1
|
Portela LMF, Constantino FB, Camargo ACL, Santos SAA, Colombelli KT, Fioretto MN, Barata LA, Silva EJR, Scarano WR, Felisbino SL, Moreno CS, Justulin LA. Early-life origin of prostate cancer through deregulation of miR-206 networks in maternally malnourished offspring rats. Sci Rep 2023; 13:18685. [PMID: 37907720 PMCID: PMC10618455 DOI: 10.1038/s41598-023-46068-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 10/27/2023] [Indexed: 11/02/2023] Open
Abstract
The Developmental Origins of Health and Disease (DOHaD) concept has provided the framework to assess how early life experiences can shape health and disease throughout the life course. While maternal malnutrition has been proposed as a risk factor for the developmental programming of prostate cancer (PCa), the molecular mechanisms remain poorly understood. Using RNA-seq data, we demonstrated deregulation of miR-206-Plasminogen (PLG) network in the ventral prostate (VP) of young maternally malnourished offspring. RT-qPCR confirmed the deregulation of the miR-206-PLG network in the VP of young and old offspring rats. Considering the key role of estrogenic signaling pathways in prostate carcinogenesis, in vitro miRNA mimic studies also revealed a negative correlation between miR-206 and estrogen receptor α (ESR1) expression in PNT2 cells. Together, we demonstrate that early life estrogenization associated with the deregulation of miR-206 networks can contribute to the developmental origins of PCa in maternally malnourished offspring. Understanding the molecular mechanisms by which early life malnutrition affects offspring health can encourage the adoption of a governmental policy for the prevention of non-communicable chronic diseases related to the DOHaD concept.
Collapse
Affiliation(s)
- Luiz M F Portela
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University (UNESP), Botucatu, SP, 18618-689, Brazil
| | - Flavia B Constantino
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University (UNESP), Botucatu, SP, 18618-689, Brazil
| | - Ana C L Camargo
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University (UNESP), Botucatu, SP, 18618-689, Brazil
| | - Sérgio A A Santos
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University (UNESP), Botucatu, SP, 18618-689, Brazil
- Cancer Signaling and Epigenetics Program, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
| | - Ketlin T Colombelli
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University (UNESP), Botucatu, SP, 18618-689, Brazil
| | - Matheus N Fioretto
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University (UNESP), Botucatu, SP, 18618-689, Brazil
| | - Luísa A Barata
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University (UNESP), Botucatu, SP, 18618-689, Brazil
| | - Erick J R Silva
- Department of Biophysics and Pharmacology, Institute of Biosciences, Unesp, Botucatu, Brazil
| | - Wellerson R Scarano
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University (UNESP), Botucatu, SP, 18618-689, Brazil
| | - Sergio L Felisbino
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University (UNESP), Botucatu, SP, 18618-689, Brazil
| | - Carlos S Moreno
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
- Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, GA, USA
| | - Luis A Justulin
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University (UNESP), Botucatu, SP, 18618-689, Brazil.
| |
Collapse
|
2
|
Probable Mechanisms of Doxorubicin Antitumor Activity Enhancement by Ginsenoside Rh2. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27030628. [PMID: 35163891 PMCID: PMC8838402 DOI: 10.3390/molecules27030628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 01/16/2022] [Accepted: 01/17/2022] [Indexed: 11/19/2022]
Abstract
Ginsenoside Rh2 increases the efficacy of doxorubicin (DOX) treatment in murine models of solid and ascites Ehrlich’s adenocarcinoma. In a solid tumor model (treatment commencing 7 days after inoculation), DOX + Rh2 co-treatment was significantly more efficacious than DOX alone. If treatment was started 24 h after inoculation, the inhibition of tumor growth of a solid tumor for the DOX + Rh2 co-treatment group was complete. Furthermore, survival in the ascites model was dramatically higher for the DOX + Rh2 co-treatment group than for DOX alone. Mechanisms underlying the combined DOX and Rh2 effects were studied in primary Ehrlich’s adenocarcinoma-derived cells and healthy mice’s splenocytes. Despite the previously established Rh2 pro-oxidant activity, DOX + Rh2 co-treatment revealed no increase in ROS compared to DOX treatment alone. However, DOX + Rh2 treatment was more effective in suppressing Ehrlich adenocarcinoma cell adhesion than either treatment alone. We hypothesize that the benefits of DOX + Rh2 combination treatment are due to the suppression of tumor cell attachment/invasion that might be effective in preventing metastatic spread of tumor cells. Ginsenoside Rh2 was found to be a modest activator in a Neh2-luc reporter assay, suggesting that Rh2 can activate the Nrf2-driven antioxidant program. Rh2-induced direct activation of Nrf2 might provide additional benefits by minimizing DOX toxicity towards non-cancerous cells.
