1
|
Abulsoud AI, Elshaer SS, Abdelmaksoud NM, Zaki MB, El-Mahdy HA, Ismail A, Al-Noshokaty TM, Fathi D, Abdel-Reheim MA, Mohammed OA, Doghish AS. Investigating the regulatory role of miRNAs as silent conductors in the management of pathogenesis and therapeutic resistance of pancreatic cancer. Pathol Res Pract 2023; 251:154855. [PMID: 37806169 DOI: 10.1016/j.prp.2023.154855] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 09/16/2023] [Accepted: 10/02/2023] [Indexed: 10/10/2023]
Abstract
Pancreatic cancer (PC) has the greatest mortality rate of all the main malignancies. Its advanced stage and poor prognosis place it at the bottom of all cancer sites. Hence, emerging biomarkers can enable precision medicine where PC therapy is tailored to each patient. This highlights the need for new, highly sensitive and specific biomarkers for early PC diagnosis. Prognostic indicators are also required to stratify PC patients. To avoid ineffective treatment, adverse events, and expenses, biomarkers are also required for patient monitoring and identifying responders to treatment. There is substantial evidence that microRNAs (miRs, miRNAs) play a critical role in regulating mRNA and, as a consequence, protein expression in normal and malignant tissues. Deregulated miRNA profiling in PC can help with diagnosis, treatment planning, and prognosis. Furthermore, knowledge of the primary effector genes and downstream pathways in PC can help pinpoint potential miRNAs for use in treatment. Different miRNA expression profiles may serve as diagnostic, prognostic markers, and therapeutic targets across the spectrum of malignant pancreatic illness. Dysregulation of miRNAs has been linked to the malignant pathophysiology of PC through affecting many cellular functions such as increasing invasive and proliferative prospect, supporting angiogenesis, cell cycle aberrance, apoptosis elusion, metastasis promotion, and low sensitivity to particular treatments. Accordingly, in the current review, we summarize the recent advances in the roles of oncogenic and tumor suppressor (TS) miRNAs in PC and discuss their potential as worthy diagnostic and prognostic biomarkers for PC, as well as their significance in PC pathogenesis and anticancer drug resistance.
Collapse
Affiliation(s)
- Ahmed I Abulsoud
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231 Cairo, Egypt; Department of Biochemistry and Biotechnology, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | - Shereen Saeid Elshaer
- Department of Biochemistry and Biotechnology, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt; Department of Biochemistry, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr city, Cairo 11823, Egypt
| | - Nourhan M Abdelmaksoud
- Department of Biochemistry and Biotechnology, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | - Mohamed Bakr Zaki
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Menoufia 32897, Egypt
| | - Hesham A El-Mahdy
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231 Cairo, Egypt.
| | - Ahmed Ismail
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231 Cairo, Egypt
| | - Tohada M Al-Noshokaty
- Department of Biochemistry and Biotechnology, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | - Doaa Fathi
- Department of Biochemistry and Biotechnology, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | - Mustafa Ahmed Abdel-Reheim
- Department of Pharmaceutical Sciences, College of Pharmacy, Shaqra University, Shaqra 11961, Saudi Arabia; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni Suef 62521, Egypt.
| | - Osama A Mohammed
- Department of Clinical Pharmacology, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia
| | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt; Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231 Cairo, Egypt.
| |
Collapse
|
2
|
Zhou J, Song G, Su M, Zhang H, Yang T, Song Z. Long noncoding RNA CASC9 promotes pancreatic cancer progression by acting as a ceRNA of miR-497-5p to upregulate expression of CCND1. ENVIRONMENTAL TOXICOLOGY 2023; 38:1251-1264. [PMID: 36947456 DOI: 10.1002/tox.23761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 01/17/2023] [Accepted: 02/20/2023] [Indexed: 05/18/2023]
Abstract
BACKGROUND Pancreatic cancer (PC) is an aggressive malignancy with poor prognosis. Accumulating studies have showed that long non-coding RNA (lncRNA) is a crucial regulator in various tumorigenesis and progression including PC. This research aims to explore the roles and molecular mechanism of lncRNA cancer susceptibility candidate 9 (CASC9) in PC. METHODS The expression levels of lncRNA CASC9 and miR-497-5p were evaluated in PC tissues and paired adjacent healthy tissues by quantitative real-time PCR. PC cell lines were transfected with lentivirus targeting lncRNA CASC9, and cells proliferation, migration and invasion tests were conducted. Dual luciferase reporter assays were also carried out to explore the relationship between lncRNA CASC9, miR-497-5p and Cyclin D1 (CCND1). RESULTS LncRNA CASC9 was significantly up-regulated in PC tissues, while miR-497-5p expression was down-regulated. Down-regulation of lncRNA CASC9 in PC cells can significantly suppress the cell aggressiveness both in vitro and in vivo; moreover, knock-down of miR-497-5p could neutralize this impact. Additionally, the luciferase activity assay has assured that CCND1 was a downstream target of miR-497-5p. CONCLUSION LncRNA CASC9 can promote the PC progression by modulating miR-497-5p/CCND1 axis, which is potential target for PC treatment.
Collapse
Affiliation(s)
- Jia Zhou
- Department of General Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
- Department of General Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guodong Song
- Department of General Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Gastrointestinal Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Mingqi Su
- Department of General Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui Zhang
- Department of General Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tingsong Yang
- Department of General Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhenshun Song
- Department of General Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
3
|
Chen S, Li L. Degradation strategy of cyclin D1 in cancer cells and the potential clinical application. Front Oncol 2022; 12:949688. [PMID: 36059670 PMCID: PMC9434365 DOI: 10.3389/fonc.2022.949688] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 07/28/2022] [Indexed: 02/02/2023] Open
Abstract
Cyclin D1 has been reported to be upregulated in several solid and hematologic tumors, promoting cancer progression. Thus, decreasing cyclin D1 by degradation could be a promising target strategy for cancer therapy. This mini review summarizes the roles of cyclin D1 in tumorigenesis and progression and its degradation strategies. Besides, we proposed an exploration of the degradation of cyclin D1 by FBX4, an F box protein belonging to the E3 ligase SKP-CUL-F-box (SCF) complex, which mediates substrate ubiquitination, as well as a postulate about the concrete combination mode of FBX4 and cyclin D1. Furthermore, we proposed a possible photodynamic therapy strategythat is based on the above concrete combination mode for treating superficial cancer.
