1
|
Wang Z, Wang R, Na Z, Liang S, Wu F, Xie H, Zhang X, Xu W, Wang X. Network Pharmacology Analysis of Liquid-Cultured Armillaria ostoyae Mycelial Metabolites and Their Molecular Mechanism of Action against Gastric Cancer. Molecules 2024; 29:1668. [PMID: 38611946 PMCID: PMC11013622 DOI: 10.3390/molecules29071668] [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: 03/01/2024] [Revised: 03/25/2024] [Accepted: 04/05/2024] [Indexed: 04/14/2024] Open
Abstract
Armillaria sp. are traditional edible medicinal mushrooms with various health functions; however, the relationship between their composition and efficacy has not yet been determined. Here, the ethanol extract of liquid-cultured Armillaria ostoyae mycelia (AOME), a pure wild Armillaria sp. strain, was analyzed using UHPLC-QTOF/MS, network pharmacology, and molecular docking techniques. The obtained extract affects various metabolic pathways, such as JAK/STAT and PI3K/AKT. The extract also contains important compounds such as 4-(dimethylamino)-N-[7-(hydroxyamino)-7-oxoheptyl] benzamide, isoliquiritigenin, and 7-hydroxycoumarin. Moreover, the extract targets key proteins, including EGFR, SCR, and IL6, to suppress the progression of gastric cancer, thereby synergistically inhibiting cancer development. The molecular docking analyses indicated that the main compounds stably bind to the target proteins. The final cell culture experimental data showed that the ethanol extract inhibited MGC-803 gastric cancer cells. In summary, our research revealed the beneficial components of AOME for treating gastric cancer and its associated molecular pathways. However, further research is needed to confirm its effectiveness and safety in gastric cancer patients.
Collapse
Affiliation(s)
- Zhishuo Wang
- School of Food Engineering, Harbin University of Commerce, Harbin 150028, China
- Key Laboratory for Food Science and Engineering, Harbin University of Commerce, Harbin 150028, China
| | - Ruiqi Wang
- School of Food Engineering, Harbin University of Commerce, Harbin 150028, China
- Key Laboratory for Food Science and Engineering, Harbin University of Commerce, Harbin 150028, China
| | - Zhiguo Na
- School of Food Engineering, Harbin University of Commerce, Harbin 150028, China
- Key Laboratory for Food Science and Engineering, Harbin University of Commerce, Harbin 150028, China
| | - Shanshan Liang
- School of Food Engineering, Harbin University of Commerce, Harbin 150028, China
- Key Laboratory for Food Science and Engineering, Harbin University of Commerce, Harbin 150028, China
| | - Fan Wu
- School of Food Engineering, Harbin University of Commerce, Harbin 150028, China
- Key Laboratory for Food Science and Engineering, Harbin University of Commerce, Harbin 150028, China
| | - Hongyao Xie
- School of Food Engineering, Harbin University of Commerce, Harbin 150028, China
- Key Laboratory for Food Science and Engineering, Harbin University of Commerce, Harbin 150028, China
| | - Xue Zhang
- School of Food Engineering, Harbin University of Commerce, Harbin 150028, China
- Key Laboratory for Food Science and Engineering, Harbin University of Commerce, Harbin 150028, China
| | - Wei Xu
- School of Food Engineering, Harbin University of Commerce, Harbin 150028, China
- Key Laboratory for Food Science and Engineering, Harbin University of Commerce, Harbin 150028, China
| | - Xin Wang
- School of Food Engineering, Harbin University of Commerce, Harbin 150028, China
- Key Laboratory for Food Science and Engineering, Harbin University of Commerce, Harbin 150028, China
| |
Collapse
|
2
|
Tian J, Cheng L, Kong E, Gu W, Jiang Y, Hao Q, Kong B, Sun L. linc00958/miR-185-5p/RSF-1 modulates cisplatin resistance and angiogenesis through AKT1/GSK3β/VEGFA pathway in cervical cancer. Reprod Biol Endocrinol 2022; 20:132. [PMID: 36056431 PMCID: PMC9438131 DOI: 10.1186/s12958-022-00995-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 07/26/2022] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Chemoresistance is one of the major obstacles that lead to poor prognosis in cervical cancer. linc00958 was reported to be an oncogene in cervical cancer. However, its role in mediating chemoresistance remains to be revealed. PURPOSE To explore the regulatory mechanisms of linc00958 in cisplatin-resistant cervical cancer cells and further validate in xenograft mice. METHODS Online bioinformatic tools were used to conduct the pre-investigation of linc00958/miR-185-5p/RSF-1 and predict the associations between RSF-1 and AKT1/GSK3β/VEGFA in cervical