Collapse
|
3
|
LncRNA MIR155HG induces M2 macrophage polarization and drug resistance of colorectal cancer cells by regulating ANXA2. Cancer Immunol Immunother 2021; 71:1075-1091. [PMID: 34562123 DOI: 10.1007/s00262-021-03055-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 09/07/2021] [Indexed: 02/06/2023]
Abstract
OBJECTIVE To investigate the effects of lncRNA MIR155HG and Annexin A2 (ANXA2) on colorectal cancer (CRC) and the mechanism of the MIR155HG/ANXA2 axis. METHODS The expressions of MIR155HG and ANXA2 in human CRC tissues were analyzed for association with pathological characteristics and prognosis of CRC patients. CRC cell lines (Caco2 and HT29) were used to study the effects of MIR155HG or ANXA2 knockdown on tumor cell behaviors and macrophage polarization as well as the effect of M2 polarization on oxaliplatin resistance of CRC cells. RNA immunoprecipitation, RNA pull-down and dual-luciferase reporter assays were applied to verify the targeting relationships among MIR155HG, miR-650 and ANXA2. Heterotopic xenograft models were established to verify the results of cell experiments. RESULTS MIR155HG and ANXA2 were highly expressed in CRC tissues/cells and of prognostic values for CRC patients. Knockdown of MIR155HG or ANXA2 suppressed M2 macrophage polarization, and proliferation, migration, invasion and oxaliplatin resistance of CRC cells. MIR155HG competed with ANXA2 for binding miR-650 and can also directly target ANXA2. Knockdown of MIR155HG or ANXA2 also inhibited M2 macrophage polarization and CRC progression in nude mice. CONCLUSION This study highlighted that MIR155HG, by regulating the miR-650/ANXA2 axis, promotes CRC progression and enhances oxaliplatin resistance in CRC cells through M2 macrophage polarization.
Collapse
|
4
|
Wang H, Zhang K, Liu J, Yang J, Tian Y, Yang C, Li Y, Shao M, Su W, Song N. Curcumin Regulates Cancer Progression: Focus on ncRNAs and Molecular Signaling Pathways. Front Oncol 2021; 11:660712. [PMID: 33912467 PMCID: PMC8072122 DOI: 10.3389/fonc.2021.660712] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 03/24/2021] [Indexed: 12/24/2022] Open
Abstract
Curcumin [(1E,6E) ‑1,7‑bis(4‑hydroxy‑3‑methoxyphenyl) hepta‑1,6‑diene‑3,5‑ dione] is a natural polyphenol derived from the rhizome of the turmeric plant Curcuma longa. Accumulated evidences have presented curcumin’s function in terms of anti-inflammatory, antioxidant properties, and especially anti-tumor activities. Studies demonstrated that curcumin could exert anti-tumor activity via multiple biological signaling pathways, such as PI3K/Akt, JAK/STAT, MAPK, Wnt/β-catenin, p53, NF-ĸB and apoptosis related signaling pathways. Moreover, Curcumin can inhibit tumor proliferation, angiogenesis, epithelial-mesenchymal transition (EMT), invasion and metastasis by regulating tumor related non-coding RNA (ncRNA) expression. In this review, we summarized the roles of curcumin in regulating signaling pathways and ncRNAs in different kinds of cancers. We also discussed the regulatory effect of curcumin through inhibiting carcinogenic miRNA and up regulating tumor suppressive miRNA. Furthermore, we aim to illustrate the cross regulatory relationship between ncRNA and signaling pathways, further to get a better understanding of the anti-tumor mechanism of curcumin, thus lay a theoretical foundation for the clinical application of curcumin in the future.