Collapse
Affiliation(s)
- Shuyi Chen
- The Sixth Student Battalion, School of Basic Medical Sciences, Fourth Military Medical University, Xi’an, China
| | - Ling Li
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi’an, China
| |
Collapse
|
4
|
Zhou C, Huang Y, Wu J, Wei Y, Chen X, Lin Z, Nie S. A narrative review of multiple mechanisms of progranulin in cancer: a potential target for anti-cancer therapy. Transl Cancer Res 2021; 10:4207-4216. [PMID: 35116716 PMCID: PMC8798827 DOI: 10.21037/tcr-20-2972] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 07/30/2021] [Indexed: 12/14/2022]
Abstract
Progranulin (PGRN) is an autocrine growth factor and has important effects on regulation of cell growth, motility, tissue repair and embryonic development. Recent years, several researches found the expression of PGRN was at higher levels in a number of cancer cells and its high levels are associated with poor outcome of patients. More and more studies investigated the role of PGRN in cancer and found PGRN exerted various biological functions in cancer cells, such as promoting proliferation, inhibiting apoptosis, inducing migration and invasion of cells, accelerating angiogenesis and enhancing the effectiveness of chemoresistance and radiation. Now the effects of PGRN have been demonstrated in several cancers, including breast cancer, lung cancer, and bladder cancer. In addition, several signaling pathways and molecules are involved in the effects of PGRN on cancer cells, including Akt, mitogen-activated protein kinase (MAPK), vascular endothelial growth factor (VEGF) and cyclin D1. Therefore, PGRN is probably a significant diagnostic and prognostic biomarker for cancer and may be a potential target for anti-cancer therapy. Here, we reviewed the advancing field of PGRN in cancer as well as several signaling pathways activated by PGRN and confirmed PGRN is a key role in cancer. Moreover, future studies are still necessary to elucidate the biological functions and signaling pathways of PGRN in cancer.
Collapse
Affiliation(s)
- Chenhui Zhou
- Department of Neurosurgery, Ningbo First Hospital, Ningbo Hospital, Zhejiang University School of Medicine, Ningbo, China
| | - Yi Huang
- Department of Neurosurgery, Ningbo First Hospital, Ningbo Hospital, Zhejiang University School of Medicine, Ningbo, China
| | - Jingmi Wu
- Department of Neurosurgery, Ningbo First Hospital, Ningbo Hospital, Zhejiang University School of Medicine, Ningbo, China
| | - Yiting Wei
- Department of Neurosurgery, Ningbo First Hospital, Ningbo Hospital, Zhejiang University School of Medicine, Ningbo, China
| | - Xiaosheng Chen
- Department of Neurosurgery, Ningbo First Hospital, Ningbo University School of Medicine, Ningbo, China
| | - Zhiqing Lin
- Department of Neurosurgery, Ningbo First Hospital, Ningbo Hospital, Zhejiang University School of Medicine, Ningbo, China
| | - Sheng Nie
- Department of Neurosurgery, Ningbo First Hospital, Ningbo Hospital, Zhejiang University School of Medicine, Ningbo, China
| |
Collapse
|
5
|
Verel-Yilmaz Y, Fernández JP, Schäfer A, Nevermann S, Cook L, Gercke N, Helmprobst F, Jaworek C, Pogge von Strandmann E, Pagenstecher A, Bartsch DK, Bartsch JW, Slater EP. Extracellular Vesicle-Based Detection of Pancreatic Cancer. Front Cell Dev Biol 2021; 9:697939. [PMID: 34368146 PMCID: PMC8343017 DOI: 10.3389/fcell.2021.697939] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 06/29/2021] [Indexed: 12/12/2022] Open
Abstract
Due to a grim prognosis, there is an urgent need to detect pancreatic ductal adenocarcinoma (PDAC) prior to metastasis. However, reliable diagnostic imaging methods or biomarkers for PDAC or its precursor lesions are still scarce. ADAM8, a metalloprotease-disintegrin, is highly expressed in PDAC tissue and negatively correlates with patient survival. The aim of our study was to determine the ability of ADAM8-positive extracellular vesicles (EVs) and cargo microRNAs (miRNAs) to discriminate precursor lesions or PDAC from healthy controls. In order to investigate enrichment of ADAM8 on EVs, these were isolated from serum of patients with PDAC (n = 52), precursor lesions (n = 7) and healthy individuals (n = 20). Nanoparticle Tracking Analysis and electron microscopy indicated successful preparation of EVs that were analyzed for ADAM8 by FACS. Additionally, EV cargo analyses of miRNAs from the same serum samples revealed the presence of miR-720 and miR-451 by qPCR and was validated in 20 additional PDAC samples. Statistical analyses included Wilcoxon rank test and ROC curves. FACS analysis detected significant enrichment of ADAM8 in EVs from patients with PDAC or precursor lesions compared to healthy individuals (p = 0.0005). ADAM8-dependent co-variates, miR-451 and miR-720 were also diagnostic, as patients with PDAC had significantly higher serum levels of miR-451 and lower serum levels of miR-720 than healthy controls and reached high sensitivity and specificity (AUC = 0.93 and 1.00, respectively) to discriminate PDAC from healthy control. Thus, detection of ADAM8-positive EVs and related cargo miR-720 and miR-451 may constitute a specific biomarker set for screening individuals at risk for PDAC.
