1
|
Wang N, Li D, Zhang T, Pachai MR, Cho WH, Khudoynazarova MN, Schoeps DM, Bao Y, Liu M, Tang L, Yelena J, Chi P, Chen Y. Loss of Kmt2c / d promotes gastric cancer initiation and confers vulnerability to mTORC1 inhibition and anti-PD1 immunotherapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.27.645747. [PMID: 40236091 PMCID: PMC11996406 DOI: 10.1101/2025.03.27.645747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
KMT2C and KMT2D ( KMT2C/D ) are frequently mutated in gastric adenocarcinoma, yet their function in cancer initiation remains poorly understood. In this study, based on the observation that loss-of-function mutations of KMT2C and KMT2D are enriched and co-occur in gastric adenocarcinoma, we developed genetically engineered mouse models to selectively knock out Kmt2c and Kmt2d in gastric epithelial cells with Tmprss2-CreER T2 . Through histological staining and single-cell RNA sequencing, we observed that Kmt2c/d loss led to nuclear dysplasia and expansion of cells with mixed gastric lineage markers. When combined with Pten deletion, Kmt2c/d loss drove rapid development of muscle-invasive gastric adenocarcinoma as early as 3 weeks post Cre-mediated gene deletion. The adenocarcinoma exhibited decreased expression of gastric lineage markers and increased expression of intestinal differentiation markers, phenocopying human gastric adenocarcinoma. Kmt2c/d knockout reduced protein synthesis but upregulated transcription of ribosomal proteins, rendering sensitivity to mTORC1 inhibitors. Additionally, Kmt2c/d knockout increased MHC-I molecule expression and enhanced antigen presentation. Combination of mTROC1 inhibition and anti-PD1 immunotherapy significantly suppressed tumor growth in immune-competent mice. Together, these findings reveal the role of Kmt2c / d loss in gastric cancer initiation and suggest the potential therapeutic strategies for KMT2C/D -deficient gastric cancer.
Collapse
|
2
|
Giri H, Biswas I, Rezaie AR. Thrombomodulin: a multifunctional receptor modulating the endothelial quiescence. J Thromb Haemost 2024; 22:905-914. [PMID: 38266676 PMCID: PMC10960680 DOI: 10.1016/j.jtha.2024.01.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/01/2024] [Accepted: 01/05/2024] [Indexed: 01/26/2024]
Abstract
Thrombomodulin (TM) is a type 1 receptor best known for its function as an anticoagulant cofactor for thrombin activation of protein C on the surface of vascular endothelial cells. In addition to its anticoagulant cofactor function, TM also regulates fibrinolysis, complement, and inflammatory pathways. TM is a multidomain receptor protein with a lectin-like domain at its N-terminus that has been shown to exhibit direct anti-inflammatory functions. This domain is followed by 6 epidermal growth factor-like domains that support the interaction of TM with thrombin. The interaction inhibits the procoagulant function of thrombin and enables the protease to regulate the anticoagulant and fibrinolytic pathways by activating protein C and thrombin-activatable fibrinolysis inhibitor. TM has a Thr/Ser-rich region immediately above the membrane surface that harbors chondroitin sulfate glycosaminoglycans, and this region is followed by a single-spanning transmembrane and a C-terminal cytoplasmic domain. The structure and physiological function of the extracellular domains of TM have been extensively studied, and numerous excellent review articles have been published. However, the physiological function of the cytoplasmic domain of TM has remained poorly understood. Recent data from our laboratory suggest that intracellular signaling by the cytoplasmic domain of TM plays key roles in maintaining quiescence by modulating phosphatase and tensin homolog signaling in endothelial cells. This article briefly reviews the structure and function of extracellular domains of TM and focuses on the mechanism and possible physiological importance of the cytoplasmic domain of TM in modulating phosphatase and tensin homolog signaling in endothelial cells.
Collapse
Affiliation(s)
- Hemant Giri
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Indranil Biswas
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Alireza R Rezaie
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA; Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA.
| |
Collapse
|
3
|
Chen J, Kang J, Yuan S, O’Connell P, Zhang Z, Wang L, Liu J, Chen R. Exploring the Mechanisms of Traditional Chinese Herbal Therapy in Gastric Cancer: A Comprehensive Network Pharmacology Study of the Tiao-Yuan-Tong-Wei decoction. Pharmaceuticals (Basel) 2024; 17:414. [PMID: 38675376 PMCID: PMC11054859 DOI: 10.3390/ph17040414] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/03/2024] [Accepted: 03/15/2024] [Indexed: 04/28/2024] Open
Abstract
The use of herbal medicine as an adjuvant therapy in the management of gastric cancer has yielded encouraging outcomes, notably in enhancing overall survival rates and extending periods of disease remission. Additionally, herbal medicines have demonstrated potential anti-metastatic effects in gastric cancer. Despite these promising findings, there remains a significant gap in our understanding regarding the precise pharmacological mechanisms, the identification of specific herbal compounds, and their safety and efficacy profiles in the context of gastric cancer therapy. In addressing this knowledge deficit, the present study proposes a comprehensive exploratory analysis of the Tiao-Yuan-Tong-Wei decoction (TYTW), utilizing an integrative approach combining system pharmacology and molecular docking techniques. This investigation aims to elucidate the pharmacological actions of TYTW in gastric pathologies. It is hypothesized that the therapeutic efficacy of TYTW in counteracting gastric diseases stems from its ability to modulate key signaling pathways, thereby influencing PIK3CA activity and exerting anti-inflammatory effects. This modulation is observed predominantly in pathways such as PI3K/AKT, MAPK, and those directly associated with gastric cancer. Furthermore, the study explores how TYTW's metabolites (agrimoniin, baicalin, corosolic acid, and luteolin) interact with molecular targets like AKT1, CASP3, ESR1, IL6, PIK3CA, and PTGS2, and their subsequent impact on these critical pathways and biological processes. Therefore, this study represents preliminary research on the anticancer molecular mechanism of TYTW by performing network pharmacology and providing theoretical evidence for further experimental investigations.
Collapse
Affiliation(s)
- Juan Chen
- Department of Gastroenterology, Beijing Nuclear Industry Hospital, Beijing 102413, China; (J.C.)
| | - Jingdong Kang
- Department of General Surgery, Beijing Nuclear Industry Hospital, Beijing 102413, China
| | - Shouli Yuan
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China;
| | - Peter O’Connell
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, D02 PN40 Dublin, Ireland
| | - Zizhu Zhang
- Department of Gastroenterology, Beijing Nuclear Industry Hospital, Beijing 102413, China; (J.C.)