cancer. RT-qPCR measured the RNA expression levels of linc00958/miR-185-5p/RSF-1 in SiHa and SiHa/DDP. Cell survival rates were evaluated by CCK8 methods after cells were exposed to differential concentrations of DDP. Dual-luciferase reporter methods were used to measure luciferase activity. Western blot measured RSF-1 protein and phosphorylated changes of AKT1/GSK3β. Immunofluorescence was employed to observe VEGFA secretion in vitro. Tube formation was applied to evaluate the in-vitro changes of angiogenesis. The SiHa/DDP cells stably transfected with pLKO-sh-NC or pLKO-sh-linc00958 plasmids, were injected into mice, establishing xenograft models. The changes in mice weight and tumor volumes were recorded. H&E staining and Immunohistochemistry (IHC) method was further performed. RESULTS linc00958 expression was higher in SiHa/DDP cells. High linc00958 expression was associated with low overall survival. In SiHa/DDP cells linc00958/miR-185-5p/RSF-1 axis inhibited the cellular resistance to cisplatin and suppressed VEGFA and the tube formation through AKT1/GSK3β/VEGFA pathway. The knockdown of linc00958 inhibited RSF-1 and Ki67, curbing tumor growth; it also inhibited VEGFA and CD34, decreasing angiogenesis in mice. CONCLUSION linc00958/miR-185-5p/RSF-1 modulates cisplatin resistance and angiogenesis through AKT1/GSK3β/VEGFA pathway in cervical cancer.
Collapse
Affiliation(s)
- Jing Tian
- grid.411918.40000 0004 1798 6427Department of Gynecological Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, People’s Republic of China
- grid.411918.40000 0004 1798 6427Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, TianjinTianjin, 300060 China
| | - Lei Cheng
- Department of Gynecology Oncology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, 266035 China
| | - Enqi Kong
- grid.410587.fShandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250021 China
| | - Wenjin Gu
- grid.415468.a0000 0004 1761 4893Department of Gynecological Oncology, Qingdao Central Hospital, The Second Affiliated Hospital of Medical College of Qingdao University, Qingdao, 266042 China
| | - Yuanyuan Jiang
- grid.415468.a0000 0004 1761 4893Department of Gynecological Oncology, Qingdao Central Hospital, The Second Affiliated Hospital of Medical College of Qingdao University, Qingdao, 266042 China
| | - Quan Hao
- grid.411918.40000 0004 1798 6427Department of Gynecological Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, People’s Republic of China
- grid.411918.40000 0004 1798 6427Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, TianjinTianjin, 300060 China
| | - Beihua Kong
- grid.27255.370000 0004 1761 1174Department of Obstetrics and Gynecology, Cheeloo College of Medicine, Shandong University, Qilu hospital, Jinan, 250012 China
| | - Li Sun
- grid.415468.a0000 0004 1761 4893Department of Gynecological Oncology, Qingdao Central Hospital, The Second Affiliated Hospital of Medical College of Qingdao University, Qingdao, 266042 China
| |
Collapse
|
3
|
Long Noncoding RNA Mediated Regulation in Human Embryogenesis, Pluripotency, and Reproduction. Stem Cells Int 2022; 2022:8051717. [PMID: 35103065 PMCID: PMC8800634 DOI: 10.1155/2022/8051717] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 12/27/2021] [Indexed: 12/12/2022] Open
Abstract
Long noncoding RNAs (lncRNAs), a class of noncoding RNAs with more than 200 bp in length, are produced by pervasive transcription in mammalian genomes and regulate gene expression through various action mechanisms. Accumulating data indicate that lncRNAs mediate essential biological functions in human development, including early embryogenesis, induction of pluripotency, and germ cell development. Comprehensive analysis of sequencing data highlights that lncRNAs are expressed in a stage-specific and human/primate-specific pattern during early human development. They contribute to cell fate determination through interacting with almost all classes of cellular biomolecules, including proteins, DNA, mRNAs, and microRNAs. Furthermore, the expression of a few of lncRNAs is highly associated with the pathogenesis and progression of many reproductive diseases, suggesting that they could serve as candidate biomarkers for diagnosis or novel targets for treatment. Here, we review research on lncRNAs and their roles in embryogenesis, pluripotency, and reproduction. We aim to identify the underlying molecular mechanisms essential for human development and provide novel insight into the causes and treatments of human reproductive diseases.