Collapse
Affiliation(s)
- Haijun Wang
- Department of Pathology, Key Laboratory of Clinical Molecular Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China.,School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Ke Zhang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Jia Liu
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Jie Yang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Yidan Tian
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Chen Yang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Yushan Li
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Minglong Shao
- Department of Mental Health, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Wei Su
- Department of Pathology, Key Laboratory of Clinical Molecular Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Na Song
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China.,Institute of Precision Medicine, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
5
|
Greene J, Baird AM, Lim M, Flynn J, McNevin C, Brady L, Sheils O, Gray SG, McDermott R, Finn SP. Differential CircRNA Expression Signatures May Serve as Potential Novel Biomarkers in Prostate Cancer. Front Cell Dev Biol 2021; 9:605686. [PMID: 33718350 PMCID: PMC7946979 DOI: 10.3389/fcell.2021.605686] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 02/05/2021] [Indexed: 12/17/2022] Open
Abstract
Circular RNAs (circRNAs), a recently discovered non-coding RNA, have a number of functions including the regulation of miRNA expression. They have been detected in a number of malignancies including prostate cancer (PCa). The differential expression pattern of circRNAs associated with PCa and androgen receptor (AR) status was investigated in this study. circRNA profiling was performed using a high throughout microarray assay on a panel of prostate cell lines, which consisted of normal, benign, and malignant cells (n = 9). circRNAs were more commonly significantly up-regulated (p < 0.05) than downregulated in malignant cell lines (n = 3,409) vs. benign cell lines (n = 2,949). In a grouped analysis based on AR status, there were 2,127 down-regulated circRNAs in androgen independent cell lines compared to 2,236 in androgen dependent cell lines, thus identifying a potential circRNA signature reflective of androgen dependency. Through a bioinformatics approach, the parental genes associated with the top 10 differentially expressed circRNAs were identified such as hsa_circ_0064644, whose predicted parental gene target is RBMS3, and hsa_circ_0060539, whose predicted gene target is SDC4. Furthermore, we identified three circRNAs associated with the parental gene Caprin1 (hsa_circ_0021652, hsa_circ_0000288, and hsa_circ_0021647). Other studies have shown the importance of Caprin1 in PCa cell survival and drug resistance. Given the modified circRNA expression signatures identified here, these hypothesis generating results suggest that circRNAs may serve as potential putative diagnostic and predictive markers in PCa. However, further validation studies are required to assess the true potential of these markers in the clinical setting.