| | - Lina Wang
- Pharmacy Department, Beijing Water Resources Hospital, Beijing 100036, China
| | - Junying Liu
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, D02 PN40 Dublin, Ireland
| | - Rongfeng Chen
- National Center for Occupational Safety and Health, National Health Commission, Beijing 102308, China
| |
Collapse
|
4
|
Nie K, Zheng Z, Li X, Chang Y, Liu F, Wang X. Explore the active ingredients and potential mechanisms of JianPi QingRe HuaYu Methods in the treatment of gastric inflammation-cancer transformation by network pharmacology and experimental validation. BMC Complement Med Ther 2023; 23:411. [PMID: 37964307 PMCID: PMC10644588 DOI: 10.1186/s12906-023-04232-0] [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: 03/28/2023] [Accepted: 10/20/2023] [Indexed: 11/16/2023] Open
Abstract
BACKGROUND JianPi QingRe HuaYu Methods (JQH) have been long used to treat chronic atrophic gastritis (CAG) and precancerous lesions of gastric cancer (PLGC). However, whether JQH can inhibit the transformation of gastritis to gastric cancer (GC) remains unclear. METHODS Herein, we first retrieved the active ingredients and targets of JQH from the TCMSP database and the targets related to the gastric inflammation-cancer transformation from public databases. Differentially expressed genes (DEGs) related to gastric inflammation-cancer transformation were identified from the Gene Expression Omnibus (GEO) database. Then, we obtained the potential therapeutic targets of JQH in treating gastric inflammation-cancer transformation by intersecting drugs and disease targets. The Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and protein-protein interaction (PPI) analyses of the potential therapeutic targets were conducted using R software. Next, we conducted molecular docking and in vitro experiments to validate our results. RESULTS We obtained 214 potential therapeutic targets of JQH by intersecting drugs and disease targets. We found that the potential mechanisms of JQH in treating gastric inflammation-cancer transformation might be related to JAK-STAT, Wnt, p53 and VEGF signaling pathways. The molecular docking indicated that quercetin, as the main active ingredient of JQH, might inhibit gastric inflammation-cancer transformation by binding with specific receptors. Our experimental results showed that quercetin inhibited cells proliferation (P < 0.001), promoted cell apoptosis (P < 0.001), reduced the secretion of pro-inflammatory cytokines (P < 0.001) and promoted the secretion of anti-inflammatory cytokines (P < 0.001) in MNNG-induced GES-1 cells. Furthermore, quercetin inhibited cells proliferation (P < 0.001) and reduced mRNA and protein level of markers of PLGC (P < 0.001) in CDCA-induced GES-1 cells. CONCLUSION These results provide the material basis and regulatory mechanisms of JQH in treating gastric inflammation-cancer transformation.
Collapse
Affiliation(s)
- Kechao Nie
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
- School of Health Science, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Zhihua Zheng
- The Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
- Department of Gastroenterology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518000, China
| | - Xiushen Li
- Shenzhen University General Hospital, Shenzhen, 518060, China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Yonglong Chang
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - FengBin Liu
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Xiaoyu Wang
- School of Health Science, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| |
Collapse
|
5
|
Wang G, Ren Z, Zhao Y, Li Y. A nine-gene signature as prognostic biomarker in gastric cancer by bioinformatics analysis. Clin Transl Oncol 2023; 25:3296-3306. [PMID: 37041435 DOI: 10.1007/s12094-023-03180-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 03/28/2023] [Indexed: 04/13/2023]
Abstract
PURPOSE The prognosis of advanced gastric cancer (GC) remains poor. It is urgent and necessary to find suitable prognostic markers. miR-619-5p is highly expressed in GC. However, the value of miR-619-5p and its target genes as prognostic biomarkers of GC is unclear. METHODS RT-PCR was performed to verify the expression of miR-619-5p in GC cell lines and their exosomes. Western blotting and transmission electron microscope were used to identify exosomes. The target genes of miR-619-5p were predicted by RNA22 and TargetScan. The differentially expressed genes (DEGs) and prognosis-related genes (PRGs) were obtained using The Cancer Genome Atlas (TCGA) database. The DAVID database was used to analyse pathway enrichment and functional annotation of common target genes. The STRING database and Cytoscape software were used to screen key genes and visualize their functional modules. The survival analysis was conducted using TCGA and Kaplan-Meier Plotter (KMP) databases. Finally, a prognostic model was constructed on the foundation of the key genes to assess the reliability of the screening process. RESULTS The expression of miR-619-5p in GC cells and their exosomes was proved to be significantly higher than that in normal cell lines. There are 129 common target genes involved in 3 pathways and 28 functional annotations. Finally, nine key target genes of GC (BRCA1, RAD51, KIF11, ERCC6L, BRIP1, TIMELESS, CDC25A, CLSPN and NCAPG2) were identified, and a prognostic model was successfully constructed with a good predictive ability. CONCLUSIONS The model of 9-gene signature could effectively predict the prognosis of GC, and have great potential to be novel prognostic factors and therapeutic targets for patients with GC.
Collapse
Affiliation(s)
- Guan Wang
- Key Laboratory of Digestive System Tumours of Gansu Province, The Second Clinical Medical College of Lanzhou University, Second Hospital of Lanzhou University, 82 Cuiying Gate, Lanzhou, 730030, Gansu, China
| | - Zhijian Ren
- Key Laboratory of Digestive System Tumours of Gansu Province, The Second Clinical Medical College of Lanzhou University, Second Hospital of Lanzhou University, 82 Cuiying Gate, Lanzhou, 730030, Gansu, China
| | - Yang Zhao
- Key Laboratory of Digestive System Tumours of Gansu Province, The Second Clinical Medical College of Lanzhou University, Second Hospital of Lanzhou University, 82 Cuiying Gate, Lanzhou, 730030, Gansu, China
| | - Yumin Li
- Key Laboratory of Digestive System Tumours of Gansu Province, The Second Clinical Medical College of Lanzhou University, Second Hospital of Lanzhou University, 82 Cuiying Gate, Lanzhou, 730030, Gansu, China.
| |
Collapse
|
6
|
Wu S, Abdullah Al-Maskri AA, Li Q, Liu J, Cai S. A Novel miRNA Detection Method Using Loop-Mediated Isothermal Amplification. Int J Anal Chem 2023; 2023:6624884. [PMID: 37732283 PMCID: PMC10508998 DOI: 10.1155/2023/6624884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/22/2023] [Accepted: 08/29/2023] [Indexed: 09/22/2023] Open
Abstract
A novel ligation-based loop-mediated isothermal amplification has been developed for miRNA detection. Two stem-loop structure DNA linker A/B probes which hybridized with miRNA were designed to establish a rapid and ultrasensitive miRNA-LAMP system for miRNA detection. Target miR-200a was used to template the ligation of Linker A/B probes with SplintR Ligase and used as a dumbbell-shaped amplicon. By adding BIP/FIP and Bst 2.0 DNA polymerase, the LAMP reaction was carried out, which brought greatly improved amplification efficiency. The double-stranded DNA fluorescent dye EvaGreen was added for the detection of amplification product to achieve the quantification of the target miRNA. This method can detect miRNA in a linear range of seven orders of magnitude, with a detection limit of 100 fM. Therefore, this ultrasensitive miRNA-LAMP assay provides a new path for the highly sensitive quantitative analysis of miRNA, thereby bringing convenience to clinical diagnosis and prognostic research.
