1
|
Wang NN, Song Y, Yan X, Liu X, Wu R, Cao M, Li C. Regulatory roles of miRNA-530 in the post-transcriptional regulation of NF-κB signaling pathway through targeted modulation of IκBα in Sebastesschlegelii. FISH & SHELLFISH IMMUNOLOGY 2024; 149:109604. [PMID: 38710343 DOI: 10.1016/j.fsi.2024.109604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 04/17/2024] [Accepted: 05/02/2024] [Indexed: 05/08/2024]
Abstract
MicroRNAs (miRNAs) are a crucial type of non-coding RNAs involved in post-transcriptional regulation. The playing essential regulatory roles in the NF-κB signaling pathway and modulate the host immune response to diverse pathogens by targeting IκBα. However, the regulatory mechanism of miRNAs in relation with IκBα in Sebastes schlegelii remains unclear. In our study, we identified two copies of IkBα gene in black rockfish (Sebastes schlegelii), namely IkBα1 and IkBα2. Moreover, we have discovered that miRNA-530 can activate the NF-κB signaling pathway by inhibiting the expression of IκBα, thereby inducing the inflammatory response. This project comprehensively investigated the interactive regulatory roles of miRNA-530 in the NF-κB signaling pathway at both cellular and in vivo levels, while also elucidating the regulatory relationships between miRNA-530 and IκBα. In conclusion, our research confirmed that miRNA-530 can target the 3'UTR region of IκBα, resulting in a decrease in the expression of IκBα at the post-transcriptional level and inhibiting its translation. The findings contribute to the understanding of the regulatory network of non-coding RNA in teleosts and its subsequent regulation of the NF-κB signaling pathway by miRNAs.
Collapse
Affiliation(s)
- Ning Ning Wang
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China
| | - Yize Song
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China
| | - Xu Yan
- College of Life Sciences, Qingdao University, Qingdao, 266071, China
| | - Xiantong Liu
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China
| | - Ruixue Wu
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China
| | - Min Cao
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China
| | - Chao Li
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China.
| |
Collapse
|
2
|
Jin J, Lin J, Xu A, Lou J, Qian C, Li X, Wang Y, Yu W, Tao H. CCL2: An Important Mediator Between Tumor Cells and Host Cells in Tumor Microenvironment. Front Oncol 2021; 11:722916. [PMID: 34386431 PMCID: PMC8354025 DOI: 10.3389/fonc.2021.722916] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 07/09/2021] [Indexed: 12/12/2022] Open
Abstract
Tumor microenvironment (TME) formation is a major cause of immunosuppression. The TME consists of a considerable number of macrophages and stromal cells that have been identified in multiple tumor types. CCL2 is the strongest chemoattractant involved in macrophage recruitment and a powerful initiator of inflammation. Evidence indicates that CCL2 can attract other host cells in the TME and direct their differentiation in cooperation with other cytokines. Overall, CCL2 has an unfavorable effect on prognosis in tumor patients because of the accumulation of immunosuppressive cell subtypes. However, there is also evidence demonstrating that CCL2 enhances the anti-tumor capability of specific cell types such as inflammatory monocytes and neutrophils. The inflammation state of the tumor seems to have a bi-lateral role in tumor progression. Here, we review works focusing on the interactions between cancer cells and host cells, and on the biological role of CCL2 in these processes.
Collapse
Affiliation(s)
- Jiakang Jin
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Jinti Lin
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Ankai Xu
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Jianan Lou
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Chao Qian
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Xiumao Li
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Yitian Wang
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Wei Yu
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Huimin Tao
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
3
|
Song C, Wang X, Zhao X, Ai J, Qi Y, Chen A. MicroRNA-325-3p contributes to colorectal carcinoma by targeting cytokeratin 18. Oncol Lett 2021; 21:248. [PMID: 33664812 PMCID: PMC7882876 DOI: 10.3892/ol.2021.12509] [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: 08/31/2020] [Accepted: 12/23/2020] [Indexed: 12/12/2022] Open
Abstract
Colorectal carcinoma (CRC) is one of the most common malignant tumors. The present study aimed to investigate a non-invasive molecular marker that can evaluate the diagnosis and potential molecular mechanism of CRC. Microarray assays and reverse transcription-quantitative PCR analysis demonstrated that microRNA (miR)-325-3p expression was significantly increased in both tissues and serum samples of patients with CRC. In addition, miR-325-3p expression in the tissues and serum was significantly associated with differentiation, TNM stage and lymph node metastasis. The results of the dual-luciferase reporter assay and western blot analysis revealed that cytokeratin 18 (CK18) is a target gene of miR-325-3p. Furthermore, treatment with transforming growth factor (TGF)-β increased miR-325-3p expression in a time-dependent manner. Conversely, TGF-β decreased CK18 expression at 48 and 72 h. Western blot analysis demonstrated that TGF-β1 significantly decreased the expression of the epithelial marker, CK18, and increased the expression of the mesenchymal markers, α-SMA and vimentin. Notably, these effects were reversed following inhibition of miR-325-3p expression. Taken together, the results of the present study suggest that miR-325-3p is a key regulator of TGF-β-induced CK18 downregulation. Thus, elevated levels of miR-325-3p is an important factor affecting epithelial-to-mesenchymal transition, and is likely to be a molecular marker in the progression of CRC and act as a potential therapeutic target.