Collapse
|
4
|
Long non-coding RNA UCA1 enhances cervical cancer cell proliferation and invasion by regulating microRNA-299-3p expression. Oncol Lett 2021; 22:772. [PMID: 34589151 PMCID: PMC8442166 DOI: 10.3892/ol.2021.13033] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 08/16/2021] [Indexed: 12/24/2022] Open
Abstract
The long non-coding RNA, urothelial cancer-associated 1 (UCA1) is an important regulator in several tumors. However, to the best of our knowledge, the clinical roles of UCA1 in cervical cancer remain unclear. Thus, the present study aimed to investigate the function and mechanism of UCA1 in cervical cancer. Reverse transcription-quantitative PCR analysis was performed to detect UCA1 and microRNA (miR)-299-3p expression in cervical cancer tissues and cell lines. The Cell Counting Kit-8 and Transwell assays were performed to assess cell proliferation and invasion, respectively. Furthermore, the dual-luciferase reporter assay was performed to confirm the association between UCA1 and miR-299-3p. Rescue experiments were performed to determine the mechanism of the UCA1/miR-299-3p axis. The results demonstrated that UCA1 expression was upregulated in cervical cancer tissues and cell lines. Furthermore, overexpression of UCA1 enhanced the proliferation and invasion of cervical cancer cells, the effects of which were reversed following UCA1 knockdown. Notably, UCA1 interacted with miR-299-3p and negatively regulated miR-299-3p expression. In addition, miR-299-3p expression was downregulated in cervical cancer tissues and cell lines. Overexpression of miR-299-3p suppressed the proliferation and invasion of cervical cancer cells, reversing the effects of UCA1 knockdown on cervical cancer cell proliferation. Taken together, the results of the present study suggest that UCA1 promotes cell proliferation and invasion by regulating miR-299-3p expression in cervical cancer.
Collapse
|
5
|
Lamsisi M, Wakrim L, Bouziyane A, Benhessou M, Oudghiri M, Laraqui A, Elkarroumi M, Ennachit M, El Mzibri M, Ennaji MM. The Biological Significance of Long noncoding RNAs Dysregulation and their Mechanism of Regulating Signaling Pathways in Cervical Cancer. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2021; 10:75-101. [PMID: 34703793 PMCID: PMC8496250 DOI: 10.22088/ijmcm.bums.10.2.75] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 08/01/2021] [Indexed: 12/19/2022]
Abstract
Despite the remarkable decrease in cervical cancer incidence due to the availability of the HPV vaccine and implementation of screening programs for early detection in developed countries, this cancer remains a major health problem globally, especially in developing countries where most of the cases and mortality occur. Therefore, more understanding of molecular mechanisms of cervical cancer development might lead to the discovery of more effective diagnosis and treatment options. Research on long noncoding RNAs (lncRNAs) demonstrates the important roles of these molecules in many physiological processes and diseases, especially cancer. In the present review, we discussed the significance of lncRNAs altered expression in cervical cancer, highlighting their roles in regulating highly conserved signaling pathways, such as mitogen-activated protein kinase (MAPK), Wnt/β-catenin, Notch, and phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT) pathways and their association with the progression of cervical cancer in order to bring more insight and understanding of this disease and their potential implications in cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Maryame Lamsisi
- Team of Virology, Oncology and Medical Biotechnologies, Laboratory of Virology, Microbiology, Quality, and Biotechnologies/ ETB. Faculty of Science and Techniques Mohammedia, Hassan II University of Casablanca, Morocco.
| | - Lahcen Wakrim
- Laboratory of Virology, Pasteur Institute of Morocco. Casablanca, Morocco.
| | - Amal Bouziyane
- Team of Virology, Oncology and Medical Biotechnologies, Laboratory of Virology, Microbiology, Quality, and Biotechnologies/ ETB. Faculty of Science and Techniques Mohammedia, Hassan II University of Casablanca, Morocco.
- University Mohammed VI of Health Science, Casablanca, Morocco.
| | - Mustapha Benhessou
- Team of Virology, Oncology and Medical Biotechnologies, Laboratory of Virology, Microbiology, Quality, and Biotechnologies/ ETB. Faculty of Science and Techniques Mohammedia, Hassan II University of Casablanca, Morocco.