Collapse
Affiliation(s)
- Saiwei Wu
- Department of Pharmacy, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Abdu Ahmed Abdullah Al-Maskri
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Qun Li
- Department of Pharmacy, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Jiatong Liu
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Sheng Cai
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| |
Collapse
|
7
|
Hu Q, Huang T. Regulation of the Cell Cycle by ncRNAs Affects the Efficiency of CDK4/6 Inhibition. Int J Mol Sci 2023; 24:ijms24108939. [PMID: 37240281 DOI: 10.3390/ijms24108939] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/12/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023] Open
Abstract
Cyclin-dependent kinases (CDKs) regulate cell division at multiple levels. Aberrant proliferation induced by abnormal cell cycle is a hallmark of cancer. Over the past few decades, several drugs that inhibit CDK activity have been created to stop the development of cancer cells. The third generation of selective CDK4/6 inhibition has proceeded into clinical trials for a range of cancers and is quickly becoming the backbone of contemporary cancer therapy. Non-coding RNAs, or ncRNAs, do not encode proteins. Many studies have demonstrated the involvement of ncRNAs in the regulation of the cell cycle and their abnormal expression in cancer. By interacting with important cell cycle regulators, preclinical studies have demonstrated that ncRNAs may decrease or increase the treatment outcome of CDK4/6 inhibition. As a result, cell cycle-associated ncRNAs may act as predictors of CDK4/6 inhibition efficacy and perhaps present novel candidates for tumor therapy and diagnosis.
Collapse
Affiliation(s)
- Qingyi Hu
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Tao Huang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
8
|
Tang Y, Dong L, Zhang C, Li X, Li R, Lin H, Qi Y, Tang M, Peng Y, Liu C, Zhou J, Hou N, Liu W, Yang G, Yang X, Teng Y. PRMT5 acts as a tumor suppressor by inhibiting Wnt/β-catenin signaling in murine gastric tumorigenesis. Int J Biol Sci 2022; 18:4329-4340. [PMID: 35864961 PMCID: PMC9295066 DOI: 10.7150/ijbs.71581] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 05/21/2022] [Indexed: 11/05/2022] Open
Abstract
Previous studies have demonstrated the in vitro oncogenic role of protein arginine methyltransferase 5 (PRMT5) in gastric cancer cell lines. The in vivo function of PRMT5 in gastric tumorigenesis, however, is still unexplored. Here, we showed that Prmt5 deletion in mouse gastric epithelium resulted in spontaneous tumorigenesis in gastric antrum. All Prmt5-deficient mice displayed intestinal-type gastric cancer within 4 months of age. Of note, 20% (2/10) of Prmt5 mutants finally developed into invasive gastric cancer by 8 months of age. Gastric cancer caused by PRMT5 loss exhibited the increase in Lgr5+ stem cells, which are proposed to contribute to both the gastric tumorigenesis and progression in mouse models. Consistent with the notion that Lgr5 is the target of Wnt/β-catenin signaling, whose activation is the most predominant driver for gastric tumorigenesis, Prmt5 mutant gastric cancer showed the activation of Wnt/β-Catenin signaling. Furthermore, in human gastric cancer samples, PRMT5 deletion and downregulation were frequently observed and associated with the poor prognosis. We propose that as opposed to the tumor-promoting role of PRMT5 well-established in the progression of various cancer types, PRMT5 functions as a tumor suppressor in vivo, at least during gastric tumor formation.
Collapse
Affiliation(s)
- Yuling Tang
- State Key Laboratory of Proteomics, Beijing Proteome Research Centre, National Centre for Protein Sciences, Beijing Institute of Lifeomics, Beijing 102206, China.,Laboratory Animal Center, the Academy of Military Medical Sciences, Beijing 100071, China
| | - Lei Dong
- State Key Laboratory of Proteomics, Beijing Proteome Research Centre, National Centre for Protein Sciences, Beijing Institute of Lifeomics, Beijing 102206, China
| | - Chong Zhang
- State Key Laboratory of Proteomics, Beijing Proteome Research Centre, National Centre for Protein Sciences, Beijing Institute of Lifeomics, Beijing 102206, China
| | - Xiubin Li
- Department of Urology, the Third Medical Center of Chinese PLA General Hospital, Beijing 100039, China
| | - Rongyu Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Centre, National Centre for Protein Sciences, Beijing Institute of Lifeomics, Beijing 102206, China
| | - Huisang Lin
- State Key Laboratory of Proteomics, Beijing Proteome Research Centre, National Centre for Protein Sciences, Beijing Institute of Lifeomics, Beijing 102206, China
| | - Yini Qi
- State Key Laboratory of Proteomics, Beijing Proteome Research Centre, National Centre for Protein Sciences, Beijing Institute of Lifeomics, Beijing 102206, China
| | - Mingchuan Tang
- State Key Laboratory of Proteomics, Beijing Proteome Research Centre, National Centre for Protein Sciences, Beijing Institute of Lifeomics, Beijing 102206, China
| | - Yanli Peng
- State Key Laboratory of Proteomics, Beijing Proteome Research Centre, National Centre for Protein Sciences, Beijing Institute of Lifeomics, Beijing 102206, China
| | - Chuan Liu
- State Key Laboratory of Proteomics, Beijing Proteome Research Centre, National Centre for Protein Sciences, Beijing Institute of Lifeomics, Beijing 102206, China
| | - Jian Zhou
- State Key Laboratory of Proteomics, Beijing Proteome Research Centre, National Centre for Protein Sciences, Beijing Institute of Lifeomics, Beijing 102206, China
| | - Ning Hou
- State Key Laboratory of Proteomics, Beijing Proteome Research Centre, National Centre for Protein Sciences, Beijing Institute of Lifeomics, Beijing 102206, China
| | - Wenjia Liu
- State Key Laboratory of Proteomics, Beijing Proteome Research Centre, National Centre for Protein Sciences, Beijing Institute of Lifeomics, Beijing 102206, China
| | - Guan Yang
- State Key Laboratory of Proteomics, Beijing Proteome Research Centre, National Centre for Protein Sciences, Beijing Institute of Lifeomics, Beijing 102206, China
| | - Xiao Yang
- State Key Laboratory of Proteomics, Beijing Proteome Research Centre, National Centre for Protein Sciences, Beijing Institute of Lifeomics, Beijing 102206, China
| | - Yan Teng
- State Key Laboratory of Proteomics, Beijing Proteome Research Centre, National Centre for Protein Sciences, Beijing Institute of Lifeomics, Beijing 102206, China
| |
Collapse
|
9
|
Lee JS, Kim GH, Lee JH, Ryu JY, Oh EJ, Kim HM, Kwak S, Hur K, Chung HY. MicroRNA-365a/b-3p as a Potential Biomarker for Hypertrophic Scars. Int J Mol Sci 2022; 23:ijms23116117. [PMID: 35682793 PMCID: PMC9181131 DOI: 10.3390/ijms23116117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 05/25/2022] [Accepted: 05/28/2022] [Indexed: 11/29/2022] Open
Abstract
The clinical aspects of hypertrophic scarring vary according to personal constitution and body part. However, the mechanism of hypertrophic scar (HS) formation remains unclear. MicroRNAs (miRNAs) are known to contribute to HS formation, however, their detailed role remains unknown. In this study, candidate miRNAs were identified and analyzed as biomarkers of hypertrophic scarring for future clinical applications. HSfibroblasts and normal skin fibroblasts from patients were used for profiling and validation of miRNAs. An HS mouse model with xenografted human skin on nude mice was established. The miRNA expression between normal human, normal mouse, and mouse HS skin tissues was compared. Circulating miRNA expression levels in the serum of normal mice and mice with HSs were also analyzed. Ten upregulated and twenty-one downregulated miRNAs were detected. Among these, miR-365a/b-3p and miR-16-5p were identified as candidate miRNAs with statistically significant differences; miR-365a/b-3p was significantly upregulated (p = 0.0244). In mouse studies, miR-365a/b-3p expression levels in skin tissue and serum were higher in mice with HSs than in the control group. These results indicate that miRNAs contribute to hypertrophic scarring and that miR-365a/b-3p may be considered a potential biomarker for HS formation.
