthy controls. Then, we conducted in vitro and in vivo functional assays with the identified miRNAs and their predicted targets. We found that, compared with healthy controls, 35 miRNAs were upregulated and 5 downregulated in exosomes isolated from the plasma of MB patients. We further found that the expression of miR-101-3p and miR-423-5p was significantly higher in plasma exosomes from MB patients than in healthy controls in an expanded cohort and these exosomal miRNAs could be delivered to tumor cells via exosomes. An in vitro functional analysis of miR-101-3p and miR-423-5p showed that treating MB cells with the corresponding mimics significantly inhibited the proliferation, colony-forming ability, migratory ability, and invasive capacity of tumor cells, and promoted cell apoptosis. Additionally, miR-101-3p and miR-423-5p were found to act as tumor suppressors by directly targeting a common gene, FOXP4, which encodes a transcription factor with a vital role in embryonic development and tumorigenesis. Moreover, miR-101-3p also targeted EZH2, a histone methyltransferase, to reinforce its tumor inhibitory effects. Using a xenograft nude mouse model of MB, we further identified that the overexpression of miR-101-3p and miR-423-5p inhibited tumorigenesis in vivo. Our findings provide novel insights into the functions of exosomal miRNAs in mediating MB progression and suggest a potential therapeutic approach for the treatment of children with MB.
Collapse
Affiliation(s)
- Ping Xue
- Institute of Pediatrics, Children's Hospital of Fudan University, and the Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
- Department of Neurosurgery, Children's Hospital of Fudan University, Shanghai, China
| | - Saihua Huang
- Institute of Pediatrics, Children's Hospital of Fudan University, and the Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
- National Health Commission (NHC) Key Laboratory of Neonatal Diseases (Fudan University), Shanghai, 201102, China
| | - Xiao Han
- Institute of Pediatrics, Children's Hospital of Fudan University, and the Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
- National Health Commission (NHC) Key Laboratory of Neonatal Diseases (Fudan University), Shanghai, 201102, China
| | - Caiyan Zhang
- Institute of Pediatrics, Children's Hospital of Fudan University, and the Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Lan Yang
- Institute of Pediatrics, Children's Hospital of Fudan University, and the Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Wenfeng Xiao
- Institute of Pediatrics, Children's Hospital of Fudan University, and the Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Jinrong Fu
- General Department, Children's Hospital of Fudan University, Shanghai, China
| | - Hao Li
- Department of Neurosurgery, Children's Hospital of Fudan University, Shanghai, China.
| | - Yufeng Zhou
- Institute of Pediatrics, Children's Hospital of Fudan University, and the Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China.
- National Health Commission (NHC) Key Laboratory of Neonatal Diseases (Fudan University), Shanghai, 201102, China.
| |
Collapse
|
23
|
EZH2 as a new therapeutic target in brain tumors: Molecular landscape, therapeutic targeting and future prospects. Biomed Pharmacother 2021; 146:112532. [PMID: 34906772 DOI: 10.1016/j.biopha.2021.112532] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 12/02/2021] [Accepted: 12/08/2021] [Indexed: 12/20/2022] Open
Abstract
Brain tumors are responsible for high mortality and morbidity worldwide. The brain tumor treatment depends on identification of molecular pathways involved in progression and malignancy. Enhancer of zeste homolog 2 (EZH2) has obtained much attention in recent years in field of cancer therapy due to its aberrant expression and capacity in modulating expression of genes by binding to their promoter and affecting methylation status. The present review focuses on EZH2 signaling in brain tumors including glioma, glioblastoma, astrocytoma, ependymomas, medulloblastoma and brain rhabdoid tumors. EZH2 signaling mainly participates in increasing proliferation and invasion of cancer cells. However, in medulloblastoma, EZH2 demonstrates tumor-suppressor activity. Furthermore, EZH2 can regulate response of brain tumors to chemotherapy and radiotherapy. Various molecular pathways can function as upstream mediators of EZH2 in brain tumors including lncRNAs and miRNAs. Owing to its enzymatic activity, EZH2 can bind to promoter of target genes to induce methylation and affects their expression. EZH2 can be considered as an independent prognostic factor in brain tumors that its upregulation provides undesirable prognosis. Both anti-tumor agents and gene therapies such as siRNA have been developed for targeting EZH2 in cancer therapy.