- School of Medicine and Pharmacy, University Hassan II of Casablanca, Morocco.
| | - Mounia Oudghiri
- Immunology and Biodiversity laboratory, Faculty of Sciences Ain chock, Hassan II University of Casablanca, Morocco.
| | - Abdelilah Laraqui
- Research and Biosafety Laboratory, Mohammed V Military Hospital, University Mohammed V of Rabat, Morocco.
| | - Mohamed Elkarroumi
- School of Medicine and Pharmacy, University Hassan II of Casablanca, Morocco.
| | - Mohammed Ennachit
- School of Medicine and Pharmacy, University Hassan II of Casablanca, Morocco.
| | | | - Moulay Mustapha Ennaji
- Corresponding author: Faculty of Science and Techniques Mohammedia, University Hassan II of Casablanca, Morocco. E-mail:
| |
Collapse
|
6
|
Pei XD, He ZL, Yao HL, Xiao JS, Li L, Gu JZ, Shi PZ, Wang JH, Jiang LH. 6-Shogaol from ginger shows anti-tumor effect in cervical carcinoma via PI3K/Akt/mTOR pathway. Eur J Nutr 2021; 60:2781-2793. [PMID: 33416981 DOI: 10.1007/s00394-020-02440-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 11/04/2020] [Indexed: 12/13/2022]
Abstract
PURPOSE 6-Shogaol, an active phenolic compound from ginger (Zingiber officinale), can inhibit the growth of a variety of human cancer cells. Nevertheless, its underlying molecular mechanisms in cervical cancer remain unclear. In this study, we systematically examine the inhibitory effect of 6-shogaol on cervical cancer in vitro and in vivo. METHODS Cell proliferation was assessed by CCK8 assay and colony formation assay in HeLa and SiHa cells. We analyzed cell cycle and apoptosis through flow cytometry. GFP-LC3 puncta and transmission electron microscopy were used to observe autophagic bodies. Wound-healing assay and transwell assay were used for evaluating the migration of cells. Western blot was applied to detect protein expression levels. RESULTS 6-Shogaol could suppress cell proliferation and migration, cause cell cycle arrest in the G2/M phase in HeLa and SiHa cells. Moreover, 6-shogaol triggered the apoptosis process through the mitochondrial pathway by downregulating the expression levels of p-PI3K, p-Akt and p-mTOR. Further research indicated that the induction of apoptosis by 6-shogaol was remarkably decreased after the treatment of ROS scavenger and PI3K agonist. Additionally, 6-shogaol increased the number of LC3-positive puncta and autophagic bodies per cell in both HeLa and SiHa cells. Pretreatment of cells with Bafilomycin A1, an autophagy inhibitor, accelerated 6-shogaol mediated cell apoptosis, suggesting that induction of autophagy by 6-shogaol is suppressive to apoptosis. Furthermore, in vivo data revealed that 6-shogaol significantly inhibited tumor growth and cell proliferation in tumor tissues. CONCLUSION These findings suggested that 6-shogaol could be developed as a functional food ingredient, which is potentially used as therapeutic agents for patients with cervical cancer.
Collapse
Affiliation(s)
- Xiao-Dong Pei
- College of Light Industry and Food Engineering, Guangxi University, Nanning, 530004, Guangxi, People's Republic of China
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, People's Republic of China
| | - Zhi-Long He
- College of Light Industry and Food Engineering, Guangxi University, Nanning, 530004, Guangxi, People's Republic of China
| | - Hong-Liang Yao
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangdong, 510260, People's Republic of China
| | - Jun-Song Xiao
- Beijing Higher Institution Engineering Research Center of Food Additives and Ingredients, Beijing Technology and Business University-BTBU, Beijing, 100048, People's Republic of China
| | - Lan Li
- School of Basic Medical Sciences, Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, People's Republic of China
| | - Jian-Zhong Gu
- School of Basic Medical Sciences, Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, People's Republic of China
| | - Pei-Zhao Shi
- School of Basic Medical Sciences, Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, People's Republic of China
| | - Jin-Hua Wang
- School of Basic Medical Sciences, Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, People's Republic of China
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Li-He Jiang
- College of Light Industry and Food Engineering, Guangxi University, Nanning, 530004, Guangxi, People's Republic of China.
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, People's Republic of China.
- School of Basic Medical Sciences, Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, People's Republic of China.