Collapse
Affiliation(s)
- Chuanfang Song
- Department of Gastroenterology, Hongqi Hospital Affiliated to Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Xiujie Wang
- Department of Gastroenterology, Hongqi Hospital Affiliated to Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Xinxin Zhao
- Department of Gastroenterology, Hongqi Hospital Affiliated to Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Jiang Ai
- Department of Gastroenterology, Hongqi Hospital Affiliated to Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Yixuan Qi
- Department of Gastroenterology, Hongqi Hospital Affiliated to Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Aidong Chen
- Department of Gastroenterology, Hongqi Hospital Affiliated to Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| |
Collapse
|
4
|
Cheng J, Yang A, Cheng S, Feng L, Wu X, Lu X, Zu M, Cui J, Yu H, Zou L. Circulating miR-19a-3p and miR-483-5p as Novel Diagnostic Biomarkers for the Early Diagnosis of Gastric Cancer. Med Sci Monit 2020; 26:e923444. [PMID: 32487978 PMCID: PMC7297033 DOI: 10.12659/msm.923444] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Background MicroRNAs (miRNAs) are attracting substantial interest as promising noninvasive biomarkers for gastric cancer (GC). Our study aimed to identify circulating miRNAs that are potential noninvasive markers for precancerous lesions and early gastric cancers (EGCs). Material/Methods Plasma specimens were obtained from 58 gastritis subjects, 54 patients with precancerous lesions, and 38 EGC patients for study. Results Significant differences in the plasma expression levels of miR-19a-3p, miR-22-3p, miR-146a-5p, and miR-483-5p (all P<0.05) were observed between EGC patients and gastritis subjects. Multivariable analysis showed that age (OR, 1.054; 95% CI, 1.006–1.104), miR-19a-3p expression (OR, 3.676; 95% CI, 1.914–7.061), and miR-483-5p expression (OR, 1.589; 95% CI, 1.242–2.033) were independently associated with EGCs and precancerous lesions. A combined diagnostic model incorporating these 3 variables for the prediction of EGCs and precancerous lesions was derived. The area under the receiver operating characteristic curve (AUC) of the model was 0.84; the sensitivity was 87.7% and the specificity was 62.8% at the cutoff value of −0.08. Conclusions Plasma miR-19a-3p and miR-483-5p are promising and powerful noninvasive markers for the early detection of GC. Patients are more willing to undergo noninvasive diagnostic procedures than gastroscopy for cancer screening, economizing limited medical resources.
Collapse
Affiliation(s)
- Jieyao Cheng
- Department of Gastroenterology, Peking Union Medical College Hospital, Beijing, China (mainland)
| | - Aiming Yang
- Department of Gastroenterology, Peking Union Medical College Hospital, Beijing, China (mainland)
| | - Shujun Cheng
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (mainland)
| | - Lin Feng
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (mainland)
| | - Xi Wu
- Department of Gastroenterology, Peking Union Medical College Hospital, Beijing, China (mainland)
| | - Xinghua Lu
- Department of Gastroenterology, Peking Union Medical College Hospital, Beijing, China (mainland)
| | - Ming Zu
- Department of Gastroenterology, Peking Union Medical College Hospital, Beijing, China (mainland)
| | - Jianfang Cui
- Department of Gastroenterology, Peking Union Medical College Hospital, Beijing, China (mainland)
| | - Hang Yu
- Department of Gastroenterology, Peking Union Medical College Hospital, Beijing, China (mainland)
| | - Long Zou
- Department of Gastroenterology, Peking Union Medical College Hospital, Beijing, China (mainland)
| |
Collapse
|
5
|
Calabrese G, Dolcimascolo A, Caruso G, Forte S. miR-19a Is Involved In Progression And Malignancy Of Anaplastic Thyroid Cancer Cells. Onco Targets Ther 2019; 12:9571-9583. [PMID: 32009794 PMCID: PMC6859471 DOI: 10.2147/ott.s221733] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 08/13/2019] [Indexed: 12/15/2022] Open
Abstract
Background MicroRNAs (miRNAs) are endogenous, single-stranded, non-coding RNAs acting as negative regulators of gene expression involved in a number of physiological processes. MiRNAs' expression is commonly dysregulated in many types of human tumor diseases and cancers, including thyroid cancers, and is often involved in tumor initiation and progression. miR-19a, a member of miR-17-92 cluster, has been demonstrated to promote cell growth in anaplastic thyroid cancer (ATC), the most advanced and aggressive thyroid cancer. Purpose In this work, we investigate the potential contribution of miR-19a in thyroid cancer cells poor prognosis and de-differentiation. Methods We directly modulated the expression of miR-19a in papillary (PTC) and anaplastic thyroid carcinoma cell lines through transfection of specific miR-19a mimic or inhibitor. Further, we performed gene expression analysis of specific genes to evaluate miR-19a association with cell cycle, differentiation, and poor prognosis. Results Our data indicate that miR-19a overexpression in PTC cells significantly promotes cell growth, decreases the expression of differentiation genes and activates poor prognosis genes. Its inhibition in ATC cells reduces cell proliferation and the expression of genes related to poor prognosis but does not affect differentiation. Conclusion Our findings reveal the existence of functional associations between miR-19a expression and thyroid cancer progression and malignancy suggesting miR-19a as a novel candidate therapeutic target for ATC.