Collapse
Affiliation(s)
- Joon Seok Lee
- Department of Plastic and Reconstructive Surgery, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (J.S.L.); (J.H.L.); (J.Y.R.); (E.J.O.); (H.M.K.)
| | - Gyeong Hwa Kim
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Daegu 41199, Korea;
- CMRI, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Jong Ho Lee
- Department of Plastic and Reconstructive Surgery, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (J.S.L.); (J.H.L.); (J.Y.R.); (E.J.O.); (H.M.K.)
| | - Jeong Yeop Ryu
- Department of Plastic and Reconstructive Surgery, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (J.S.L.); (J.H.L.); (J.Y.R.); (E.J.O.); (H.M.K.)
| | - Eun Jung Oh
- Department of Plastic and Reconstructive Surgery, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (J.S.L.); (J.H.L.); (J.Y.R.); (E.J.O.); (H.M.K.)
- CMRI, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Hyun Mi Kim
- Department of Plastic and Reconstructive Surgery, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (J.S.L.); (J.H.L.); (J.Y.R.); (E.J.O.); (H.M.K.)
- CMRI, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Suin Kwak
- BK21 FOUR KNU Convergence Educational Program of Biomedical Science for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41199, Korea;
| | - Keun Hur
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Daegu 41199, Korea;
- CMRI, School of Medicine, Kyungpook National University, Daegu 41944, Korea
- BK21 FOUR KNU Convergence Educational Program of Biomedical Science for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41199, Korea;
- Correspondence: (K.H.); (H.Y.C.); Tel.: +82-53-420-4821 (K.H.); +82-53-420-5692 (H.Y.C.); Fax: +82-53-422-1466 (K.H.); +82-53-425-3879 (H.Y.C.)
| | - Ho Yun Chung
- Department of Plastic and Reconstructive Surgery, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (J.S.L.); (J.H.L.); (J.Y.R.); (E.J.O.); (H.M.K.)
- CMRI, School of Medicine, Kyungpook National University, Daegu 41944, Korea
- BK21 FOUR KNU Convergence Educational Program of Biomedical Science for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41199, Korea;
- Kyungpook National University Bio-Medical Research Institute, Kyungpook National University, Daegu 41944, Korea
- Correspondence: (K.H.); (H.Y.C.); Tel.: +82-53-420-4821 (K.H.); +82-53-420-5692 (H.Y.C.); Fax: +82-53-422-1466 (K.H.); +82-53-425-3879 (H.Y.C.)
| |
Collapse
|
10
|
Fan X, Zhang L, Huang J, Zhong Y, Fan Y, Zhou T, Lu M. An Integrated Immune-Related Bioinformatics Analysis in Glioma: Prognostic Signature's Identification and Multi-Omics Mechanisms' Exploration. Front Genet 2022; 13:889629. [PMID: 35601497 PMCID: PMC9114310 DOI: 10.3389/fgene.2022.889629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 04/18/2022] [Indexed: 12/05/2022] Open
Abstract
As the traditional treatment for glioma, the most common central nervous system malignancy with poor prognosis, the efficacy of high-intensity surgery combined with radiotherapy and chemotherapy is not satisfactory. The development of individualized scientific treatment strategy urgently requires the guidance of signature with clinical predictive value. In this study, five prognosis-related differentially expressed immune-related genes (PR-DE-IRGs) (CCNA2, HMGB2, CASP3, APOBEC3C, and BMP2) highly associated with glioma were identified for a prognostic model through weighted gene co-expression network analysis, univariate Cox and lasso regression. Kaplan-Meier survival curves, receiver operating characteristic curves and other methods have shown that the model has good performance in predicting the glioma patients' prognosis. Further combined nomogram provided better predictive performance. The signature's guiding value in clinical treatment has also been verified by multiple analysis results. We also constructed a comprehensive competing endogenous RNA (ceRNA) regulatory network based on the protective factor BMP2 to further explore its potential role in glioma progression. Numerous immune-related biological functions and pathways were enriched in a high-risk population. Further multi-omics integrative analysis revealed a strong correlation between tumor immunosuppressive environment/IDH1 mutation and signature, suggesting that their cooperation plays an important role in glioma progression.
Collapse
Affiliation(s)
- Xin Fan
- Department of Emergency Medicine, Shangrao Hospital Affiliated to Nanchang University, Shangrao People’s Hospital, Shangrao, China
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Lingling Zhang
- School of Stomatology, Nanchang University, Nanchang, China
| | - Junwen Huang
- The First Clinical Medical College of Nanchang University, Nanchang, China
| | - Yun Zhong
- The First Clinical Medical College of Nanchang University, Nanchang, China
| | - Yanting Fan
- The First Clinical Medical College of Nanchang University, Nanchang, China
| | - Tong Zhou
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Min Lu
- Department of Emergency Medicine, Shangrao Hospital Affiliated to Nanchang University, Shangrao People’s Hospital, Shangrao, China
| |
Collapse
|
11
|
Qi Y, Chen J, Duan J, Kang L, Wang K, Chen Z, Xu B, Gu R. Major vault protein attenuates cardiomyocyte injury in doxorubicin-induced cardiomyopathy through activating AKT. BMC Cardiovasc Disord 2022; 22:77. [PMID: 35246039 PMCID: PMC8896232 DOI: 10.1186/s12872-022-02517-9] [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: 08/30/2021] [Accepted: 02/18/2022] [Indexed: 11/30/2022] Open
Abstract
Background Doxorubicin (DOX) has limited chemotherapy application for malignancies due to cardiotoxicity. The pathogenesis of DOX-induced cardiomyopathy (DiCM) is yet to be elucidated. Increasing studies proved that activation of AKT prevented cardiomyocyte apoptosis and cardiac dysfunction in response to DOX insult. Our previous studies indicated that major vault protein (MVP) deficiency was accompanied by suppressed phosphorylation of AKT in metabolic diseases. This study aimed to investigate the role and underlying mechanism of MVP on cardiomyocyte apoptosis in DiCM. Methods Mice were intraperitoneally injected with DOX 5 mg/kg, once a week for 5 weeks, the total cumulative dose was 25 mg/kg. Cardiomyocyte-specific MVP overexpression was achieved using an adeno-associated virus system under the cTnT promoter after the fourth DOX injection. Cardiac function was examined by echocardiography followed by euthanasia. Tissue and serum were collected for morphology analysis and biochemical examination. Results Herein, we found that MVP expression was upregulated in DOX-treated murine hearts. Cardiac-specific MVP overexpression alleviated DOX-induced cardiac dysfunction, oxidative stress and fibrosis. Mechanistically, MVP overexpression activated AKT signaling and decreased cardiomyocyte apoptosis in DiCM. Conclusions Based on these findings, we supposed that MVP was a potential therapeutic agent against DiCM. Supplementary Information The online version contains supplementary material available at 10.1186/s12872-022-02517-9.