- Medical College, Guangxi University, Nanning, 530004, Guangxi, People's Republic of China.
| |
Collapse
|
7
|
Song H, Liu Y, Liang H, Jin X, Liu L. SPINT1-AS1 Drives Cervical Cancer Progression via Repressing miR-214 Biogenesis. Front Cell Dev Biol 2021; 9:691140. [PMID: 34350182 PMCID: PMC8326843 DOI: 10.3389/fcell.2021.691140] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 06/28/2021] [Indexed: 01/15/2023] Open
Abstract
Accumulating evidences have revealed the dysregulated expressions and critical roles of non-coding RNAs in various malignancies, including cervical cancer. Nevertheless, our knowledge about the vast majority of non-coding RNAs is still lacking. Here we identified long non-coding RNA (lncRNA) SPINT1-AS1 as a novel cervical cancer-associated lncRNA. SPINT1-AS1 was increased in cervical cancer and correlated with advanced stage and poor prognosis. SPINT1-AS1 was a direct downstream target of miR-214, a well-known tumor suppressive microRNA (miRNA) in cervical cancer. Intriguingly, SPINT1-AS1 was also found to repress miR-214 biogenesis via binding DNM3OS, the primary transcript of miR-214. The interaction between SPINT1-AS1 and DNM3OS repressed the binding of DROSHA and DGCR8 to DNM3OS, blocked DNM3OS cleavage, and therefore repressed mature miR-214 biogenesis. The expression of SPINT1-AS1 was significantly negatively correlated with miR-214 in cervical cancer tissues, supporting the reciprocal repression between SPINT1-AS1 and miR-214 in vivo. Through downregulating mature miR-214 level, SPINT1-AS1 upregulated the expression of β-catenin, a target of miR-214. Thus, SPINT1-AS1 further activated Wnt/β-catenin signaling in cervical cancer. Functionally, SPINT1-AS1 drove cervical cancer cellular proliferation, migration, and invasion in vitro, and also tumorigenesis in vivo. Deletion of the region mediating the interaction between SPINT1-AS1 and DNM3OS, overexpression of miR-214, and inhibition of Wnt/β-catenin signaling all reversed the roles of SPINT1-AS1 in cervical cancer. Collectively, these findings identified SPINT1-AS1 as a novel cervical cancer-associated oncogenic lncRNA which represses miR-214 biogenesis and activates Wnt/β-catenin signaling, highlighting its potential as prognostic biomarker and therapeutic target for cervical cancer.
Collapse
Affiliation(s)
- Hongjuan Song
- Department of Gynecology, Xuzhou Maternal and Child Health Care Hospital, Xuzhou, China.,Department of Gynecology, Xuzhou Renci Hospital, Xuzhou, China
| | - Yuan Liu
- Department of Gynecology, Xuzhou Maternal and Child Health Care Hospital, Xuzhou, China
| | - Hui Liang
- Department of Cervical Disease, Xuzhou Maternal and Child Health Care Hospital, Xuzhou, China
| | - Xin Jin
- Medical Department, Xuzhou Central Hospital, Xuzhou, China
| | - Liping Liu
- Department of Research and Development, Shanghai Lichun Biotechnology Co., Ltd., Shanghai, China
| |
Collapse
|
8
|
Xu J, Ma J, Guan B, Li J, Wang Y, Hu S. LncRNA HCP5 promotes malignant cell behaviors in esophageal squamous cell carcinoma via the PI3K/AKT/mTOR signaling. Cell Cycle 2021; 20:1374-1388. [PMID: 34190001 DOI: 10.1080/15384101.2021.1944512] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The role of lncRNA HCP5 in esophageal squamous cell carcinoma (ESCC) remains unknown despite its involvement in different malignancies. MTT assay, EdU assay, TUNEL assay, transwell assay, and sphere formation assay were conducted to reveal ESCC cell viability, proliferation, apoptosis, migration, invasion, and stemness characteristics. FISH and subcellular fraction assays were performed to reveal the subcellular location of HCP5 in ESCC cells. Luciferase reporter assay and RIP assay were conducted to explore the downstream axis of HCP5. Our findings revealed that HCP5 expression was at a higher level in ESCC tissues and cells compared to that in control tissues and cells. Additionally, HCP5 promoted ESCC cellular activities by promoting proliferation, migration, invasion ability and stemness characteristics of ESCC cells as well as suppressing cell apoptosis. Furthermore, we found that HCP5 bound with miR-139-5p to upregulate PDE4A via the competing endogenous RNA network in ESCC cells. Importantly, HCP5 was discovered to stimulate the PI3K/AKT/mTOR signaling by regulating the downstream target genes. Finally, rescue assays indicated that HCP5 promoted ESCC cell growth by activating the PDE4A-medaited PI3K/AKT/mTOR pathway. HCP5 promotes ESCC cellular development by modulating the miR-139-5p/PDE4A pathway and stimulating the PI3K/AKT/mTOR signaling pathway, which may be conducive for the improvement of ESCC treatment.