Collapse
Affiliation(s)
- Giovanna Calabrese
- Department of Biomedical and Biotechnological Sciences, Physiology Section, University of Catania, Catania 95123, Italy
| | - Anna Dolcimascolo
- Department of Biomedical and Biotechnological Sciences, Physiology Section, University of Catania, Catania 95123, Italy
| | - Giuseppe Caruso
- Department of Laboratories, Oasi Research Institute, IRCCs, Troina, EN 94018, Italy
| | - Stefano Forte
- Molecular Biology Unit, IOM Ricerca, Viagrande, CT 95029, Italy
| |
Collapse
|
6
|
Efficient isolation, biophysical characterisation and molecular composition of extracellular vesicles secreted by primary and immortalised cells of reproductive origin. Theriogenology 2019; 135:121-137. [PMID: 31207473 DOI: 10.1016/j.theriogenology.2019.06.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 05/25/2019] [Accepted: 06/04/2019] [Indexed: 12/27/2022]
Abstract
Effective communication between the maternal reproductive tract, gametes and the pre-implantation embryo is essential for the successful establishment of pregnancy. Recent studies have recognised extracellular vesicles (EVs) as potent vehicles for intercellular communication, potentially via their transport of microRNAs (miRNAs). The aim of the current investigation was to determine the size, concentration and electrical surface properties (zeta potential) of EVs secreted by; (1) primary cultures of porcine oviductal epithelial cells (POECs) from the isthmus and ampullary regions of the female reproductive tract; (2) Ishikawa and RL95-2 human endometrial epithelial cell line cultures; and (3) the non-reproductive epithelial cell line HEK293T. In addition, this study investigated whether EVs secreted by POECs contained miRNAs. All cell types were cultured in EV-depleted medium for 24 or 48 h. EVs were successfully isolated from conditioned culture media using size exclusion chromatography. Nanoparticle tracking analysis (NTA) was performed to evaluate EV size, concentration and zeta potential. QRT-PCR was performed to quantify the expression of candidate miRNAs (miR-103, let-7a, miR-19a, miR-203, miR-126, miR-19b, RNU44, miR-92, miR-196a, miR-326 and miR-23a). NTA confirmed the presence of EVs with diameters of 50-150 nm in all cell types. EV size distribution was significantly different between cell types after 24 and 48 h of cell culture and the concentration of EVs secreted by POECs and Ishikawa cells was also time dependent. The distribution of EVs with specific electrokinetic potential measurements varied between cell types, indicating that EVs of differing cellular origin have varied membrane components. In addition, EVs secreted by POECs exhibited significantly different time dependant changes in zeta potential. QRT-PCR confirmed the presence of miR-103, let-7a, miR-19a, miR-203, miR-126, and miR-19b in EVs secreted by POECs (CT ≥ 29). Bioinformatics analysis suggests that these miRNAs are involved in cell proliferation, innate immune responses, apoptosis and cellular migration. In conclusion, reproductive epithelial cells secrete distinct populations of EVs containing miRNAs, which potentially act in intercellular communication in order to modulate the periconception events leading to successful establishment of pregnancy.