Collapse
Affiliation(s)
- John Greene
- Department of Histopathology and Morbid Anatomy, School of Medicine, Trinity College, Dublin, Ireland.,Department of Medical Oncology, Tallaght University Hospital, Dublin, Ireland
| | - Anne-Marie Baird
- School of Medicine, Trinity Translational Medicine Institute, Trinity College, Dublin, Ireland
| | - Marvin Lim
- Department of Histopathology and Morbid Anatomy, School of Medicine, Trinity College, Dublin, Ireland.,Department of Medical Oncology, Tallaght University Hospital, Dublin, Ireland
| | - Joshua Flynn
- School of Medicine, Trinity Translational Medicine Institute, Trinity College, Dublin, Ireland
| | - Ciara McNevin
- Department of Histopathology and Morbid Anatomy, School of Medicine, Trinity College, Dublin, Ireland.,Department of Medical Oncology, Tallaght University Hospital, Dublin, Ireland
| | - Lauren Brady
- Department of Histopathology and Morbid Anatomy, School of Medicine, Trinity College, Dublin, Ireland
| | - Orla Sheils
- Department of Histopathology and Morbid Anatomy, School of Medicine, Trinity College, Dublin, Ireland.,School of Medicine, Trinity Translational Medicine Institute, Trinity College, Dublin, Ireland
| | - Steven G Gray
- School of Medicine, Trinity Translational Medicine Institute, Trinity College, Dublin, Ireland.,Thoracic Oncology Research Group, Trinity Translational Medicine Institute, St. James's Hospital, Dublin, Ireland
| | - Raymond McDermott
- Department of Medical Oncology, Tallaght University Hospital, Dublin, Ireland.,Department of Medical Oncology, St. Vincent's University Hospital, Dublin, Ireland
| | - Stephen P Finn
- Department of Histopathology and Morbid Anatomy, School of Medicine, Trinity College, Dublin, Ireland.,Thoracic Oncology Research Group, Trinity Translational Medicine Institute, St. James's Hospital, Dublin, Ireland.,Department of Histopathology, St. James's Hospital, Dublin, Ireland
| |
Collapse
|
6
|
Li B, Chen Z, Luo X, Zhang C, Chen H, Wang S, Zhao M, Ma H, Liu J, Cheng M, Yang Y, Yan H. Butylphthalide Inhibits Autophagy and Promotes Multiterritory Perforator Flap Survival. Front Pharmacol 2021; 11:612932. [PMID: 33584290 PMCID: PMC7878674 DOI: 10.3389/fphar.2020.612932] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 12/14/2020] [Indexed: 11/13/2022] Open
Abstract
Multiterritory perforator flap is an important plastic surgery technique, yet its efficacy can be limited by partial necrosis at the choke Ⅱ zone. Butylphthalide (NBP) has been used for many diseases but has not been studied in the multiterritory perforator flap. With the effect of NBP, we observed increasing in capillary density, inhibition of autophagy and oxidative stress, and a reduction in apoptosis of cells, all consistent with increased flap survival. However, the protective effect of NBP on multiterritory perforator flap was lost following administration of the autophagy agonist rapamycin (Rap). Through the above results, we assumed that NBP promotes flap survival by inhibiting autophagy. Thus, this study has found a new pharmacological effect of NBP on the multiterritory perforator by inhibiting autophagy to prevent distal postoperative necrosis and exert effects on angiogenesis, oxidative stress, and apoptosis within the flap.
Collapse
Affiliation(s)
- Baolong Li
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Zhengtai Chen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Xiaobin Luo
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Chenxi Zhang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Hongyu Chen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Shuxuan Wang
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mengyao Zhao
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Haiwei Ma
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Junling Liu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Mengshi Cheng
- Respiratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,The First Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Yanyan Yang
- Infectious Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,The First Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Hede Yan
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
| |
Collapse
|
7
|
Lyu J, Sun Y, Li X, Ma H. MicroRNA-206 inhibits the proliferation, migration and invasion of colorectal cancer cells by regulating the c-Met/AKT/GSK-3β pathway. Oncol Lett 2020; 21:147. [PMID: 33633805 PMCID: PMC7877959 DOI: 10.3892/ol.2020.12408] [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: 06/24/2020] [Accepted: 11/13/2020] [Indexed: 12/16/2022] Open
Abstract
An imbalance in microRNA (miRNA/miR) expression is closely associated with tumorigenesis and progression. miR-206 is downregulated in different types of tumors, including colorectal cancer (CRC). However, the effects of miR-206 on the progression of CRC, and its underlying molecular mechanisms are yet to be elucidated. The present study aimed to investigate the effects of miR-206 on the proliferation, migration and invasion of colorectal cancer cells, and determine its potential molecular mechanism. The results of the present study demonstrated that the expression levels of miR-206 and c-Met were affected in HCT116 and SW480 cells by transfected with miR-206 mimic, inhibitor or small interfering RNA-c-Met. A Dual-luciferase reporter assay was performed to identify the miRNA targets. Cell proliferation, migration and invasion assays were also performed. The results demonstrated that overexpression of miR-206 significantly decreased the viability of HCT116 and SW480 cells. The results of the Transwell assay indicated that the cell migratory and invasive abilities were inhibited following transfection with miR-206 mimic. As a target of miR-206, knockdown of c-Met significantly suppressed cell viability, migration and invasion. In addition, c-Met knockdown or overexpression of miR-206 inhibited activation of the AKT/GSK-3β pathway. Collectively, these results suggest that miR-206 suppresses the proliferation, migration and invasion of CRC cells by targeting the c-Met/AKT/GSK-3β pathway.