Collapse
Affiliation(s)
- Yesim Verel-Yilmaz
- Department of Visceral, Thoracic and Vascular Surgery, Philipps University Marburg, Marburg, Germany
| | - Juan Pablo Fernández
- Department of Visceral, Thoracic and Vascular Surgery, Philipps University Marburg, Marburg, Germany
| | - Agnes Schäfer
- Department of Neurosurgery, Philipps University Marburg, Marburg, Germany
| | - Sheila Nevermann
- Department of Visceral, Thoracic and Vascular Surgery, Philipps University Marburg, Marburg, Germany
| | - Lena Cook
- Department of Neurosurgery, Philipps University Marburg, Marburg, Germany
| | - Norman Gercke
- Department of Visceral, Thoracic and Vascular Surgery, Philipps University Marburg, Marburg, Germany
| | - Frederik Helmprobst
- Department of Neuropathology, Philipps University Marburg, Marburg, Germany.,Core Facility-Mouse Pathology and Electron Microscopy (MPEM), Philipps University Marburg, Marburg, Germany
| | - Christian Jaworek
- Department of Neurosurgery, Philipps University Marburg, Marburg, Germany
| | | | - Axel Pagenstecher
- Department of Neuropathology, Philipps University Marburg, Marburg, Germany
| | - Detlef K Bartsch
- Department of Visceral, Thoracic and Vascular Surgery, Philipps University Marburg, Marburg, Germany
| | - Jörg W Bartsch
- Department of Neurosurgery, Philipps University Marburg, Marburg, Germany
| | - Emily P Slater
- Department of Visceral, Thoracic and Vascular Surgery, Philipps University Marburg, Marburg, Germany
| |
Collapse
|
6
|
Fathi M, Ghafouri-Fard S, Abak A, Taheri M. Emerging roles of miRNAs in the development of pancreatic cancer. Biomed Pharmacother 2021; 141:111914. [PMID: 34328099 DOI: 10.1016/j.biopha.2021.111914] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 06/16/2021] [Accepted: 07/06/2021] [Indexed: 01/02/2023] Open
Abstract
Pancreatic cancer is a fatal cancer which is expected to exceed breast cancer as the third foremost source of cancer mortality by 2025. This cancer has been associated with several somatic genetic aberrations including mutations in the KRAS, CDKN2A/p16, TP53, and SMAD4. In addition, epigenetic alterations have been shown to affect development of this cancer. miRNAs are among the mostly appreciated epigenetic factors in this regard. Several oncomiRs such as miR-212, miR 506, miR-196b, miR-221-3p, miR-301a-3p, miR-23a and miR-29a have been found to promote proliferation of pancreatic cancer cells and block apoptotic pathways in these cells. On the other hand, miR-451a, miR-506, miR-142, miR-216b, miR-519d-3p, miR-1181, miR-340, miR-143-3p, miR-203a-3p, miR-455, miR-15a, miR-135a and miR-202 are among tumor suppressor miRNAs that modulate proliferation and cell cycle transition in these cells. In the current paper, we will discuss the role of oncomiRs and tumor suppressor miRNAs in the evolution of pancreatic cancer. Moreover, we will summarize the application of miRNAs as diagnostic and prognostic markers in pancreatic cancer. These studies have shown the ability of miRNAs to be served as non-invasive markers for pancreatic cancer.
Collapse
Affiliation(s)
- Mohadeseh Fathi
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Atefe Abak
- Men's Health and Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Taheri
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
7
|
Meng Y, Yang L, Wei X, Luo H, Hu Y, Tao X, He J, Zheng X, Xu Q, Luo K, Yu G, Luo Q. CCT5 interacts with cyclin D1 promoting lung adenocarcinoma cell migration and invasion. Biochem Biophys Res Commun 2021; 567:222-229. [PMID: 34217974 DOI: 10.1016/j.bbrc.2021.04.105] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 04/25/2021] [Indexed: 10/21/2022]
Abstract
Cyclin D1 (CCND1) has been identified as a metastatic promoter in various tumors including lung adenocarcinoma (LUAD), a subtype of non small cell lung cancer (NSCLC). The previous observation revealed that CCND1 was upregulated in NSCLC and predicted poor prognosis of LUAD patients. In this study, we examined a chaperonin containing TCP1 subunit 5 (CCT5) protein interacts with CCND1 in LUAD. Immunofluorescence demonstrated the co-localization of CCT5 and CCND1 protein in LUAD cells. CCT5 expression was detected with both immunohistochemistry (IHC) and bioinformatics analyses. Similar with the expression pattern of CCND1, CCT5 displayed a high level in LUAD tissues compared to non cancerous lung specimens. Patients with high CCT5 expression showed a significant shorter overall survival relative to those with low expression level. Furthermore, upregulated CCT5 exhibited significant positive correlation with TNM stage of LUAD patients in both IHC analyses and bioinformatics. Knocking down CCT5 remarkably inhibited LUAD cell migration and invasion in vitro by inactivating PI3K/AKT and its downstream EMT signals, which could abrogated the accelerated migration and invasion caused by CCND1 overexpression. In summary, our study discovered a highly expressed protein CCT5 in LUAD which interacted with CCND1 and promoted migration and invasion of LUAD cells by positively moderating PI3K/AKT-induced EMT pathway.
Collapse
Affiliation(s)
- Yiliang Meng
- Department of Oncology, Baise People's Hospital, Guangxi, Baise, 33000, Guangxi, China
| | - Liu Yang
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 510095, PR China
| | - Xiao Wei
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 510095, PR China
| | | | - Yingying Hu
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 510095, PR China
| | - Xingyu Tao
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 510095, PR China
| | - Jingjing He
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 510095, PR China
| | - Xuesong Zheng
- Department of Oncology, Baise People's Hospital, Guangxi, Baise, 33000, Guangxi, China
| | - Qunying Xu
- Department of Oncology, Baise People's Hospital, Guangxi, Baise, 33000, Guangxi, China
| | - Kunxiang Luo
- Department of Neurosurgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi, BaiSe, 533000, China
| | - Guifang Yu
- Department of Oncology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Qisheng Luo
- Department of Neurosurgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi, BaiSe, 533000, China.