Collapse
|
7
|
Wang H, Hu Z, Chen L. Enhanced plasma miR-26a-5p promotes the progression of bladder cancer via targeting PTEN. Oncol Lett 2018; 16:4223-4228. [PMID: 30197668 PMCID: PMC6126335 DOI: 10.3892/ol.2018.9163] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 05/22/2018] [Indexed: 12/17/2022] Open
Abstract
The current study aimed to evaluate the expression and specific role of miR-26a-5p in the progression of bladder cancer (BC). Reverse transcription-quantitative polymerase chain reaction analysis was performed to evaluate the level of miR-26a-5p in BC cancer and healthy controls. The present data showed that plasma miR-26a-5p was significantly increased in BC patients. Furthermore, BC tissues exhibited greater levels of miR-26a-5p compared with adjacent non-neoplastic tissues-26a-5p. Compared with BC patients at Ta-T1 stage, the level of miR-26a-5p was significantly elevated in BC patients ≥T2. BC patients at G3 stage demonstrated a higher plasma miR-26a-5p level compared with those at G1/2 stage. Receiver operating characteristic (ROC) analysis indicated that miR-26a-5p could differentiate BC patients from controls. Additionally, Kaplan-Meier analysis demonstrated that plasma miR-26a-5p negatively correlated with survival of BC patients. Dual luciferase reporter assay indicated that miR-26a-5p significantly suppressed the relative luciferase activity of pmirGLO-PTEN-3′UTR compared with the control. In conclusion, the current study indicated novel data that the levels of plasma miR-26a-5p was significantly increased in BC patients. Furthermore, the present study suggested that determination of plasma miR-26a-5p level could help to distinguish BC patients from healthy controls via targeting PTEN.
Collapse
Affiliation(s)
- Hui Wang
- Department of Nephrology, Qilu Hospital, Shandong University, Jinan, Shandong 250014, P.R. China.,Department of Nephrology, The Fourth Hospital of Jinan City, Jinan, Shandong 250031, P.R. China
| | - Zhao Hu
- Department of Nephrology, Qilu Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Li Chen
- Department of Endocrinology, Qilu Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| |
Collapse
|
8
|
Deng Y, Zhou X, Xiang X, Ou Y, He J. Effect of miRNA-19a on gastrointestinal motility in rats with functional dyspepsia. Exp Ther Med 2018; 15:4875-4879. [PMID: 29805508 PMCID: PMC5952082 DOI: 10.3892/etm.2018.6009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Accepted: 02/01/2018] [Indexed: 12/17/2022] Open
Abstract
The effect of microRNA (miRNA)-19a on gastrointestinal motility in rats with functional dyspepsia was investigated. Fifty adult Sprague-Dawley (SD) rats were randomly divided into 5 groups, 10 rats in each group, one group as the normal group, one group as the model group, and the other three groups were divided into negative control group, miRNA-19a mimic group and miRNA-19a inhibitor group. All rats were intraperitoneally injected with miRNA-19a scramble, miRNA-19a mimic and miRNA-19a inhibitor. Except the normal group, the functional dyspepsia model rat was established by proper clipping tail stimulation. The gastric emptying rate, intestinal propulsive ratio, serum motilin and vasoactive intestinal peptide of rats in each group were measured. The level of miRNA-19a expression in each group was detected by reverse transcription-polymerase chain reaction (RT-PCR). The gastric emptying rate, intestinal propulsive ratio and serum motilin in model group were significantly lower than those in normal group, and vasoactive intestinal peptide was higher in model group than that in normal group (P<0.05). The expression of miRNA-19a in model group was significantly higher than that in normal group (P<0.05). After intraperitoneal injection of miRNA-19a mimic, the expression of miRNA-19a was increased; gastric emptying rate, intestinal propulsive ratio and serum motilin were significantly reduced in model group, and vasoactive intestinal peptide was increased (P<0.05). After intraperitoneal injection of miRNA-19a inhibitor, the expression of miRNA-19a was remarkably decreased; gastric emptying rate, intestinal propulsive ratio and serum motilin were further increased in model group, and vasoactive intestinal peptide was decreased (P<0.05). In conclusion, the expression of miRNA-19a in rats with functional dyspepsia is higher than that in normal rats, and the reduced miRNA-19a expression can ameliorate the gastrointestinal motility in rats with functional dyspepsia.