Collapse
Affiliation(s)
- Jiayu Lyu
- Department of First General Surgery, The Fifth Hospital of Harbin, Harbin, Heilongjiang 150040, P.R. China
| | - Yao Sun
- Department of Neurology, General Hospital of Heilongjiang Province Land Reclamation Bureau, Harbin, Heilongjiang 150088, P.R. China
| | - Xizhi Li
- Department of Neurology, Binzhou Medical University Hospital, Binzhou, Shandong 256603, P.R. China
| | - Huili Ma
- Department of Emergency Surgical Trauma Center, Binzhou Medical University Hospital, Binzhou, Shandong 256603, P.R. China
| |
Collapse
|
8
|
Hong W, Ying H, Lin F, Ding R, Wang W, Zhang M. lncRNA LINC00460 Silencing Represses EMT in Colon Cancer through Downregulation of ANXA2 via Upregulating miR-433-3p. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 19:1209-1218. [PMID: 32069703 PMCID: PMC7019044 DOI: 10.1016/j.omtn.2019.12.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 11/29/2019] [Accepted: 12/09/2019] [Indexed: 12/17/2022]
Abstract
Colon cancer (CC), one of the major causes of tumor-associated death, is often presented with a heterogenic pool of cells with unique differentiation patterns. This study explored the functions that LINC00460 displayed in CC by regulating microRNA-433-3p (miR-433-3p) and Annexin A2 (ANXA2). LINC00460 expression was either silenced or overexpressed in HCT-116 and LOVO cells to explore the functional roles of LINC00460 in CC. The relationship between miR-433-3p and LINC00460/ANXA2 was analyzed using dual-luciferase reporter assay, RNA-pull down, and RNA immunoprecipitation (RIP) assays. Cell proliferation, metastasis, invasion, and apoptosis were examined in vitro, and tumorigenicity was evaluated in vivo following LINC00460 silencing. Additionally, the regulatory mechanisms were investigated using LINC00460 and ANXA2 gain- or loss-of-function experiments. We found that LINC00460 was expressed highly in CC. Downregulation of LINC00460 inhibited cell invasion and proliferation in vitro and restrained tumor growth in vivo. Moreover, LINC00460 was able to specifically bind to miR-433-3p to increase the expression of ANXA2. Furthermore, LINC00460 downregulated the E-cadherin expression and upregulated the vimentin and N-cadherin expression by upregulating ANXA2, therefore inducing epithelial-mesenchymal transition. These findings suggested that LINC00460 might function as an oncogenic long non-coding RNA (lncRNA) in CC development and could be explored as a potential biomarker and therapeutic target for CC.
Collapse
Affiliation(s)
- Weiwen Hong
- Department of Anus & Intestine Surgery, Taizhou First People's Hospital, Taizhou 318020, P.R. China
| | - Hongan Ying
- General Department, Taizhou First People's Hospital, Taizhou 318020, P.R. China
| | - Feng Lin
- Department of General Surgery, Taizhou First People's Hospital, Taizhou 318020, P.R. China
| | - Ruliang Ding
- Department of Anus & Intestine Surgery, Taizhou First People's Hospital, Taizhou 318020, P.R. China
| | - Weiya Wang
- Department of Anus & Intestine Surgery, Taizhou First People's Hospital, Taizhou 318020, P.R. China
| | - Meng Zhang
- Department of General Surgery, Taizhou First People's Hospital, Taizhou 318020, P.R. China.