Collapse
Affiliation(s)
- Yu Qi
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Jianzhou Chen
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Junfeng Duan
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Lina Kang
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Kun Wang
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Ziwei Chen
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Biao Xu
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China. .,State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210023, China.
| | - Rong Gu
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China.
| |
Collapse
|
12
|
Huang F, Xiang Y, Li T, Huang Y, Wang J, Zhang HM, Li HH, Dai ZT, Li JP, Li H, Zhou J, Liao XH. Metformin and MiR-365 synergistically promote the apoptosis of gastric cancer cells via MiR-365-PTEN-AMPK axis. Pathol Res Pract 2022; 230:153740. [PMID: 35007850 DOI: 10.1016/j.prp.2021.153740] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 12/10/2021] [Accepted: 12/16/2021] [Indexed: 01/19/2023]
Abstract
Metformin is an oral biguanide used to treat diabetes. Recent study showed it may interfere was related to cancer progression and has a positive effect on cancer prevention and treatment, which attracts a new hot research topic. Here we show that Metformin suppressed the proliferation but induced apoptosis of gastric cells. Notably, Metformin enhanced gastriccell apoptosis via modulating AMPK signaling. Furthermore, Metformin and miR-365 synergistically promote the apoptosis of gastric cancer cells by miR-365-PTEN-AMPK axis. Our study unraveled a novel signaling axis in the regulation in gastric cancer, which could be amplified by the application of metformin. The new effect of metformin potentiates its novel therapeutic application in the future. AVAILABILITY OF DATA AND MATERIALS: The data generated during this study are included in this article and its supplementary information files are available from the corresponding author on reasonable request.
Collapse
Affiliation(s)
- Feng Huang
- Institute of Biology and Medicine, Wuhan University of Science and Technology, 430000, PR China.
| | - Yuan Xiang
- Department of Medical Laboratory, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Hubei 430014, PR China.
| | - Ting Li
- Institute of Biology and Medicine, Wuhan University of Science and Technology, 430000, PR China.
| | - You Huang
- Institute of Biology and Medicine, Wuhan University of Science and Technology, 430000, PR China.
| | - Jun Wang
- Institute of Biology and Medicine, Wuhan University of Science and Technology, 430000, PR China.
| | - Hui-Min Zhang
- Institute of Biology and Medicine, Wuhan University of Science and Technology, 430000, PR China.
| | - Han-Han Li
- Institute of Biology and Medicine, Wuhan University of Science and Technology, 430000, PR China.
| | - Zhou-Tong Dai
- Institute of Biology and Medicine, Wuhan University of Science and Technology, 430000, PR China.
| | - Jia-Peng Li
- Institute of Biology and Medicine, Wuhan University of Science and Technology, 430000, PR China.
| | - Hui Li
- Institute of Biology and Medicine, Wuhan University of Science and Technology, 430000, PR China.
| | - Jun Zhou
- Institute of Biology and Medicine, Wuhan University of Science and Technology, 430000, PR China.
| | - Xing-Hua Liao
- Institute of Biology and Medicine, Wuhan University of Science and Technology, 430000, PR China.
| |
Collapse
|
13
|
Gao Y, Miao Y, Zhang W, Ru X, Hou L. MicroRNA-365 induces apoptosis and inhibits invasion of human myeloma cells by targeting homeobox A9 (HOXA9). ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2021; 85:103627. [PMID: 33621688 DOI: 10.1016/j.etap.2021.103627] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 02/04/2021] [Accepted: 02/18/2021] [Indexed: 06/12/2023]
Abstract
The aberrant micro-RNA (miR) expression has been reported to play a vital role in proliferation and tumorigenesis and of several human cancers. MicroRNA-365 (miR-365) has been shown to exhibit tumor-suppressive or oncogenic role in several human cancers. Nonetheless, little is known about its growth regulatory role in human multiple myeloma. The present study characterized the regulatory control exercised by miR-365 in multiple myeloma. The results showed significant (P < 0.05) upregulation of miR-365 in myeloma tissues and cell lines. Overexpression of miR-365 significantly (P < 0.05) suppressed the proliferation and inhibition of miR-365 promoted the proliferation of the human myeloma cells. The tumor-suppressive effects of miR-365 were found to be the result of apoptosis in the IM-9 myeloma cells. The miR-365 overexpression also suppressed the invasion of the IM-9 myeloma cells. The homeobox gene, HOXA9 was identified as the molecular regulatory target of miR-365 in human myeloma. The overexpression of miR-365 was shown to cause suppression of HOXA9. The silencing of HOXA9 could also suppress the growth of the IM-9 myeloma cells while as the overexpression of HOXA9 could abolish the tumor-suppressive effects of miR-365. The in vivo study revealed that miR-365 inhibits the growth of the xenografted tumors. Nonetheless, the inhibition of miR-365 promotes the growth of the xenografted tumors. To sum up, the current study suggests the tumor-suppressive effects of miR-365 in human myeloma and highlights the applicability of miR-365 as vital therapeutic target against this fatal malignancy.
Collapse
Affiliation(s)
- Ying Gao
- Department of Hematology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, 710068, China
| | - Yudi Miao
- Department of Hematology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, 710068, China
| | - Weihua Zhang
- Department of Hematology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, 710068, China
| | - Xingli Ru
- Department of Hematology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, 710068, China
| | - LiMin Hou
- Department of Hematology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, 710068, China.
| |
Collapse
|
14
|
Xie M, Wu Z, Ying S, Liu L, Zhao C, Yao C, Zhang Z, Luo C, Wang W, Zhao D, Zhang J, Qiu W, Wang Y. Sublytic C5b-9 induces glomerular mesangial cell proliferation via ERK1/2-dependent SOX9 phosphorylation and acetylation by enhancing Cyclin D1 in rat Thy-1 nephritis. Exp Mol Med 2021; 53:572-590. [PMID: 33811247 PMCID: PMC8102557 DOI: 10.1038/s12276-021-00589-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 02/07/2021] [Accepted: 02/09/2021] [Indexed: 02/01/2023] Open
Abstract
Glomerular mesangial cell (GMC) proliferation is a histopathological alteration in human mesangioproliferative glomerulonephritis (MsPGN) or in animal models of MsPGN, e.g., the rat Thy-1 nephritis (Thy-1N) model. Although sublytic C5b-9 assembly on the GMC membrane can trigger cell proliferation, the mechanisms are still undefined. We found that sublytic C5b-9-induced rat GMC proliferation was driven by extracellular signal-regulated kinase 1/2 (ERK1/2), sry-related HMG-box 9 (SOX9), and Cyclin D1. Here, ERK1/2 phosphorylation was a result of the calcium influx-PKC-α-Raf-MEK1/2 axis activated by sublytic C5b-9, and Cyclin D1 gene transcription was enhanced by ERK1/2-dependent SOX9 binding to the Cyclin D1 promoter (-582 to -238 nt). In addition, ERK1/2 not only interacted with SOX9 in the cell nucleus to mediate its phosphorylation at serine residues 64 (a new site identified by mass spectrometry) and 181 (a known site), but also indirectly induced SOX9 acetylation by elevating the expression of general control non-repressed protein 5 (GCN5), which together resulted in Cyclin D1 synthesis and GMC proliferation. Moreover, our in vivo experiments confirmed that silencing these genes ameliorated the lesions of Thy-1N rats and reduced SOX9 phosphorylation, acetylation and Cyclin D1 expression. Furthermore, the renal tissue sections of MsPGN patients also showed higher phosphorylation or expression of ERK1/2, SOX9, and Cyclin D1. In summary, these findings suggest that sublytic C5b-9-induced GMC proliferation in rat Thy-1N requires SOX9 phosphorylation and acetylation via enhanced Cyclin D1 gene transcription, which may provide a new insight into human MsPGN pathogenesis.