Collapse
Affiliation(s)
- Yinzhi Deng
- Department of Gastroenterology, The Central Hospital of Enshi Autonomous Prefecture, Enshi, Hubei 445000, P.R. China
| | - Xiangyu Zhou
- Department of Gastroenterology, The Central Hospital of Enshi Autonomous Prefecture, Enshi, Hubei 445000, P.R. China
| | - Xingchao Xiang
- Department of Gastroenterology, The Central Hospital of Enshi Autonomous Prefecture, Enshi, Hubei 445000, P.R. China
| | - Yangli Ou
- Department of Gastroenterology, The Central Hospital of Enshi Autonomous Prefecture, Enshi, Hubei 445000, P.R. China
| | - Jianhua He
- Department of Gastroenterology, The Central Hospital of Enshi Autonomous Prefecture, Enshi, Hubei 445000, P.R. China
| |
Collapse
|
9
|
He L, Qu L, Wei L, Chen Y, Suo J. Reduction of miR‑132‑3p contributes to gastric cancer proliferation by targeting MUC13. Mol Med Rep 2017; 15:3055-3061. [PMID: 28339011 PMCID: PMC5428394 DOI: 10.3892/mmr.2017.6347] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 01/16/2017] [Indexed: 02/07/2023] Open
Abstract
Abnormal expression of epidermal growth factor receptor (EGFR) signaling and microRNAs (miRNAs) has been widely seen in gastric cancer. The present study focused on the miRNAs that regulate human epidermal growth factor receptor (HER) activation through mucin 13 (MUC13). The protein level of MUC13 was demonstrated to be significantly increased in gastric cancer tissues compared with normal tissues by western blot analysis and immunohistochemistry. TargetScan bioinformatic predictions indicated that miRNA (miR)-212-3p and miR-132-3p may bind to the 3′-untranslated region of MUC13. Further investigation revealed that miR-132-3p was significantly decreased in gastric cancer tissues compared with normal tissues, whereas miR-212-3p expression was unaffected. Luciferase assays and western blot confirmed MUC13 as a target gene of miR-132-3p. Inhibition of miR-132-3p enhanced gastric cancer cell migration through activation of HER2, extracellular signal-regulated kinase (ERK) and Akt serine/threonine kinase (Akt) signaling, which was a similar effect to that of MUC13 overexpression. In summary, reduction of miR-132-3p may contribute to gastric cancer proliferation by targeting MUC13.
Collapse
Affiliation(s)
- Liang He
- Department of Gastrointestinal Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Linlin Qu
- Department of Laboratory Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Lijing Wei
- Department of Laboratory Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yan Chen
- Department of Gastrointestinal Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Jian Suo
- Department of Gastrointestinal Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
10
|
Ma Q, Peng Z, Wang L, Li Y, Wang K, Zheng J, Liang Z, Liu T. miR-19a correlates with poor prognosis of clear cell renal cell carcinoma patients via promoting cell proliferation and suppressing PTEN/SMAD4 expression. Int J Oncol 2016; 49:2589-2599. [DOI: 10.3892/ijo.2016.3746] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 10/10/2016] [Indexed: 11/06/2022] Open
|
11
|
MiR-19a promotes cell proliferation and invasion by targeting RhoB in human glioma cells. Neurosci Lett 2016; 628:161-6. [PMID: 27329239 DOI: 10.1016/j.neulet.2016.06.031] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Revised: 06/06/2016] [Accepted: 06/17/2016] [Indexed: 01/30/2023]
Abstract
MicroRNA-19a (miR-19a) is upregulated in different types of cancers, including gliomas, but its specific role and function in gliomas have yet to be fully elucidated. In this study, we found that miR-19a was significantly upregulated in human glioma tissues and cell lines. Overexpression of miR-19a by a miR-19a mimic promoted glioma cell proliferation and invasion. In contrast, miR-19a inhibitor suppressed cell proliferation and invasion. Furthermore, by a dual-luciferase reporter assay and expression analysis, we determined that Ras homolog family member B was a direct target of miR-19a. Knockdown of Ras homolog family member B could block cell proliferation and invasion induced by the miR-19a mimic. In conclusion, our study demonstrated that miR-19a upregulation is common in gliomas and that suppression of miR-19a expression inhibits cell proliferation and invasion, which indicates that miR-19a may act as an oncogene in gliomas.
Collapse
|
12
|
He G, Li LI, Guan E, Chen J, Qin YI, Xie Y. Fentanyl inhibits the progression of human gastric carcinoma MGC-803 cells by modulating NF-κB-dependent gene expression in vivo. Oncol Lett 2016; 12:563-571. [PMID: 27347181 DOI: 10.3892/ol.2016.4619] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 03/24/2016] [Indexed: 12/13/2022] Open
Abstract
Fentanyl is widely used to treat acute and chronic pain. Previous in vitro studies by the present authors demonstrated that fentanyl inhibits the progression of the MGC-803 human gastric carcinoma cell line by affecting apoptosis-related genes, including nuclear factor-kappa B (NF-κB) and phosphatase and tensin homolog. In the present study, the effects of fentanyl on NF-κB-dependent gene expression were investigated in vivo. Nude mice were inoculated with an MGC-803 cell suspension, and mice that developed subcutaneous tumors measuring >1.0×1.0 cm were selected for study. Mice were administered intraperitoneal injections of fentanyl (0.05 mg/kg, group F1; 0.1 mg/kg, group F2; 0.2 mg/kg, group F3; and 0.4 mg/kg, group F4) for 14 consecutive days. Non-fentanyl-treated mice (group C) and normal saline-treated mice (group N) served as the control groups. Tumor growth was monitored by calculating the time-shift of the growth curve. Morphological changes were also observed using microscopy. The expression of NF-κB, B-cell lymphoma-2 (Bcl-2), B-cell associated X protein (Bax), vascular endothelial growth factor-A (VEGF-A) and matrix metalloproteinase-9 (MMP-9) in the subcutaneous tumor tissue was also analyzed by reverse transcription-polymerase chain reaction and western blot analysis, and confirmed using immunohistochemistry. The relative tumor volumes of groups F1, F2, F3 and F4 were significantly reduced compared with groups C and N. Furthermore, subcutaneous tumor cells exhibited nuclear swelling, chromatin condensation, reduced chromatin and nuclear fragmentation in the F1, F2, F3 and F4 groups. The number of NF-κB+, Bcl-2+, VEGF-A+ and MMP-9+ subcutaneous tumor cells was reduced, whereas the number of Bax+ cells was increased in the F1, F2, F3 and F4 groups. Additionally, in these groups, tumor expression of NF-κB, Bcl-2, VEGF-A and MMP-9 transcripts and proteins was downregulated, while Bax messenger RNA and protein expression levels were upregulated. The results revealed that fentanyl inhibits the growth of subcutaneous human gastric carcinoma tumors in mice. Therefore, it could be hypothesized that this antineoplastic activity may result from the inhibition of NF-κB activation, suppression of downstream VEGF-A and MMP-9 expression, and normalization of the pro-apoptotic Bax/Bcl-2 ratio.