| |
Collapse
|
9
|
Yin W, Chen J, Wang G, Zhang D. MicroRNA‑106b functions as an oncogene and regulates tumor viability and metastasis by targeting LARP4B in prostate cancer. Mol Med Rep 2019; 20:951-958. [PMID: 31173237 PMCID: PMC6625195 DOI: 10.3892/mmr.2019.10343] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 03/07/2019] [Indexed: 01/10/2023] Open
Abstract
Prostate cancer (PCa) is the most common malignancy among males worldwide, and is one of the leading causes of cancer‑related mortality. MicroRNAs (miRs) are a type of endogenous, noncoding RNA that serve a key role in pathological processes, and have been demonstrated to be involved in the formation and progression of PCa. Previous studies have reported that miR‑106b acts as an oncogene; however, the specific effects of miR‑106b on PCa have not been fully elucidated. The present study aimed to investigate the role and underlying molecular mechanisms of miR‑106b in the initiation and progression of PCa. In this study, miR‑106b was reported to be overexpressed and la‑related protein 4B (LARP4B) was downregulated in PCa tissues compared with paracancerous tissues. In addition, LARP4B was identified as a target gene of miR‑106b by bioinformatics prediction analysis and a dual luciferase reporter gene assay. Furthermore, MTT, wound healing and Transwell assays were performed to evaluate PCa cell viability, and migration and invasive abilities. The data revealed that inhibition of miR‑106b significantly suppressed the viability, migration and invasion of PCa cells. In addition, inhibition of miR‑106b significantly suppressed the mRNA and protein expression of cancer‑related genes, including matrix metalloproteinase‑2, cluster of differentiation 44 and Ki‑67, and increased that of the tumor suppressor, mothers against decapentaplegic homolog 2. Collectively, the findings of the present study indicated that miR‑106b may target LAR4B to inhibit cancer cell viability, migration and invasion, and may be considered as a novel therapeutic target in PCa.
Collapse
Affiliation(s)
- Weiqi Yin
- Department of Urology, Ningbo First Hospital, Ningbo, Zhejiang 315010, P.R. China
| | - Junfeng Chen
- Department of Urology, Ningbo First Hospital, Ningbo, Zhejiang 315010, P.R. China
| | - Guoyao Wang
- Department of Urology, Ningbo First Hospital, Ningbo, Zhejiang 315010, P.R. China
| | - Dongxu Zhang
- Department of Urology, Ningbo First Hospital, Ningbo, Zhejiang 315010, P.R. China
| |
Collapse
|
10
|
ANXA2 Silencing Inhibits Proliferation, Invasion, and Migration in Gastric Cancer Cells. JOURNAL OF ONCOLOGY 2019; 2019:4035460. [PMID: 31186633 PMCID: PMC6521490 DOI: 10.1155/2019/4035460] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 03/15/2019] [Accepted: 03/28/2019] [Indexed: 12/14/2022]
Abstract
Annexin A2 (ANXA2) has been well known to associate with the progress of malignant tumor. However, the biological behavior of ANXA2 in gastric cancer (GC) remains unclear. We made a hypothesis in transcriptome level from TCGA datasets. Then, we used immunohistochemical staining to quantify the expression level of ANXA2 protein in GC tissues compared with adjacent tissues. Quantitative real-time PCR and western blot were used for analyzing ANXA2 expression in human GC (SGC-7901, MKN-45, BGC-823, and AGS) cell lines. We investigated the effect of a lentivirus-mediated knock-down of ANXA2 on the proliferation, invasion and migration of gastric cancer AGS cells. Cell proliferation was examined by MTT and colony formation tests. Cell apoptosis and cycle were measured by flow cytometry. Migration and invasion were detected by transwell assay. We found that high expression of ANXA2 can increase the mobility of cancer cells from TCGA datasets. ANXA2 was upregulated in GC tissues compared with adjacent tissues. AGS cell line displayed significantly higher expression of ANXA2 among the four GC cell lines. In addition, ANXA2 silencing led to a weakened ability of proliferation, invasion, and migration in GC cells; targeting of ANXA2 may be a potential therapeutic strategy for GC patients.
Collapse
|