| |
Collapse
|
8
|
Chen W, Zhu Y, Zhang W, Zhang H, Zhou Y, Sun P, Wu G. CDC42EP3 is a key promoter involved in the development and progression of gastric cancer. Carcinogenesis 2021; 42:1179-1188. [PMID: 34111280 DOI: 10.1093/carcin/bgab048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 05/19/2021] [Accepted: 06/08/2021] [Indexed: 12/13/2022] Open
Abstract
Gastric cancer (GC) is one of the most prevalent cancers and severely endangers human health. Due to the low rate of diagnosis, most patients with gastric cancer are diagnosed as advanced. CDC42 effector protein 3 (CDC42EP3) has been revealed to be involved in several types of human cancers' development and progression. However, the function of CDC42EP3 in GC is not yet clear. CDC42EP3 expression was detected by immunohistochemistry (IHC), qRT-PCR and Western blot assay in tumor tissues and cell lines of GC. CDC42EP3 knockdown cell models were constructed by lentivirus transfection. Cell proliferation was evaluated by the MTT assay. The wound-healing assay and the transwell assay were utilized to assess the cell migration. Also, the cell apoptosis and the cell cycle were evaluated by flow cytometry. Moreover, the mechanism was investigated by Human Apoptosis Antibody Array. The in vivo experiments were conducted to verify the effects of CDC42EP3 knockdown on the tumor growth of GC. The expression level of CDC42EP3 was up-regulated in tumor tissues. High CDC42EP3 expression was positively related to more advanced tumor grade. CDC42EP3 knockdown inhibited cell proliferation and migration, promoted cell apoptosis and suppressed the tumor growth. On the other hand, it was also found that the silencing of CDC42EP3 inhibited HSP27 and IGF-1sR expression as well as promoted Caspase3, p53, TNF-α, TNF-β, TRAILR-1 and TRAILR-2 expression. CDC42EP3 was revealed to work as a tumor promoter in the development and progression of GC, which could be a promising therapeutic target for the therapy of GC.
Collapse
Affiliation(s)
- Wenchao Chen
- Department of gastrointestinal surgery, Henan Provincial People's Hospital, Zhengzhou, Henan, 450003, China.,Zhengzhou University People's Hospital, Zhengzhou, Henan, 450003, China.,Henan University People's Hospital, Zhengzhou, Henan, 450003, China
| | - Yuanzeng Zhu
- Department of gastrointestinal surgery, Henan Provincial People's Hospital, Zhengzhou, Henan, 450003, China.,Zhengzhou University People's Hospital, Zhengzhou, Henan, 450003, China.,Henan University People's Hospital, Zhengzhou, Henan, 450003, China
| | - Wei Zhang
- Department of gastrointestinal surgery, Henan Provincial People's Hospital, Zhengzhou, Henan, 450003, China.,Zhengzhou University People's Hospital, Zhengzhou, Henan, 450003, China.,Henan University People's Hospital, Zhengzhou, Henan, 450003, China
| | - Han Zhang
- Department of gastrointestinal surgery, Henan Provincial People's Hospital, Zhengzhou, Henan, 450003, China.,Zhengzhou University People's Hospital, Zhengzhou, Henan, 450003, China.,Henan University People's Hospital, Zhengzhou, Henan, 450003, China
| | - Yang Zhou
- Department of gastrointestinal surgery, Henan Provincial People's Hospital, Zhengzhou, Henan, 450003, China.,Zhengzhou University People's Hospital, Zhengzhou, Henan, 450003, China.,Henan University People's Hospital, Zhengzhou, Henan, 450003, China
| | - Peichun Sun
- Department of gastrointestinal surgery, Henan Provincial People's Hospital, Zhengzhou, Henan, 450003, China.,Zhengzhou University People's Hospital, Zhengzhou, Henan, 450003, China.,Henan University People's Hospital, Zhengzhou, Henan, 450003, China
| | - Gang Wu
- Department of gastrointestinal surgery, Henan Provincial People's Hospital, Zhengzhou, Henan, 450003, China.,Zhengzhou University People's Hospital, Zhengzhou, Henan, 450003, China.,Henan University People's Hospital, Zhengzhou, Henan, 450003, China
| |
Collapse
|
9
|
Quercetin induces apoptosis and enhances gemcitabine therapeutic efficacy against gemcitabine-resistant cancer cells. Anticancer Drugs 2021; 31:684-692. [PMID: 32282368 DOI: 10.1097/cad.0000000000000933] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Quercetin, an abundant flavonoid found in various fruits and vegetables, displays multiple biological activities, including anticancer effects. Therefore, quercetin is receiving increasing attention as a potential adjuvant anticancer treatment. Gemcitabine (GEM) resistance is a major issue for clinicians and patients with advanced cancers, making it crucial to determine ways to bolster its effects. In this study, we explored the anticancer effects and mechanistic actions of quercetin in GEM-resistant cancer cells. Pancreatic cancer (BxPC-3, PANC-1) and hepatocellular carcinoma (HepG2, Huh-7) cell lines were studied. Proliferation assays showed that quercetin had cytotoxic effects on GEM-resistant cell lines (HepG2 and PANC-1), and flow cytometric analysis indicated a significant pro-apoptotic effect on these cell lines. GEM treatment, in combination with quercetin, resulted in increased anticancer effects compared with GEM alone. Quercetin led to S phase arrest in GEM-resistant cell lines, and western blot analysis revealed tumour protein p53 upregulation and cyclin D1 downregulation. This study provides mechanistic insight into the anticancer effects of quercetin and suggests that quercetin adjuvant treatment may benefit patients who are resistant to GEM therapy.
Collapse
|
10
|
Ren H, Mai G, Liu Y, Xiang R, Yang C, Su W. Eukaryotic Translation Initiation Factor 3 Subunit B Is a Promoter in the Development and Progression of Pancreatic Cancer. Front Oncol 2021; 11:644156. [PMID: 33996561 PMCID: PMC8116711 DOI: 10.3389/fonc.2021.644156] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 02/12/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Pancreatic cancer (PC) is a malignant tumor with hidden incidence, high degree of malignancy, rapid disease progression, and poor prognosis. Eukaryotic translation initiation factor 3 subunit B (EIF3B) is necessary for tumor growth, which is an alternative therapeutic target for many cancers. However, little is known about the relationship between EIF3B and PC. Methods: The expression of EIF3B in PC was detected by immunohistochemistry. EIF3B knockdown cell models were constructed by lentivirus infection. The MTT assay, the wound-healing assay, the transwell assay, the flow cytometry, and the Human Apoptosis Antibody Array was used to detect the effects of EIF3B knockdown on cell proliferation, cell migration, cell apoptosis, and cell cycle in vitro. Also, the effects of EIF3B knockdown on the tumor growth of PC were determined in vivo. Results: This study showed that the expression level of EIF3B was significantly up-regulated in PC tumor tissues and associated with pathological grade. In vitro, EIF3B knockdown inhibited the PC cell proliferation and migration, and the apoptosis levels were obviously promoted by regulating apoptosis-related proteins including Bcl-2, HSP27, HSP60, Survivin, sTNF-R2, TNF-α, TNF-β, TRAILR-3, TRAILR-4, and XIAP. Furthermore, the tumor growth of PC was inhibited after the knockdown of EIF3B in vivo. Conclusion: EIF3B was up-regulated in PC and was a promoter in the development and progression of PC, which could be considered as a therapeutic target for the treatment of PC.