Collapse
Affiliation(s)
- Mengxiao Xie
- grid.89957.3a0000 0000 9255 8984Department of Immunology, and Key Laboratory of Immunological Environment and Disease, Nanjing Medical University, 101 Longmian Road, Nanjing, Jiangsu 211166 China ,grid.412676.00000 0004 1799 0784Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu 210029 China
| | - Zhijiao Wu
- grid.89957.3a0000 0000 9255 8984Department of Immunology, and Key Laboratory of Immunological Environment and Disease, Nanjing Medical University, 101 Longmian Road, Nanjing, Jiangsu 211166 China
| | - Shuai Ying
- grid.89957.3a0000 0000 9255 8984Department of Immunology, and Key Laboratory of Immunological Environment and Disease, Nanjing Medical University, 101 Longmian Road, Nanjing, Jiangsu 211166 China
| | - Longfei Liu
- grid.89957.3a0000 0000 9255 8984Department of Immunology, and Key Laboratory of Immunological Environment and Disease, Nanjing Medical University, 101 Longmian Road, Nanjing, Jiangsu 211166 China ,grid.89957.3a0000 0000 9255 8984Department of Central Laboratory, The Affiliated Huaian No. 1 People’s Hospital, Nanjing Medical University, One West Huanghe Road, Huai’an, Jiangsu 223300 China
| | - Chenhui Zhao
- grid.412676.00000 0004 1799 0784Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu 210029 China
| | - Chunlei Yao
- grid.412676.00000 0004 1799 0784Department of Nephrology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu 210029 China
| | - Zhiwei Zhang
- grid.89957.3a0000 0000 9255 8984Department of Immunology, and Key Laboratory of Immunological Environment and Disease, Nanjing Medical University, 101 Longmian Road, Nanjing, Jiangsu 211166 China
| | - Can Luo
- grid.89957.3a0000 0000 9255 8984Department of Immunology, and Key Laboratory of Immunological Environment and Disease, Nanjing Medical University, 101 Longmian Road, Nanjing, Jiangsu 211166 China
| | - Wenbo Wang
- grid.89957.3a0000 0000 9255 8984Department of Immunology, and Key Laboratory of Immunological Environment and Disease, Nanjing Medical University, 101 Longmian Road, Nanjing, Jiangsu 211166 China
| | - Dan Zhao
- grid.89957.3a0000 0000 9255 8984Department of Immunology, and Key Laboratory of Immunological Environment and Disease, Nanjing Medical University, 101 Longmian Road, Nanjing, Jiangsu 211166 China
| | - Jing Zhang
- grid.89957.3a0000 0000 9255 8984Department of Immunology, and Key Laboratory of Immunological Environment and Disease, Nanjing Medical University, 101 Longmian Road, Nanjing, Jiangsu 211166 China
| | - Wen Qiu
- grid.89957.3a0000 0000 9255 8984Department of Immunology, and Key Laboratory of Immunological Environment and Disease, Nanjing Medical University, 101 Longmian Road, Nanjing, Jiangsu 211166 China ,grid.89957.3a0000 0000 9255 8984Key Laboratory of Antibody Technology of Ministry of Health, Nanjing Medical University, Nanjing, Jiangsu 211166 China
| | - Yingwei Wang
- grid.89957.3a0000 0000 9255 8984Department of Immunology, and Key Laboratory of Immunological Environment and Disease, Nanjing Medical University, 101 Longmian Road, Nanjing, Jiangsu 211166 China ,grid.89957.3a0000 0000 9255 8984Key Laboratory of Antibody Technology of Ministry of Health, Nanjing Medical University, Nanjing, Jiangsu 211166 China
| |
Collapse
|
15
|
Jiang ZB, Ma BQ, Feng Z, Liu SG, Gao P, Yan HT. miR-365 inhibits the progression of gallbladder carcinoma and predicts the prognosis of Gallbladder carcinoma patients. Cell Cycle 2021; 20:308-319. [PMID: 33459111 DOI: 10.1080/15384101.2021.1874694] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Gallbladder carcinoma (GBC) is one of the most common fatal biliary tract tumors in the world. Its 3-year survival rate is 30% and the recurrence rate remains very high. miR-365 was downregulated in numerous tumors and worked as tumor suppressor gene. However, the role of miR-365 in GBC was unclear. In this study, our results found that the expression of miR-365 in GBC tissues was reduced rather than that in non-cancerous tissues. miR-365 overexpression inhibited the proliferation, metastasis and expansion of GBC CSCs. Mechanically, bioinformatic and luciferase reporter analysis identified Ras-related C3 botulinum toxin substrate 1 (RAC1) as a direct target of miR-365. Overexpression of miR-365 in GBC cells reduced the RAC1 mRNA and protein expression. The special RAC1 inhibitor EHop-106 abolished the discrepancy of growth, metastasis and self-renewal ability between miR-365-overexpression GBC cells and their control cells, which further demonstrated that RAC1 was involved in miR-365-disrupted GBC cells growth, metastasis and self-renewal. More importantly, reduced expression of miR-365 was a predictor of poor prognosis of GBC patients. In conclusion, miR-365 inhibited GBC cell growth, metastasis and self-renewal capacity by directly targeting RAC1, and may therefore prove to be a novel prognosis biomarker for GBC patients.