Collapse
|
24
|
Wu J, Zhang F, Zhang J, Sun Z, Hao C, Cao H, Wang W. A Novel miRNA-Based Model Can Predict the Prognosis of Clear Cell Renal Cell Carcinoma. Technol Cancer Res Treat 2021; 20:15330338211027923. [PMID: 34159861 PMCID: PMC8237220 DOI: 10.1177/15330338211027923] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is one of the most prevalent renal malignant cancer, whose survival rate and quality of life of patients are still not satisfactory. Nevertheless, the TNM staging system currently used in clinical cannot make accurate survival predictions and precise treatment decisions for ccRCC patients. Therefore, there is an urgent need for more reliable biomarkers to identify high-risk subgroups of ccRCC patients to guide timely intervention and treatment. Recently, MiRNAs have been shown to be closely related to the procession of a variety of tumors, and they have high stability in various tissues, which makes them suggested to have the potential as a prognostic biomarker of ccRCC. In this study, by analyzing and processing the miRNAs expression profile of ccRCC patients from the TCGA database, we finally constructed an excellent miRNAs signature and verified it through a variety of methods. In order to build a more accurate and reliable clinical predictive model, we integrated the miRNAs signature with other prognostic-related clinical parameters to construct a nomogram. Functional enrichment analysis showed that miRNAs in the signature may regulate the genes involved in the Hippo signaling pathway, Tight junction, and Wnt signaling pathway to cause different prognoses of ccRCC patients, which may provide a reference for subsequent basic research and targeted therapy. To conclude, our study constructed a useful miRNAs signature, which allows the prognosis stratification for ccRCC patients and thereby guides the timely and effective interventions on high-risk patients. At the same time, this study also found the potential biological pathways involved in the procession of ccRCC.
Collapse
Affiliation(s)
- Jiyue Wu
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China.,Institute of Urology, Capital Medical University, Beijing, China
| | - Feilong Zhang
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China.,Institute of Urology, Capital Medical University, Beijing, China
| | - Jiandong Zhang
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China.,Institute of Urology, Capital Medical University, Beijing, China
| | - Zejia Sun
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China.,Institute of Urology, Capital Medical University, Beijing, China
| | - Changzhen Hao
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China.,Institute of Urology, Capital Medical University, Beijing, China
| | - Huawei Cao
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China.,Institute of Urology, Capital Medical University, Beijing, China
| | - Wei Wang
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China.,Institute of Urology, Capital Medical University, Beijing, China
| |
Collapse
|
25
|
TSCCA: A tensor sparse CCA method for detecting microRNA-gene patterns from multiple cancers. PLoS Comput Biol 2021; 17:e1009044. [PMID: 34061840 PMCID: PMC8195367 DOI: 10.1371/journal.pcbi.1009044] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 06/11/2021] [Accepted: 05/05/2021] [Indexed: 12/22/2022] Open
Abstract
Existing studies have demonstrated that dysregulation of microRNAs (miRNAs or miRs) is involved in the initiation and progression of cancer. Many efforts have been devoted to identify microRNAs as potential biomarkers for cancer diagnosis, prognosis and therapeutic targets. With the rapid development of miRNA sequencing technology, a vast amount of miRNA expression data for multiple cancers has been collected. These invaluable data repositories provide new paradigms to explore the relationship between miRNAs and cancer. Thus, there is an urgent need to explore the complex cancer-related miRNA-gene patterns by integrating multi-omics data in a pan-cancer paradigm. In this study, we present a tensor sparse canonical correlation analysis (TSCCA) method for identifying cancer-related miRNA-gene modules across multiple cancers. TSCCA is able to overcome the drawbacks of existing solutions and capture both the cancer-shared and specific miRNA-gene co-expressed modules with better biological interpretations. We comprehensively evaluate the performance of TSCCA using a set of simulated data and matched miRNA/gene expression data across 33 cancer types from the TCGA database. We uncover several dysfunctional miRNA-gene modules with important biological functions and statistical significance. These modules can advance our understanding of miRNA regulatory mechanisms of cancer and provide insights into miRNA-based treatments for cancer. MicroRNAs (miRNAs) are a class of small non-coding RNAs. Previous studies have revealed that miRNA-gene regulatory modules play key roles in the occurrence and development of cancer. However, little has been done to discover miRNA-gene regulatory modules from a pan-cancer view. Thus, it is urgently needed to develop new methods to explore the complex cancer-related miRNA-gene patterns by integrating multi-omics data of multi-cancers. To build the connections between miRNA-gene regulatory modules across different cancer types, we propose a tensor sparse canonical correlation analysis (TSCCA) method. Our specific contributions are two-fold: (1) We propose a sparse statistical learning model TSCCA and an efficient block-coordinate descent algorithm to solve it. (2) We apply TSCCA to a multi-omics data set of 33 cancer types from TCGA and identify some cancer-related miRNA-gene modules with important biological functions and statistical significance.