Collapse
Affiliation(s)
- Jianyu Xu
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Jianli Ma
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Bixi Guan
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Jian Li
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Yan Wang
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Songliu Hu
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| |
Collapse
|
9
|
Yang D, Ma X, Xu J, Jia K, Liu X, Zhang P. Zfx-induced upregulation of UBE2J1 facilitates endometrial cancer progression via PI3K/AKT pathway. Cancer Biol Ther 2021; 22:238-247. [PMID: 33632059 DOI: 10.1080/15384047.2021.1883186] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Emerging documents revealed that E2 enzyme family has been implicated in regulating the progression of numerous human cancers. Ubiquitin-conjugating enzyme E2 J1 (UBE2J1), a member of E2 enzyme family, has been reported to participate in the biological process of medulloblastoma, while little is known about its functionality in endometrial cancer (EC). Gene expression at the mRNA and protein levels were identified using RT-qPCR and western blot analysis, separately. The alteration on cell proliferation, adhesion, migration, invasion, and epithelial-mesenchymal transition (EMT) process was determined through 5-Ethynyl-2'-deoxyuridine, cell adhesion, wound healing and transwell assays as well as western blot analysis. The role of UBE2J1 in xenograft tumor in mice was determined. Luciferase reporter and chromatin immunoprecipitation assays were conducted to reveal the undering mechanism of UBE2J1. Our results indicated that UBE2J1 displayed high level in EC tissues and cells and predicted poor prognosis of EC patients. In addition, UBE2J1 depletion inhibited cell proliferation, adhesion, motion, EMT process invitro, and repressed tumor growth invivo. Rescue assays manifested that ethyl 2-amino-6-chloro-4-(1-cyano-2-ethoxy-2-oxoethyl)-4H-chromene-3-carboxylate treatment reversed the effects of UBE2J1 on PI3K/AKT pathway activation and malignant phenotypes of EC cells. Finally, zinc finger X-chromosomal protein (zfx), with high expression in EC tissues, was verified to activate UBE2J1 transcription by binding to UBE2J1 promoter. In conclusion, all facts signified that zfx-induced upregulation of UBE2J1 accelerated the progression of EC via regulating the PI3K/AKT signaling pathway, which suggested that UBE2J1 might be of great significance in probing into the underlying therapeutic strategies of EC.
Collapse
Affiliation(s)
- Dexin Yang
- Institute of Science and Technology for Brain-inspired Intelligence (ISTBI), Fudan University, Shanghai, China
| | - Xin Ma
- Department of Gynaecology and Obstetrics, Zhangjiagang First People's Hospital, Zhangjiagang, China
| | - Jie Xu
- Department of Gynaecology and Obstetrics, Zhangjiagang First People's Hospital, Zhangjiagang, China
| | - Ke Jia
- Department of Gynaecology and Obstetrics, Zhangjiagang First People's Hospital, Zhangjiagang, China
| | - Xiaoli Liu
- Department of Gynaecology and Obstetrics, Zhangjiagang First People's Hospital, Zhangjiagang, China
| | - Ping Zhang
- Department of Gynaecology and Obstetrics, Zhangjiagang First People's Hospital, Zhangjiagang, China
| |
Collapse
|
10
|
Liu H, Song M, Sun X, Zhang X, Miao H, Wang Y. T-box transcription factor TBX1, targeted by microRNA-6727-5p, inhibits cell growth and enhances cisplatin chemosensitivity of cervical cancer cells through AKT and MAPK pathways. Bioengineered 2021; 12:565-577. [PMID: 33557670 PMCID: PMC8806341 DOI: 10.1080/21655979.2021.1880732] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Cervical cancer (CC) is the fourth most common cancers among women worldwide. T-box transcription factor 1 (TBX1), a member of the T-box family, has anti-tumor effects in some types of cancer, but its role in CC is yet unknown. The aim of this study is to investigate the functions and underlying mechanisms of TBX1 in CC. Online database UALCAN showed that TBX1 was down-regulated in CC tissues compared with normal tissues and patients with lower TBX1 expression level had a poor prognosis. TBX1 overexpression significantly decreased the proliferation, migration, and invasion of Hela and SiHa cells. Conversely, cell apoptosis and chemosensitivity to cisplatin were promoted in TBX1-overexpressing CC cells. Moreover, up-regulation of TBX1 inhibited both AKT and MAPK signaling pathways. Furthermore, dual luciferase report assay indicated that TBX1 could directly bind to miR-6727-5p. In addition, TBX1 expression was inhibited by miR-6727-5p mimic and up-regulated by miR-6727-5p inhibitor. Knockdown of TBX1 reversed the inhibitory effect of the miR-6727-5p inhibitor on CC cells. This study demonstrates that TBX1, a target gene of miR-6727-5p, acts as a tumor suppressor in CC, indicating that TBX1 may be a new target for CC therapy.