Collapse
Affiliation(s)
- Ze-Bin Jiang
- Department of General Surgery, Gansu Provincial Hospital , Gansu, China
| | - Bing-Qiang Ma
- Department of General Surgery, Gansu Provincial Hospital , Gansu, China
| | - Zongfeng Feng
- Department of General Surgery, Cao County People's Hospital , Heze, Shandong Province, China
| | - Shao-Guang Liu
- Department of Emergency Surgery, Gansu Provincial Hospital , Gansu, China
| | - Peng Gao
- Department of General Surgery, Gansu Provincial Hospital , Gansu, China
| | - Hui-Ting Yan
- Department of Nursing Department, Gansu Provincial Hospital , Gansu, China
| |
Collapse
|
16
|
Hao D, Wang X, Wang X, Thomsen B, Yang Y, Lan X, Huang Y, Chen H. MicroRNA bta-miR-365-3p inhibits proliferation but promotes differentiation of primary bovine myoblasts by targeting the activin A receptor type I. J Anim Sci Biotechnol 2021; 12:16. [PMID: 33431058 PMCID: PMC7802253 DOI: 10.1186/s40104-020-00528-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 11/18/2020] [Indexed: 12/29/2022] Open
Abstract
Background MicroRNAs act as post-transcriptional regulators that repress translation or degrade mRNA transcripts. Each microRNA has many mRNA targets and each mRNA may be targeted by several microRNAs. Skeletal muscles express a plethora of microRNA genes that regulate muscle development and function by controlling the expression of protein-coding target genes. To expand our understanding of the role of microRNA, specifically bta-miR-365-3p, in muscle biology, we investigated its functions in regulating primary bovine myoblast proliferation and differentiation. Results Firstly, we found that bta-miR-365-3p was predominantly expressed in skeletal muscle and heart tissue in Chinese Qinchuan beef cattle. Quantitative PCR and western blotting results showed that overexpression of bta-miR-365-3p significantly reduced the expression levels of cyclin D1 (CCND1), cyclin dependent kinase 2 (CDK2) and proliferating cell nuclear antigen (PCNA) but stimulated the expression levels of muscle differentiation markers, i.e., MYOD1, MYOG at both mRNA and protein level. Moreover, downregulation of bta-miR-365-3p increased the expression of CCND1, CDK2 and PCNA but decreased the expression of MYOD1 and MYOG at both mRNA and protein levels. Furthermore, flow cytometry, EdU proliferation assays and immunostaining results showed that increased levels of bta-miR-365-3p suppressed cell proliferation but promoted myotube formation, whereas decreased levels of bta-miR-365-3p resulted in the opposite consequences. Finally, we identified that activin A receptor type I (ACVR1) could be a direct target of bta-miR-365-3p. It was demonstrated that bta-miR-365-3p can bind to the 3’UTR of ACVR1 gene to regulate its expression based on dual luciferase gene reporter assays. Consistently, knock-down of ACVR1 was associated with decreased expressions of CDK2, CCND1 and PCNA but increased expression of MYOG and MYOD1 both at mRNA and protein level. Conclusion Collectively, these data suggested that bta-miR-365-3p represses proliferation but promotes differentiation of bovine myoblasts through several biological mechanisms involving downregulation of ACVR1. Supplementary Information The online version contains supplementary material available at 10.1186/s40104-020-00528-0.
Collapse
Affiliation(s)
- Dan Hao
- College of Animal Science and Technology, Northwest A&F University, Shaanxi Key Laboratory of Animal Genetics, Breeding and Reproduction, Yangling, 712100, Shaanxi, China.,Department of Molecular Biology and Genetics, Aarhus University, 8000, Aarhus C, Denmark
| | - Xiaogang Wang
- College of Animal Science and Technology, Northwest A&F University, Shaanxi Key Laboratory of Animal Genetics, Breeding and Reproduction, Yangling, 712100, Shaanxi, China
| | - Xiao Wang
- Quantitative Genomics, Bioinformatics and Computational Biology Group, Department of Applied Mathematics and Computer Science, Technical University of Denmark, Richard Petersens Plads, Building 324, 2800, Kongens Lyngby, Denmark
| | - Bo Thomsen
- Department of Molecular Biology and Genetics, Aarhus University, 8000, Aarhus C, Denmark
| | - Yu Yang
- College of Animal Science and Technology, Northwest A&F University, Shaanxi Key Laboratory of Animal Genetics, Breeding and Reproduction, Yangling, 712100, Shaanxi, China
| | - Xianyong Lan
- College of Animal Science and Technology, Northwest A&F University, Shaanxi Key Laboratory of Animal Genetics, Breeding and Reproduction, Yangling, 712100, Shaanxi, China
| | - Yongzhen Huang
- College of Animal Science and Technology, Northwest A&F University, Shaanxi Key Laboratory of Animal Genetics, Breeding and Reproduction, Yangling, 712100, Shaanxi, China
| | - Hong Chen
- College of Animal Science and Technology, Northwest A&F University, Shaanxi Key Laboratory of Animal Genetics, Breeding and Reproduction, Yangling, 712100, Shaanxi, China.
| |
Collapse
|
17
|
Polymorphisms in Pepsinogen C and miRNA Genes Associate with High Serum Pepsinogen II in Gastric Cancer Patients. Microorganisms 2021; 9:microorganisms9010126. [PMID: 33430456 PMCID: PMC7827830 DOI: 10.3390/microorganisms9010126] [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: 11/18/2020] [Accepted: 01/03/2021] [Indexed: 12/18/2022] Open
Abstract
Background: Pepsinogen (PG) II (PGII) is a serological marker used to estimate the risk of gastric cancer but how PGII expression is regulated is largely unknown. It has been suggested that PGII expression, from the PGC (Progastricsin) gene, is regulated by microRNAs (miRNA), but how PGII levels vary with Helicobacter pylori (H. pylori) infection and miRNAs genotype remains unclear. Methods: Serum levels of PGI and PGII were determined in 80 patients with gastric cancer and persons at risk for gastric cancer (74 first-degree relatives of patients, 62 patients with autoimmune chronic atrophic gastritis, and 2 patients with dysplasia), with and without H. pylori infection. As control from the general population, 52 blood donors were added to the analyses. Associations between PGII levels and genetic variants in PGC and miRNA genes in these groups were explored based on H. pylori seropositivity and the risk for gastric cancer. The two-dimensional difference in gel electrophoresis (2D-DIGE) and the NanoString analysis of messenger RNA (mRNAs) from gastric cancer tissue were used to determine the pathways associated with increased PGII levels. Results: PGII levels were significantly higher in patients with gastric cancer, and in those with H. pylori infection, than in other patients or controls. A PGI/PGII ratio ≤ 3 was found better than PGI < 25 ng/mL to identify patients with gastric cancer (15.0% vs. 8.8%). For two genetic variants, namely rs8111742 in miR-Let-7e and rs121224 in miR-365b, there were significant differences in PGII levels between genotype groups among patients with gastric cancer (p = 0.02 and p = 0.01, respectively), but not among other study subjects. Moreover, a strict relation between rs9471643 C-allele with H. pylori infection and gastric cancer was underlined. Fold change in gene expression of mRNA isolated from gastric cancer tissue correlated well with polymorphism, H. pylori infection, increased PGII level, and pathway for bacteria cell entry into the host. Conclusions: Serum PGII levels depend in part on an interaction between H. pylori and host miRNA genotypes, which may interfere with the cut-off of PGI/PGII ratio used to identify persons at risk of gastric cancer. Results reported new findings regarding the relation among H. pylori, PGII-related host polymorphism, and genes involved in this interaction in the gastric cancer setting.