Collapse
|
26
|
Elbadawy M, Sato Y, Mori T, Goto Y, Hayashi K, Yamanaka M, Azakami D, Uchide T, Fukushima R, Yoshida T, Shibutani M, Kobayashi M, Shinohara Y, Abugomaa A, Kaneda M, Yamawaki H, Usui T, Sasaki K. Anti-tumor effect of trametinib in bladder cancer organoid and the underlying mechanism. Cancer Biol Ther 2021; 22:357-371. [PMID: 34034619 DOI: 10.1080/15384047.2021.1919004] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Bladder cancer (BC), a main neoplasm of urinary tract, is usually inoperable and unresponsive to chemotherapy. As a novel experimental model for muscle-invasive BC, we previously established a culture method of dog BC organoids. In the present study, the detailed in vitro and in vivo anti-tumor effects of trametinib were investigated by using this model. In each BC organoid strain, epidermal growth factor receptor (EGFR)/ERK signaling was upregulated compared with normal bladder cells. Trametinib even at a low concentration inhibited the cell viability of BC organoids and the activation of ERK through decreasing expression of c-Myc, ELK1, SIK1, and PLA2G4A. Trametinib arrested cell cycle of BC with few apoptosis. Dual treatment of BC organoids with trametinib and YAP inhibitor, verteporfin extremely inhibited the cell viability with apoptosis induction. Moreover, trametinib induced basal to luminal differentiation of BC organoids by upregulating luminal markers and downregulating basal ones. In vivo, trametinib decreased the tumor growth of BC organoids in mice and the xenograft-derived organoids from trametinib-administered mice showed enhanced sensitivity to carboplatin due to MSH2 upregulation. Our data suggested a new strategy of trametinib-YAP inhibitor or trametinib-carboplatin combination as a promising treatment of BC.
Collapse
Affiliation(s)
- Mohamed Elbadawy
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Japan.,Department of Pharmacology, Faculty of Veterinary Medicine, Benha University, Toukh, Egypt
| | - Yomogi Sato
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Japan
| | - Takashi Mori
- Laboratory of Veterinary Clinical Oncology, Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan.,Center for Highly Advanced Integration of Nano and Life Sciences, Gifu University (G-CHAIN), Gifu, Japan
| | - Yuta Goto
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Japan
| | - Kimika Hayashi
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Japan
| | - Megumi Yamanaka
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Japan
| | - Daigo Azakami
- Department of Veterinary Clinical Oncology, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Japan
| | - Tsuyoshi Uchide
- Department of Veterinary Surgery, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Japan
| | - Ryuji Fukushima
- Animal Medical Center, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Japan
| | - Toshinori Yoshida
- Laboratory of Veterinary Pathology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Japan
| | - Makoto Shibutani
- Laboratory of Veterinary Pathology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Japan
| | - Mio Kobayashi
- Laboratory of Veterinary Pathology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Japan
| | - Yuta Shinohara
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Japan.,Pet Health & Food Division, Iskara Industry CO., LTD, Chuo-ku, Japan
| | - Amira Abugomaa
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Japan.,Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Masahiro Kaneda
- Laboratory of Veterinary Anatomy, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Japan
| | - Hideyuki Yamawaki
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, Towada, Japan
| | - Tatsuya Usui
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Japan
| | - Kazuaki Sasaki
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Japan
| |
Collapse
|
27
|
Bao D, Li M, Zhou D, Zhuang C, Ge Z, Wei Q, Zhang L. miR-130b-3p is high-expressed in polycystic ovarian syndrome and promotes granulosa cell proliferation by targeting SMAD4. J Steroid Biochem Mol Biol 2021; 209:105844. [PMID: 33582305 DOI: 10.1016/j.jsbmb.2021.105844] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 02/04/2021] [Accepted: 02/09/2021] [Indexed: 01/01/2023]
Abstract