Collapse
Affiliation(s)
- Haixia Liu
- Department of Gynecology, The Affiliated Hospital of Qingdao University , Qingdao, Shandong, P.R. China.,Department of Gynecology, The Third People's Hospital of Qingdao , Qingdao, Shandong, P.R. China
| | - Mei Song
- Department of Gynecology Oncology, The Affiliated Central Hospital of Qingdao University , Qingdao, Shandong, P.R. China
| | - Xiaoyan Sun
- Department of Gynecology, The Third People's Hospital of Qingdao , Qingdao, Shandong, P.R. China
| | - Xin Zhang
- Department of Gynecology, The Third People's Hospital of Qingdao , Qingdao, Shandong, P.R. China
| | - Huayan Miao
- Department of Gynecology, The Third People's Hospital of Qingdao , Qingdao, Shandong, P.R. China
| | - Yankui Wang
- Department of Gynecology, The Affiliated Hospital of Qingdao University , Qingdao, Shandong, P.R. China
| |
Collapse
|
11
|
The emerging role of non-coding RNAs in the regulation of PI3K/AKT pathway in the carcinogenesis process. Biomed Pharmacother 2021; 137:111279. [PMID: 33493969 DOI: 10.1016/j.biopha.2021.111279] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/07/2021] [Accepted: 01/12/2021] [Indexed: 02/07/2023] Open
Abstract
The PI3K/AKT pathway is an intracellular signaling pathway with an indispensable impact on cell cycle control. This pathway is functionally related with cell proliferation, cell survival, metabolism, and quiescence. The crucial role of this pathway in the development of cancer has offered this pathway as a target of novel anti-cancer treatments. Recent researches have demonstrated the role of microRNAs (miRNAs) and long noncoding RNAs (lncRNAs) in controlling the PI3K/AKT pathway. Some miRNAs such as miR-155-5p, miR-328-3p, miR-125b-5p, miR-126, miR-331-3p and miR-16 inactivate this pathway, while miR-182, miR-106a, miR-193, miR-214, miR-106b, miR-93, miR-21 and miR-103/107 enhance activity of this pathway. Expression levels of PI3K/AKT-associated miRNAs could be used to envisage the survival of cancer patients. Numerous lncRNAs such as GAS5, FER1L4, LINC00628, PICART1, LOC101928316, ADAMTS9-AS2, SLC25A5-AS1, MEG3, AB073614 and SNHG6 interplay with this pathway. Identification of the impact of miRNAs and lncRNAs in the control of the activity of PI3K/AKT pathway would enhance the efficacy of targeted therapies against this pathway. Moreover, each of the mentioned miRNAs and lncRNAs could be used as a putative therapeutic candidate for the interfering with the carcinogenesis. In the current study, we review the role of miRNAs and lncRNAs in controlling the PI3K/AKT pathway and their contribution to carcinogenesis.