Collapse
Affiliation(s)
- Guodong He
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China; Department of Anesthesiology, The First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - L I Li
- Department of Anesthesiology, The First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Enjian Guan
- Department of Anesthesiology, The First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Jing Chen
- Department of Anesthesiology, The First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Y I Qin
- Department of Anesthesiology, The First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Yubo Xie
- Department of Anesthesiology, The First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
13
|
Song B, Shu Y, Cui T, Fu P. Allicin inhibits human renal clear cell carcinoma progression via suppressing HIF pathway. Int J Clin Exp Med 2015; 8:20573-20580. [PMID: 26884975 PMCID: PMC4723820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 11/01/2015] [Indexed: 06/05/2023]
Abstract
Hypoxia-inducible factor 1-alpha (HIF-1α) protects hypoxic cells from apoptosis or necrosis under ischemic and anoxic conditions. Allicin is characterized by the anti-cancer characteristics. This study aims to explore whether allicin is involved in renal clear cell carcinoma progression through HIF-1α. A total of 40 RCC tissues and 39 normal renal tissues were collected H&E and immunohistochemistry were applied to study morphology changes. MTT assay and flow cytometry (FCM) were used to analyze cell viability and apoptosis. In vitro colony formation assay and wound healing assay were conducted to explore cell migration. The protein levels of Bcl-2, VEGF and HIF-1α were increased in RCC tissues. More importantly, treatment with allicin significantly decreased HIF-1α protein level, thereby reducing Bcl-2 and VEGF expression. In addition, allicin also obviously enhanced apoptotic cells. And colony formation capacity and cell migration rate were reduced in RCC-9863 cells treated with allicin. Further study revealed that overexpression of HIF-1α could partially repress allicin-induced downstream effects. To conclude, allicin inhibits human renal clear cell carcinoma progression partially by suppressing HIF pathway.
Collapse
Affiliation(s)
- Bin Song
- Department of Nephrology, The People’s Hospital of DeyangDeyang, 618000, China
| | - Ying Shu
- Department of Nephrology, Division of Internal Medicine, West China Hospital of Sichuan UniversityChengdu, 610041, China
| | - Tianlei Cui
- Department of Nephrology, Division of Internal Medicine, West China Hospital of Sichuan UniversityChengdu, 610041, China
| | - Ping Fu
- Department of Nephrology, Division of Internal Medicine, West China Hospital of Sichuan UniversityChengdu, 610041, China
| |
Collapse
|
14
|
Akhtar S, Hartmann P, Karshovska E, Rinderknecht FA, Subramanian P, Gremse F, Grommes J, Jacobs M, Kiessling F, Weber C, Steffens S, Schober A. Endothelial Hypoxia-Inducible Factor-1α Promotes Atherosclerosis and Monocyte Recruitment by Upregulating MicroRNA-19a. Hypertension 2015; 66:1220-6. [PMID: 26483345 DOI: 10.1161/hypertensionaha.115.05886] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 09/25/2015] [Indexed: 12/12/2022]
Abstract
Chemokines mediate monocyte adhesion to dysfunctional endothelial cells (ECs) and promote arterial inflammation during atherosclerosis. Hypoxia-inducible factor (HIF)-1α is expressed in various cell types of atherosclerotic lesions and is associated with lesional inflammation. However, the impact of endothelial HIF-1α in atherosclerosis is unclear. HIF-1α was detectable in the nucleus of ECs covering murine and human atherosclerotic lesions. To study the role of endothelial HIF-1α in atherosclerosis, deletion of the Hif1a gene was induced in ECs from apolipoprotein E knockout mice (EC-Hif1a(-/-)) by Tamoxifen injection. The formation of atherosclerotic lesions, the lesional macrophage accumulation, and the expression of CXCL1 in ECs were reduced after partial carotid ligation in EC-Hif1a(-/-) compared with control mice. Moreover, the lesion area and the lesional macrophage accumulation were decreased in the aortas of EC-Hif1a(-/-) mice compared with control mice during diet-induced atherosclerosis. In