Collapse
Affiliation(s)
- Haoyuan Ren
- Department of Gastrointestinal Surgery, People's Hospital of Deyang City, Deyang, China
| | - Gang Mai
- Department of Gastrointestinal Surgery, People's Hospital of Deyang City, Deyang, China
| | - Yong Liu
- Department of Gastrointestinal Surgery, People's Hospital of Deyang City, Deyang, China
| | - Rongchao Xiang
- Department of Gastrointestinal Surgery, People's Hospital of Deyang City, Deyang, China
| | - Chong Yang
- Organ Transplantation Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Wenjie Su
- Department of Anesthesiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.,Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| |
Collapse
|
11
|
miR-720 Regulates Insulin Secretion by Targeting Rab35. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6662612. [PMID: 33880375 PMCID: PMC8046542 DOI: 10.1155/2021/6662612] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 03/22/2021] [Accepted: 03/25/2021] [Indexed: 12/12/2022]
Abstract
miRNAs pose a good prospect in the diagnosis and treatment of type 2 diabetes (T2D). This study is aimed at investigating whether miR-720 targets Rab35 to regulate insulin secretion in MIN6 cells and its molecular mechanism and the clinical value of miR-720 as a specific biomarker of T2D. Fifty-five samples of new diagnosis T2D patients and normal control were collected. Levels of miR-720, fasting blood glucose, insulin, and other indicators of glucose and lipid metabolism were determined. We increased and decreased the miR-720 expression using miR-720 mimic and inhibitor to identify the effect of miR-720 on insulin secretion in MIN6 cells, respectively. Then, we used miR-720 mimic, miR-720 inhibitor, and dual luciferase reporter gene assays to prove miR-720 which regulates insulin secretion by targeting Rab35 in MIN6 cells. In addition, we overexpressed and silenced the Rab35 gene to detect the expression of PI3K, Akt, and mTOR in MIN6 cells by RT-PCR and western blot. In this study, circulating miR-720 was significantly higher in the T2D group than the control group, and miR-270 was positive correlated with FBG, while negatively correlated with FINS. The overexpression of miR-720 inhibited insulin secretion, and miR-720 downregulation promoted insulin secretion. miR-720 regulated insulin secretion by targeting Rab35 in MIN6 cells. Compared with the control group, the expression of PI3K, Akt, and mTOR was significantly decreased by the overexpression of the Rab35 gene, while the silencing Rab35 gene could induce the expression of PI3K, Akt, and mTOR. Furthermore, miR-720 mimic could activate the PI3K pathway. We conclude that miR-720 may be a potential biomarker for the diagnosis of T2D. Increase of miR-720 reduced the Rab35 expression then activate the PI3K/Akt/mTOR signal pathway, thus inhibiting insulin secretion.
Collapse
|
12
|
Nelson SR, Roche S, Cotter M, Garcia PA, Reitmeier D, Zollbrecht E, O'Neill F, Clynes M, Doolan P, Mehta JP, Swan N, Larkin A, Walsh N. Genomic Profiling and Functional Analysis of let-7c miRNA-mRNA Interactions Identify SOX13 to Be Involved in Invasion and Progression of Pancreatic Cancer. JOURNAL OF ONCOLOGY 2020; 2020:2951921. [PMID: 33424970 PMCID: PMC7775161 DOI: 10.1155/2020/2951921] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 12/01/2020] [Accepted: 12/11/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND Pancreatic cancer is a devastating disease; its lethality is related to rapid growth and tendency to invade adjacent organs and metastasize at an early stage. OBJECTIVE The aim of this study was to identify miRNAs and their gene targets involved in the invasive phenotype in pancreatic cancer to better understand the biological behaviour and the rapid progression of this disease. METHODS miRNA profiling was performed in isogenic matched high invasive and low-invasive subclones derived from the MiaPaCa-2 cell line and validated in a panel of pancreatic cancer cell lines, tumour, and normal pancreas. Online miRNA target prediction algorithms and gene expression arrays were used to predict the target genes of the differentially expressed miRNAs. miRNAs and potential target genes were subjected to overexpression and knockdown approaches and downstream functional assays to determine their pathological role in pancreatic cancer. RESULTS Differential expression analysis revealed 10 significantly dysregulated miRNAs associated with invasive capacity (Student's t-tests; P value <0.05; fold change = ±2). The expression of top upregulated miR-135b and downregulated let-7c miRNAs correlated with the invasive abilities of eight pancreatic cancer cell lines and displayed differential expression in pancreatic cancer and adjacent normal tissue specimens. Ectopic overexpression of let-7c decreased proliferation, invasion, and colony formation. Integrated analysis of miRNA-mRNA using in silico algorithms and experimental validation databases identified four putative gene targets of let-7c. One of these targets, SOX13, was found to be upregulated in PDAC tumour compared with normal tissue in TCGA and an independent data set by qPCR and immunohistochemistry. RNAi knockdown of SOX13 reduced the invasion and colony formation ability of pancreatic cancer cells. CONCLUSION The identification of key miRNA-mRNA gene interactions and networks provide potential diagnostic and therapeutic strategies for better treatment options for pancreatic cancer patients.