Collapse
|
12
|
Razavi ZS, Tajiknia V, Majidi S, Ghandali M, Mirzaei HR, Rahimian N, Hamblin MR, Mirzaei H. Gynecologic cancers and non-coding RNAs: Epigenetic regulators with emerging roles. Crit Rev Oncol Hematol 2020; 157:103192. [PMID: 33290823 DOI: 10.1016/j.critrevonc.2020.103192] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 11/30/2020] [Accepted: 12/01/2020] [Indexed: 12/24/2022] Open
Abstract
Gynecologic cancers involve the female genital organs, such as the vulva, vagina, cervix, endometrium, ovaries, and fallopian tubes. The occurrence and frequency of gynecologic cancer depends on personal lifestyle, history of exposure to viruses or carcinogens, genetics, body shape, and geographical habitat. For a long time, research into the molecular biology of cancer was broadly restricted to protein-coding genes. Recently it has been realized that non-coding RNAs (ncRNA), including long noncoding RNAs (LncRNAs), microRNAs, circular RNAs and piRNAs (PIWI-interacting RNAs), can all play a role in the regulation of cellular function within gynecological cancer. It is now known that ncRNAs are able to play dual roles, i.e. can exert both oncogenic or tumor suppressive functions in gynecological cancer. Moreover, several clinical trials are underway looking at the biomarker and therapeutic roles of ncRNAs. These efforts may provide a new horizon for the diagnosis and treatment of gynecological cancer. Herein, we summarize some of the ncRNAs that have been shown to be important in gynecological cancers.
Collapse
Affiliation(s)
| | - Vida Tajiknia
- Department of Surgery, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Shahab Majidi
- Student Research Committee, Fasa University of Medical Sciences, Fasa, Iran
| | - Maryam Ghandali
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hamid Reza Mirzaei
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Neda Rahimian
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences (IUMS), Tehran, Iran.
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, 02114, USA; Department of Dermatology, Harvard Medical School, Boston, MA, 02115, USA; Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, 2028, South Africa.
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
13
|
Zhang X, Wang Y, Zhao A, Kong F, Jiang L, Wang J. Long Non-Coding RNA LINC00511 Accelerates Proliferation and Invasion in Cervical Cancer Through Targeting miR-324-5p/DRAM1 Axis. Onco Targets Ther 2020; 13:10245-10256. [PMID: 33116605 PMCID: PMC7567551 DOI: 10.2147/ott.s255067] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 08/29/2020] [Indexed: 12/16/2022] Open
Abstract
Purpose Cervical cancer is the second most prevalent female malignance, and human papillomavirus (HPV) infection is the main pathogenic factor of cervical cancer. Emerging evidence has revealed that a number of long non-coding RNAs (lncRNAs) play critical roles in the tumorigenesis and progression of cervical cancer. The aim of this study was to further investigate the precise role of lncRNA LINC00511 in HPV-negative and HPV-positive cervical cancer cells and explore the potential regulatory mechanism. Methods The expression of LINC00511 in cervical cancer and cell lines was examined by RT-PCR. Fluorescence in situ hybridization analysis (FISH) assay was performed to detect the localization of LINC00511 in cervical cancer cells. Loss-of-function experiments of LINC00511 by siRNA interference were performed to assess its effects on HPV-negative and HPV-positive cervical cancer cells. Dual-luciferase reporter and RNA immunoprecipitation (RIP) assays were used to identify the target of LINC00511. Relative expression of related proteins was detected using Western blot. Results Herein, the results showed that LINC00511 was significantly up-regulated in cervical cancer and cell lines and mainly distributed in the cytoplasm of cervical cancer cells. Loss-of-function experiments indicated that silencing of LINC00511 inhibited the proliferation and invasion of both HPV-negative and HPV-positive cervical cancer cells, as well as promoted apoptosis by regulating the Bcl-2/Bax axis and Caspase 3 activation. Bioinformatic analysis, dual-luciferase reporter, and RIP assays showed that LINC00511 was a target of miR-324-5p, while DRAM1 was a direct target of miR-324-5p. The expression of miR-324-5p was down-regulated in cervical cancer, while the expression of DRAM1 was up-regulated. Moreover, the expression of LINC00511 was negatively correlated with miR-324-5p expression in cervical cancer tissues and positively correlated with DRAM1. Further, DRAM1 overexpression promoted both HPV-negative and HPV-positive cervical cancer cell proliferation and invasion, which could be reversed by miR-324-5p mimics or si-LINC00511. Conclusion Collectively, these results suggest that LINC00511 functions as a competing endogenous RNA (ceRNA) to regulate the miR-324-5p/DRAM1 axis, leading to HPV-negative and HPV-positive cervical cancer aggravation.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Oncology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, People's Republic of China
| | - Yuyan Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, People's Republic of China
| | - Anqi Zhao
- Capital Medical University, Beijing 100069, People's Republic of China
| | - Fanshuang Kong
- Department of General Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, People's Republic of China
| | - Lipeng Jiang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, People's Republic of China
| | - Jinfeng Wang
- Department of Pediatrics, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, People's Republic of China
| |
Collapse
|