vitro, mildly oxidized low-density lipoprotein or lysophosphatidic acid 20:4 increased endothelial CXCL1 expression and monocyte adhesion by inducing HIF-1α expression. Moreover, endothelial Hif1a deficiency resulted in downregulation of miR-19a in atherosclerotic arteries determined by microRNA profiling. In vitro, HIF-1α-induced miR-19a expression mediated the upregulation of CXCL1 in mildly oxidized low-density lipoprotein-stimulated ECs. These results indicate that hyperlipidemia upregulates HIF-1α expression in ECs by mildly oxidized low-density lipoprotein-derived unsaturated lysophosphatidic acid. Endothelial HIF-1α promoted atherosclerosis by triggering miR-19a-mediated CXCL1 expression and monocyte adhesion, indicating that inhibition of the endothelial HIF-1α/miR-19a pathway may be a therapeutic option against atherosclerosis.
Collapse
Affiliation(s)
- Shamima Akhtar
- From the Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany (S.A., P.H., E.K., F.-A.R., P.S., C.W., S.S., A.S.); Institute for Molecular Cardiovascular Research (S.A., A.S.), Department of Experimental Molecular Imaging (F.G., F.K.), and European Vascular Center Aachen-Maastricht (J.G., M.J.), RWTH Aachen University, Aachen, Germany; European Vascular Center Aachen-Maastricht, University Maastricht Medical Center, Maastricht, The Netherlands (J.G., M.J.); Cardiovascular Research Institute Maastricht, University Maastricht, Maastricht, The Netherlands (C.W.); and DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany (C.W., S.S., A.S.)
| | - Petra Hartmann
- From the Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany (S.A., P.H., E.K., F.-A.R., P.S., C.W., S.S., A.S.); Institute for Molecular Cardiovascular Research (S.A., A.S.), Department of Experimental Molecular Imaging (F.G., F.K.), and European Vascular Center Aachen-Maastricht (J.G., M.J.), RWTH Aachen University, Aachen, Germany; European Vascular Center Aachen-Maastricht, University Maastricht Medical Center, Maastricht, The Netherlands (J.G., M.J.); Cardiovascular Research Institute Maastricht, University Maastricht, Maastricht, The Netherlands (C.W.); and DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany (C.W., S.S., A.S.)
| | - Ela Karshovska
- From the Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany (S.A., P.H., E.K., F.-A.R., P.S., C.W., S.S., A.S.); Institute for Molecular Cardiovascular Research (S.A., A.S.), Department of Experimental Molecular Imaging (F.G., F.K.), and European Vascular Center Aachen-Maastricht (J.G., M.J.), RWTH Aachen University, Aachen, Germany; European Vascular Center Aachen-Maastricht, University Maastricht Medical Center, Maastricht, The Netherlands (J.G., M.J.); Cardiovascular Research Institute Maastricht, University Maastricht, Maastricht, The Netherlands (C.W.); and DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany (C.W., S.S., A.S.)
| | - Fatuma-Ayaan Rinderknecht
- From the Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany (S.A., P.H., E.K., F.-A.R., P.S., C.W., S.S., A.S.); Institute for Molecular Cardiovascular Research (S.A., A.S.), Department of Experimental Molecular Imaging (F.G., F.K.), and European Vascular Center Aachen-Maastricht (J.G., M.J.), RWTH Aachen University, Aachen, Germany; European Vascular Center Aachen-Maastricht, University Maastricht Medical Center, Maastricht, The Netherlands (J.G., M.J.); Cardiovascular Research Institute Maastricht, University Maastricht, Maastricht, The Netherlands (C.W.); and DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany (C.W., S.S., A.S.)
| | - Pallavi Subramanian
- From the Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany (S.A., P.H., E.K., F.-A.R., P.S., C.W., S.S., A.S.); Institute for Molecular Cardiovascular Research (S.A., A.S.), Department of Experimental Molecular Imaging (F.G., F.K.), and European Vascular Center Aachen-Maastricht (J.G., M.J.), RWTH Aachen University, Aachen, Germany; European Vascular Center Aachen-Maastricht, University Maastricht Medical Center, Maastricht, The Netherlands (J.G., M.J.); Cardiovascular Research Institute Maastricht, University Maastricht, Maastricht, The Netherlands (C.W.); and DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany (C.W., S.S., A.S.)