Collapse
Affiliation(s)
- Shannon R. Nelson
- National Institute for Cellular Biotechnology, School of Biotechnology, Dublin City University, Dublin 9, Ireland
| | - Sandra Roche
- National Institute for Cellular Biotechnology, School of Biotechnology, Dublin City University, Dublin 9, Ireland
| | - Maura Cotter
- Histopathology Department, St. Vincent's University Hospital, Elm Park, Dublin 4, Ireland
| | - Pablo Anton Garcia
- National Institute for Cellular Biotechnology, School of Biotechnology, Dublin City University, Dublin 9, Ireland
| | - Daniela Reitmeier
- National Institute for Cellular Biotechnology, School of Biotechnology, Dublin City University, Dublin 9, Ireland
| | - Elisabeth Zollbrecht
- National Institute for Cellular Biotechnology, School of Biotechnology, Dublin City University, Dublin 9, Ireland
| | - Fiona O'Neill
- National Institute for Cellular Biotechnology, School of Biotechnology, Dublin City University, Dublin 9, Ireland
| | - Martin Clynes
- National Institute for Cellular Biotechnology, School of Biotechnology, Dublin City University, Dublin 9, Ireland
| | - Padraig Doolan
- National Institute for Cellular Biotechnology, School of Biotechnology, Dublin City University, Dublin 9, Ireland
| | - Jai P. Mehta
- National Institute for Cellular Biotechnology, School of Biotechnology, Dublin City University, Dublin 9, Ireland
| | - Niall Swan
- Histopathology Department, St. Vincent's University Hospital, Elm Park, Dublin 4, Ireland
| | - AnneMarie Larkin
- National Institute for Cellular Biotechnology, School of Biotechnology, Dublin City University, Dublin 9, Ireland
| | - Naomi Walsh
- National Institute for Cellular Biotechnology, School of Biotechnology, Dublin City University, Dublin 9, Ireland
| |
Collapse
|
13
|
Yu Y, Yan R, Chen X, Sun T, Yan J. Paeonol suppresses the effect of ox-LDL on mice vascular endothelial cells by regulating miR-338-3p/TET2 axis in atherosclerosis. Mol Cell Biochem 2020; 475:127-135. [PMID: 32770325 DOI: 10.1007/s11010-020-03865-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 07/28/2020] [Indexed: 10/23/2022]
Abstract
Atherosclerosis is the common vascular disease. Vascular smooth muscle cell proliferation and vascular endothelial cell (VEC) dysfunction are involved in the causes of atherosclerosis. And oxidized low-density lipoprotein (ox-LDL)-induced vascular endothelial cells (VECs) are suitable models for studying atherosclerosis development. Paeonol was reported to repress ox-LDL-induced VEC progression. However, its detailed mechanism was not fully reported. MicroRNAs (miRNAs) acted as regulators in multiple diseases. Previous findings found that microRNA-338-3p (miR-338-3p) was overexpressed in Atherosclerosis process. However, the function and underlying mechanism of miR-338-3p in ox-LDL-treated VECs needed to be elucidated. The purpose of this research was to reveal the role of miR-338-3p in paeonol-regulated ox-LDL-induced VEC progression. Cell counting kit-8 (CCK-8) and flow cytometry were employed to determine cell viability and apoptosis, respectively. Moreover, the levels of IL-6 and IL-1β were analyzed using enzyme-linked immunosorbent assay, as well as the contents of reactive oxygen species, lactate dehydrogenase, and malonic dialdehyde were investigated using related kits. Furthermore, quantitative real-time polymerase chain reaction was carried out to determine the expression of miR-338-3p. Western blot assay was conducted to detect the level of tet methylcytosine dioxygenase 2 (TET2). Besides, the interaction between miR-338-3p and TET2 was predicted by DIANA, and then confirmed by the dual-luciferase reporter assay and RNA immunoprecipitation assay. Ox-LDL repressed mice VEC viability, and promoted apoptosis, inflammatory response, and oxidative injury. Paeonol inhibited the effect of ox-LDL on the growth of the VECs. Furthermore, paeonol regulated VEC development via downregulating miR-338-3p expression. Interestingly, miR-338-3p targeted TET2 and inhibited TET2 expression. MiR-338-3p modulated ox-LDL-treated VEC growth through suppressing TET2 expression. We demonstrated that paeonol attenuated the effect of ox-LDL on the development of mice VECs via modulating miR-338-3p/TET2 axis, providing a theoretical basis for the treatment of AS.
Collapse
Affiliation(s)
- Yunfu Yu
- Department of Cardiology, Central China Fuwai Hospital, Heart Center of Henan Provincial People's Hospital, No. 1 Yuwai Street, Zhengdong New District, Zhengzhou, 450000, Henan Province, China
| | - Rui Yan
- Department of Cardiology, Central China Fuwai Hospital, Heart Center of Henan Provincial People's Hospital, No. 1 Yuwai Street, Zhengdong New District, Zhengzhou, 450000, Henan Province, China
| | - Xiaozhen Chen
- Department of Cardiology, Central China Fuwai Hospital, Heart Center of Henan Provincial People's Hospital, No. 1 Yuwai Street, Zhengdong New District, Zhengzhou, 450000, Henan Province, China
| | - Tao Sun
- Department of Cardiology, Central China Fuwai Hospital, Heart Center of Henan Provincial People's Hospital, No. 1 Yuwai Street, Zhengdong New District, Zhengzhou, 450000, Henan Province, China
| | - Jifeng Yan
- Department of Cardiology, Central China Fuwai Hospital, Heart Center of Henan Provincial People's Hospital, No. 1 Yuwai Street, Zhengdong New District, Zhengzhou, 450000, Henan Province, China.