BACKGROUND Being one of the most prevalent metabolic and endocrine disorders, Polycystic Ovary Syndrome (PCOS) has been proven to be associated with microRNA-130b-3p (miR-130b-3p). However, the exact role played by miR-130b-3p in the pathogenesis and progression of PCOS remains unknown. Thus, this article is focused on elucidating the function of miR-130b-3p in the pathogenesis of PCOS. METHODS The expression levels of miR-130b-3p and SMAD4 in tissues and cells responsible for the development of PCOS were determined by RT-qPCR and western blot. A miR-130b-3p mimic/inhibitor or si-SMAD4 were transfected into KGN cells. The cell viability was detected by CCK-8 and EDU methods. The activity of caspase-3 was measured by caspase-3 analysis. Subsequently, apoptosis and the cell cycle were measured via flow cytometry. The correlation between SMAD4 and miR-130b-3p was confirmed using an RNA pull-down assay and a dual luciferase reporter system assay. RESULTS MiR-130b-3p was upregulated in the KGN cells and ovarian granulosa cells (GCs) of PCOS patients. It was found that miR-130b-3p overexpression or SMAD4 silencing can promote KGN cell proliferation and positive EDU rates, induce S phase arrest, inhibit apoptosis and caspase-3 activity. On the other hand, miR-130b-3p inhibitors reduce KGN cell proliferation, inhibit apoptosis and reverse the effect of si-SMAD4. CONCLUSION MiR-130b-3p directly interacts with SMAD4 to induce KGN cell proliferation, inhibit apoptosis, suggesting that miR-130b-3p expression is positively correlated with the development of PCOS. This may serve as new evidence for the abnormal proliferation of GCs in PCOS.
Collapse
Affiliation(s)
- Dongqin Bao
- Center for Reproductive Medicine, The Affiliated Shuyang Hospital of Xuzhou Medical University, Suqian City, Jiangsu Province, China.
| | - Mingan Li
- Center for Reproductive Medicine, The Affiliated Shuyang Hospital of Xuzhou Medical University, Suqian City, Jiangsu Province, China
| | - Dongxia Zhou
- Center for Reproductive Medicine, The Affiliated Shuyang Hospital of Xuzhou Medical University, Suqian City, Jiangsu Province, China
| | - Chaohui Zhuang
- Center for Reproductive Medicine, The Affiliated Shuyang Hospital of Xuzhou Medical University, Suqian City, Jiangsu Province, China
| | - Zhijuan Ge
- Center for Reproductive Medicine, The Affiliated Shuyang Hospital of Xuzhou Medical University, Suqian City, Jiangsu Province, China
| | - Qian Wei
- Center for Reproductive Medicine, The Affiliated Shuyang Hospital of Xuzhou Medical University, Suqian City, Jiangsu Province, China
| | - Limin Zhang
- Center for Reproductive Medicine, The Affiliated Shuyang Hospital of Xuzhou Medical University, Suqian City, Jiangsu Province, China
| |
Collapse
|
28
|
Regulation of Nuclear Factor-KappaB (NF-κB) signaling pathway by non-coding RNAs in cancer: Inhibiting or promoting carcinogenesis? Cancer Lett 2021; 509:63-80. [PMID: 33838282 DOI: 10.1016/j.canlet.2021.03.025] [Citation(s) in RCA: 167] [Impact Index Per Article: 55.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 02/18/2021] [Accepted: 03/24/2021] [Indexed: 12/12/2022]
Abstract
The nuclear factor-kappaB (NF-κB) signaling pathway is considered as a potential therapeutic target in cancer therapy. It has been well established that transcription factor NF-κB is involved in regulating physiological and pathological events including inflammation, immune response and differentiation. Increasing evidences suggest that deregulated NF-κB signaling can enhance cancer cell proliferation, metastasis and also mediate radio-as well as chemo-resistance. On the contrary, non-coding RNAs (ncRNAs) have been found to modulate NF-κB signaling pathway under different settings. MicroRNAs (miRNAs) can dually inhibit/induce NF-κB signaling thereby affecting the growth and migration of cancer cells. Furthermore, the response of cancer cells to radiotherapy and chemotherapy may also be regulated by miRNAs. Regulation of NF-κB by miRNAs may be mediated via binding to 3/-UTR region. Interestingly, anti-tumor compounds can increase the expression of tumor-suppressor miRNAs in inhibiting NF-κB activation and the progression of cancers. Long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs) can also effectively modulate NF-κB signaling thus affecting tumorigenesis. It is noteworthy that several studies have demonstrated that lncRNAs and circRNAs can affect miRNAs in targeting NF-κB activation. They can act as competing endogenous RNA (ceRNA) thereby reducing miRNA expression to induce NF-κB activation that can in turn promote cancer progression and malignancy.
Collapse
|