| | - Felix Gremse
- From the Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany (S.A., P.H., E.K., F.-A.R., P.S., C.W., S.S., A.S.); Institute for Molecular Cardiovascular Research (S.A., A.S.), Department of Experimental Molecular Imaging (F.G., F.K.), and European Vascular Center Aachen-Maastricht (J.G., M.J.), RWTH Aachen University, Aachen, Germany; European Vascular Center Aachen-Maastricht, University Maastricht Medical Center, Maastricht, The Netherlands (J.G., M.J.); Cardiovascular Research Institute Maastricht, University Maastricht, Maastricht, The Netherlands (C.W.); and DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany (C.W., S.S., A.S.)
| | - Jochen Grommes
- From the Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany (S.A., P.H., E.K., F.-A.R., P.S., C.W., S.S., A.S.); Institute for Molecular Cardiovascular Research (S.A., A.S.), Department of Experimental Molecular Imaging (F.G., F.K.), and European Vascular Center Aachen-Maastricht (J.G., M.J.), RWTH Aachen University, Aachen, Germany; European Vascular Center Aachen-Maastricht, University Maastricht Medical Center, Maastricht, The Netherlands (J.G., M.J.); Cardiovascular Research Institute Maastricht, University Maastricht, Maastricht, The Netherlands (C.W.); and DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany (C.W., S.S., A.S.)
| | - Michael Jacobs
- From the Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany (S.A., P.H., E.K., F.-A.R., P.S., C.W., S.S., A.S.); Institute for Molecular Cardiovascular Research (S.A., A.S.), Department of Experimental Molecular Imaging (F.G., F.K.), and European Vascular Center Aachen-Maastricht (J.G., M.J.), RWTH Aachen University, Aachen, Germany; European Vascular Center Aachen-Maastricht, University Maastricht Medical Center, Maastricht, The Netherlands (J.G., M.J.); Cardiovascular Research Institute Maastricht, University Maastricht, Maastricht, The Netherlands (C.W.); and DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany (C.W., S.S., A.S.)
| | - Fabian Kiessling
- From the Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany (S.A., P.H., E.K., F.-A.R., P.S., C.W., S.S., A.S.); Institute for Molecular Cardiovascular Research (S.A., A.S.), Department of Experimental Molecular Imaging (F.G., F.K.), and European Vascular Center Aachen-Maastricht (J.G., M.J.), RWTH Aachen University, Aachen, Germany; European Vascular Center Aachen-Maastricht, University Maastricht Medical Center, Maastricht, The Netherlands (J.G., M.J.); Cardiovascular Research Institute Maastricht, University Maastricht, Maastricht, The Netherlands (C.W.); and DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany (C.W., S.S., A.S.)
| | - Christian Weber
- From the Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany (S.A., P.H., E.K., F.-A.R., P.S., C.W., S.S., A.S.); Institute for Molecular Cardiovascular Research (S.A., A.S.), Department of Experimental Molecular Imaging (F.G., F.K.), and European Vascular Center Aachen-Maastricht (J.G., M.J.), RWTH Aachen University, Aachen, Germany; European Vascular Center Aachen-Maastricht, University Maastricht Medical Center, Maastricht, The Netherlands (J.G., M.J.); Cardiovascular Research Institute Maastricht, University Maastricht, Maastricht, The Netherlands (C.W.); and DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany (C.W., S.S., A.S.)
| | - Sabine Steffens
- From the Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany (S.A., P.H., E.K., F.-A.R., P.S., C.W., S.S., A.S.); Institute for Molecular Cardiovascular Research (S.A., A.S.), Department of Experimental Molecular Imaging (F.G., F.K.), and European Vascular Center Aachen-Maastricht (J.G., M.J.), RWTH Aachen University, Aachen, Germany; European Vascular Center Aachen-Maastricht, University Maastricht Medical Center, Maastricht, The Netherlands (J.G., M.J.); Cardiovascular Research Institute Maastricht, University Maastricht, Maastricht, The Netherlands (C.W.); and DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany (C.W., S.S., A.S.)
| | - Andreas Schober
- From the Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany (S.A., P.H., E.K., F.-A.R., P.S., C.W., S.S., A.S.); Institute for Molecular Cardiovascular Research (S.A., A.S.), Department of Experimental Molecular Imaging (F.G., F.K.), and European Vascular Center Aachen-Maastricht (J.G., M.J.), RWTH Aachen University, Aachen, Germany; European Vascular Center Aachen-Maastricht, University Maastricht Medical Center, Maastricht, The Netherlands (J.G., M.J.); Cardiovascular Research Institute Maastricht, University Maastricht, Maastricht, The Netherlands (C.W.); and DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany (C.W., S.S., A.S.).
| |
Collapse
|