| |
Collapse
|
14
|
Qian X, Chen Z, Chen SS, Liu LM, Zhang AQ. Integrated Analyses Identify Immune-Related Signature Associated with Qingyihuaji Formula for Treatment of Pancreatic Ductal Adenocarcinoma Using Network Pharmacology and Weighted Gene Co-Expression Network. J Immunol Res 2020; 2020:7503605. [PMID: 32537471 PMCID: PMC7256764 DOI: 10.1155/2020/7503605] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 04/15/2020] [Indexed: 02/06/2023] Open
Abstract
The study aimed to clarify the potential immune-related targets and mechanisms of Qingyihuaji Formula (QYHJ) against pancreatic cancer (PC) through network pharmacology and weighted gene co-expression network analysis (WGCNA). Active ingredients of herbs in QYHJ were identified by the TCMSP database. Then, the putative targets of active ingredients were predicted with SwissTargetPrediction and the STITCH databases. The expression profiles of GSE32676 were downloaded from the GEO database. WGCNA was used to identify the co-expression modules. Besides, the putative targets, immune-related targets, and the critical module genes were mapped with the specific disease to select the overlapped genes (OGEs). Functional enrichment analysis of putative targets and OGEs was conducted. The overall survival (OS) analysis of OGEs was investigated using the Kaplan-Meier plotter. The relative expression and methylation levels of OGEs were detected in UALCAN, human protein atlas (HPA), Oncomine, DiseaseMeth version 2.0 and, MEXPRESS database, respectively. Gene set enrichment analysis (GSEA) was conducted to elucidate the key pathways of highly-expressed OGEs further. OS analyses found that 12 up-regulated OGEs, including CDK1, PLD1, MET, F2RL1, XDH, NEK2, TOP2A, NQO1, CCND1, PTK6, CTSE, and ERBB2 that could be utilized as potential diagnostic indicators for PC. Further, methylation analyses suggested that the abnormal up-regulation of these OGEs probably resulted from hypomethylation, and GSEA revealed the genes markedly related to cell cycle and proliferation of PC. This study identified CDK1, PLD1, MET, F2RL1, XDH, NEK2, TOP2A, NQO1, CCND1, PTK6, CTSE, and ERBB2 might be used as reliable immune-related biomarkers for prognosis of PC, which may be essential immunotherapies targets of QYHJ.
Collapse
Affiliation(s)
- Xiang Qian
- Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Hangzhou, China
- Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou, China
- Zhejiang Cancer Hospital, Hangzhou, China
| | - Zhuo Chen
- Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Hangzhou, China
- Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou, China
- Zhejiang Cancer Hospital, Hangzhou, China
| | - Sha Sha Chen
- Department of Traditional Chinese Medicine, Taizhou Cancer Hospital, Zhejiang, China
| | - Lu Ming Liu
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, China
| | - Ai Qin Zhang
- Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Hangzhou, China
- Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou, China
- Zhejiang Cancer Hospital, Hangzhou, China
| |
Collapse
|
15
|
Qian Z, Gong L, Mou Y, Han Y, Zheng S. MicroRNA‑203a‑3p is a candidate tumor suppressor that targets thrombospondin 2 in colorectal carcinoma. Oncol Rep 2019; 42:1825-1832. [PMID: 31545460 PMCID: PMC6775819 DOI: 10.3892/or.2019.7310] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 08/16/2019] [Indexed: 12/24/2022] Open
Abstract
The aim of the present study was to investigate the role of miR-203a-3p in colorectal cancer (CRC) and identify the target gene of microRNA (miR)-203a-3p. A total of 59 sets of cancer tissues and corresponding adjacent non-tumor tissues were collected from CRC patients (aged 31–78 years) between October 2016 and May 2017. Total RNA extraction and reverse transcription-quantitative polymerase chain reaction analysis, transfection assay, and Transwell and apoptosis assays, western blot analysis, a luciferase reporter assay and immunohistochemistry were performed. miR-203a-3p was found to be significantly downregulated in CRC tissues compared with adjacent normal tissues. The overexpression of miR-203a-3p was shown to inhibit the invasion and migration of human CRC SW480 and HT29 cells, and increase their apoptosis rates. Furthermore, miR-203a-3p downregulated the expression of thrombospondin 2 (THBS2) in SW480 and HT29 cells. It was also experimentally demonstrated that miR-203a-3p binds to the 3′-untranslated region of THBS2, downregulating THBS2 expression and thereby inhibiting CRC progression and metastasis. The expression of miR-203a-3p, which serves a tumor-suppressive role, in CRC tissues was significantly downregulated. As miR-203a-3p was determined to target THBS2 to inhibit CRC progression and metastasis; thus, miR-203a-3p may be considered as a potential novel approach to treating CRC.
Collapse
Affiliation(s)
- Zhenyuan Qian
- Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, P.R. China
| | - Lijie Gong
- Department of Breast Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Yiping Mou
- Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Yong Han
- Clinical Research Institute of Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Shusen Zheng
- Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, P.R. China
| |
Collapse
|
16
|
Cheng X, Wang X, Wu Z, Tan S, Zhu T, Ding K. CORO1C expression is associated with poor survival rates in gastric cancer and promotes metastasis in vitro. FEBS Open Bio 2019; 9:1097-1108. [PMID: 30974047 PMCID: PMC6551501 DOI: 10.1002/2211-5463.12639] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 03/13/2019] [Accepted: 04/08/2019] [Indexed: 12/21/2022] Open
Abstract
Coronin-like actin-binding protein 1C (CORO1C) is a member of the WD repeat protein family that regulates actin-dependent processes by assembling F-actin. CORO1C was previously reported to promote metastasis in breast cancer and lung squamous cell carcinoma. Here, we investigated the role of CORO1C in gastric cancer. Higher expression levels of CORO1C were detected in gastric cancer tissues as compared with normal gastric tissues. In addition, CORO1C levels were found to be positively correlated with lymph node metastasis in gastric cancer patients. The expression levels of CORO1C were higher in stage III-IV gastric cancer patients (80.8%) than in stage I-II gastric cancer patients(57.1%). Gastric cancer patients positive for CORO1C expression showed lower relapse-free survival and overall survival rates. Knockdown of CORO1C dramatically suppressed total cell number, cell viability, cell colony formation, cell mitosis and cell metastasis, and promoted apoptosis of gastric cancer cells. Furthermore, cyclin D1 and vimentin were found to be positively regulated by CORO1C. As cyclin D1 and vimentin play an oncogenic role in gastric cancer, CORO1C may exert its tumor-promoting activity through these proteins.
Collapse
Affiliation(s)
- Xiao Cheng
- Department of Pathology, Anhui Medical University, Hefei, China
| | - Xiaonan Wang
- Laboratory of Pathogenic Microbiology and Immunology, Anhui Medical University, Hefei, China
| | - Zhengsheng Wu
- Department of Pathology, Anhui Medical University, Hefei, China
| | - Sheng Tan
- Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, China
| | - Tao Zhu
- Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, China
| | - Keshuo Ding
- Department of Pathology, Anhui Medical University, Hefei, China
| |
Collapse
|
17
|
Chen G, Hu M, Qu X, Wang K, Qu Y. MicroRNA‑584 directly targets CCND1 and inhibits cell proliferation and invasion in pancreatic cancer. Mol Med Rep 2018; 19:719-726. [PMID: 30431107 DOI: 10.3892/mmr.2018.9651] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 10/16/2018] [Indexed: 11/06/2022] Open
Abstract
Multiple previous studies have demonstrated that the dysregulation of microRNAs (miRNAs) is implicated in the occurrence and development of pancreatic cancer. Therefore, a further characterisation of deregulated miRNAs in pancreatic cancer may provide novel insight into the oncogenesis and progression of pancreatic cancer, which may facilitate the identification of effective therapeutic targets for treating patients with this disease. In the present study, reverse transcription‑quantitative polymerase chain reaction analysis demonstrated that the expression level of miRNA‑584‑5p (miR‑584) was significantly decreased in pancreatic cancer tissues and cell lines. It was demonstrated that restoration of miR‑584 expression significantly suppressed the proliferative and invasive ability of pancreatic cancer cells. Bioinformatics analysis predicted that cyclin D1 (CCND1) was a putative target of miR‑584. Subsequent experiments demonstrated that CCND1 was a direct target gene of miR‑584 in pancreatic cancer cells. Furthermore, the inhibition of CCND1 mimicked the suppressive effect of miR‑584 overexpression in pancreatic cancer cells. The restoration of CCND1 expression significantly abolished the inhibitory effects of miR‑584 overexpression on pancreatic cancer cells. Collectively, the present results demonstrated that miR‑584 inhibited the development of pancreatic cancer by directly targeting CCND1, suggesting that this miRNA may represent a potential therapeutic target for this fatal disease.
Collapse
Affiliation(s)
- Gang Chen
- Department of Gastroenterology, First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang 154003, P.R. China
| | - Ming Hu
- Department of General Surgery, First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang 154003, P.R. China
| | - Xiusheng Qu
- Department of Radiochemotherapy, First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang 154003, P.R. China
| | - Kaifeng Wang
- Department of Vascular Surgery, First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang 154003, P.R. China
| | - Yikun Qu
- Department of General Surgery, First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang 154003, P.R. China
| |
Collapse
|
18
|
Liu ZZ, Fang CC, Huang MJ. RNA interference-mediated Gal-3 knockdown inhibits proliferation and promotes apoptosis of pancreatic cancer cells. Shijie Huaren Xiaohua Zazhi 2018; 26:648-654. [DOI: 10.11569/wcjd.v26.i11.648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate the effect of RNA interference-mediated knockdown of galectin-3 (Gal-3) on the proliferation and apoptosis of pancreatic cancer cells and the possible mechanism involved.
METHODS Cultured Panc-1 cells were randomly divided into a control group (untreated), an NC group (transfected with control siRNA) and a Gal-3 interference group (transfected with Gal-3-siRNA). After using small interfering RNA (siRNA) technology to interfere with the expression of Gal-3 in Panc-1 cells, Gal-3 expression was detected by RT-PCR and Western blot, cell proliferation was measured by CCK-8 assay, and cell apoptosis was tested by flow cytometry. The expression of Ki67, cleaved caspase-3, and β-catenin proteins was examined by Western blot.
RESULTS Compared with the control group, there was no significant difference in the expression of Gal-3 mRNA (0.99 ± 0.08 vs 1.01 ± 0.06) or protein (0.36 ± 0.03 vs 0.34 ± 0.05) in the NC group (P > 0.05), while the expression of Gal-3 mRNA (0.38 ± 0.02 vs 1.01 ± 0.06) and protein (0.10 ± 0.01 vs 0.34 ± 0.05) in the Gal-3 interference group was significantly decreased (P < 0.05). Compared with the control group, RNA interference-mediated knockdown of Gal-3 significantly decreased cell proliferation (24 h: 0.55 ± 0.03 vs 0.71 ± 0.05; 48 h: 0.76 ± 0.05 vs 0.97 ± 0.06; 72 h: 1.08 ± 0.06 vs 1.32 ± 0.09), increased the percentage of cells in G0/G1 stage (79.48 ± 1.32 vs 71.52 ± 1.15), reduced the percentage of cells in S (14.26 ± 1.08 vs 18.24 ± 1.06 ) and G2/M stages (6.21 ± 0.78 vs 10.19 ± 1.52), increased apoptosis (13.26 ± 2.28 vs 5.82 ± 0.35), downregulated the expression of Ki67 (0.24 ± 0.02 vs 0.96 ± 0.07), cyclin D1 (0.26 ± 0.03 vs 0.88 ± 0.09), and β-catenin (0.42 ± 0.05 vs 0.87 ± 0.05), and upregulated the expression of cleaved caspase-3 (0.70 ± 0.06 vs 0.32 ± 0.03) (P < 0.05 for all). There was no significant difference in the above indexes between the NC group and control group (P > 0.05).
CONCLUSION RNA interference-mediated Gal-3 knockdown can inhibit proliferation and promote apoptosis of pancreatic cancer cells via mechanisms that may be related to the inhibition of the Wnt/ beta-catenin signaling pathway.
Collapse
Affiliation(s)
- Zhi-Zhi Liu
- Department of Gastroenterology, Luqiao Hospital of Enze Medical Center (Group) of Taizhou City, Taizhou 318050, Zhejiang Province, China
| | - Cong-Cheng Fang
- Department of Gastroenterology, Enze Medical Center (Group) Taizhou Hospital, Taizhou 318050, Zhejiang Province, China
| | - Mei-Jun Huang
- Department of Gastroenterology, Luqiao Hospital of Enze Medical Center (Group) of Taizhou City, Taizhou 318050, Zhejiang Province, China
| |
Collapse
|