Collapse
|
18
|
Zhu G, Yang S, Wang R, Lei J, Ji P, Wang J, Tao K, Yang C, Ge S, Wang L. P53/miR-154 Pathway Regulates the Epithelial-Mesenchymal Transition in Glioblastoma Multiforme Cells by Targeting TCF12. Neuropsychiatr Dis Treat 2021; 17:681-693. [PMID: 33664574 PMCID: PMC7924251 DOI: 10.2147/ndt.s273578] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 01/18/2021] [Indexed: 02/06/2023] Open
Abstract
PURPOSE Glioblastoma multiforme (GBM) is an aggressive brain tumor with a rather short survival time. Mutation of p53 has been observed and reported to play critical roles in the progression of GBM. However, the pathological mechanisms are still unclear. This study was designed to identify the role of miR-154 in mediating the biological functions of p53 in glioblastoma multiforme. METHODS In the current study, the expression of miR-154 in GBM tissue samples and cell lines with wt-p53 or mutant p53 was evaluated. The functions of miR-154 in tumor migration, invasion and epithelial-mesenchymal transition were analyzed in vitro. A luciferase reporter assay was used to identify the target of miR-154. RESULTS We found that expression of miR-154 was much lower in patient tissues with mutant p53. Further study revealed that p53 was a transcription factor of miR-154 and that the R273H mutation led to its inactivation. In addition, overexpression of miR-154 remarkably suppressed cell migration, invasion and EMT in vitro and tumor growth in vivo. Moreover, TCF12 was proven to be a direct target of miR-154, and the tumor suppressive effect of miR-154 was reversed by TCF12. CONCLUSION Overall, miR-154, which was regulated by wt-p53, inhibited migration, invasion and EMT of GBM cells by targeting TCF12, indicating that miR-154 may act as a biomarker and that the p53/miR-154/TCF12 pathway could be a potential therapeutic target for GBM.
Collapse
Affiliation(s)
- Gang Zhu
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Shirong Yang
- Department of General Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Ronglin Wang
- Department of Oncology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Jie Lei
- Department of Neurosurgery, Wuhan General Hospital of PLA, Wuhan, Hubei, People's Republic of China
| | - Peigang Ji
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Jiancai Wang
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Kai Tao
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Chen Yang
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Shunnan Ge
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Liang Wang
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| |
Collapse
|
19
|
Adebamowo SN, Adeyemo AA, Rotimi CN, Olaniyan O, Offiong R, Adebamowo CA. Genome-wide association study of prevalent and persistent cervical high-risk human papillomavirus (HPV) infection. BMC MEDICAL GENETICS 2020; 21:231. [PMID: 33225922 PMCID: PMC7682060 DOI: 10.1186/s12881-020-01156-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Accepted: 10/25/2020] [Indexed: 01/20/2023]
Abstract
BACKGROUND Genetic factors may influence the susceptibility to high-risk (hr) human papillomavirus (HPV) infection and persistence. We conducted the first genome-wide association study (GWAS) to identify variants associated with cervical hrHPV infection and persistence. METHODS Participants were 517 Nigerian women evaluated at baseline and 6 months follow-up visits for HPV. HPV was characterized using SPF10/LiPA25. hrHPV infection was positive if at least one carcinogenic HPV genotype was detected in a sample provided at the baseline visit and persistent if at least one carcinogenic HPV genotype was detected in each of the samples provided at the baseline and follow-up visits. Genotyping was done using the Illumina Multi-Ethnic Genotyping Array (MEGA) and imputation was done using the African Genome Resources Haplotype Reference Panel. Association analysis was done for hrHPV infection (125 cases/392 controls) and for persistent hrHPV infection (51 cases/355 controls) under additive genetic models adjusted for age, HIV status and the first principal component (PC) of the genotypes. RESULTS The mean (±SD) age of the study participants was 38 (±8) years, 48% were HIV negative, 24% were hrHPV positive and 10% had persistent hrHPV infections. No single variant reached genome-wide significance (p < 5 X 10- 8). The top three variants associated with hrHPV infections were intronic variants clustered in KLF12 (all OR: 7.06, p = 1.43 × 10- 6). The top variants associated with cervical hrHPV persistence were in DAP (OR: 6.86, p = 7.15 × 10- 8), NR5A2 (OR: 3.65, p = 2.03 × 10- 7) and MIR365-2 (OR: 7.71, p = 2.63 × 10- 7) gene regions. CONCLUSIONS This exploratory GWAS yielded suggestive candidate risk loci for cervical hrHPV infection and persistence. The identified loci have biological annotation and functional data supporting their role in hrHPV infection and persistence. Given our limited sample size, larger discovery and replication studies are warranted to further characterize the reported associations.
Collapse
Affiliation(s)
- Sally N Adebamowo
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, 660 West Redwood Street, Howard Hall, Room 119, Baltimore, MD, 21201, USA.
- University of Maryland Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Adebowale A Adeyemo
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Charles N Rotimi
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Olayinka Olaniyan
- Department of Obstetrics and Gynecology, National Hospital Abuja, Abuja, Nigeria
| | - Richard Offiong
- Department of Obstetrics and Gynecology, University of Abuja Teaching Hospital, Abuja, Nigeria
| | - Clement A Adebamowo
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, 660 West Redwood Street, Howard Hall, Room 119, Baltimore, MD, 21201, USA
- University of Maryland Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
- Institute of Human Virology Nigeria, Abuja, Nigeria
| |
Collapse
|
20
|
Zhang T, Zhang K, Ji K, Zhang C, Jiang Y, Zhang Q, Tian Z, Wang X, Zhang M, Li X. microRNA-365 inhibits YAP through TLR4-mediated IRF3 phosphorylation and thereby alleviates gastric precancerous lesions. Cancer Cell Int 2020; 20:549. [PMID: 33292210 PMCID: PMC7664090 DOI: 10.1186/s12935-020-01578-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 09/25/2020] [Indexed: 12/18/2022] Open
Abstract
Background Gastric carcinoma (GC) is currently one of the most common malignant tumors of the digestive system, and gastric precancerous lesions play a vital role in studying the mechanism of GC. Multiple microRNAs (miRNAs) have been documented to be potential biomarkers to indicate progression of gastric precancerous lesions. In this study, we explained the anti-cancer effect of miR-365 in gastric precancerous lesions via regulation of the TLR4/IRF3/YAP/CDX2 axis. Methods miR-365, TLR4, CDX2 and IPF3 expression was determined in GC and atrophic gastritis tissues and cells. After transfection of shRNA and overexpression plasmids, in vitro experiments detected the alteration of cell viability, apoptosis and inflammatory factors. Bioinformatics analysis, Co-IP and dual luciferase reporter gene assay were conducted to evaluate the binding between miR-365 and TLR4 as well as IRF3 and YAP. Rat models were established to explore the effect of the miR-365 and TLR4 on gastric precancerous lesions. Results miR-365 was poorly expressed in GC and atrophic gastritis tissues and GC cell lines, while TLR4, CDX2 and IRF3 were overexpressed. Of note, miR-365 was indicated to target TLR4 and thereby suppressed cancer progression and increased hemoglobin content. Interestingly, silencing of TLR4 was accompanied by decreased IRF3 phosphorylation and reduced expression with less binding between CDX2 and IRF3. Downregulation of YAP resulted in declined CDX2 expression in cancer cells. Moreover, the inhibitory role of miR-365 was further confirmed in animal models. Conclusion Taken together, miR-365-mediated TLR4 inhibition reduces IRF3 phosphorylation and YAP-mediated CDX2 transcription to impede progression of gastric precancerous lesions.
Collapse
Affiliation(s)
- Tianqi Zhang
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, No. 16, Jiangsu Road, Shinan District, Qingdao, 266000, Shandong, People's Republic of China
| | - Kunpeng Zhang
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, No. 16, Jiangsu Road, Shinan District, Qingdao, 266000, Shandong, People's Republic of China
| | - Kaiyue Ji
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, No. 16, Jiangsu Road, Shinan District, Qingdao, 266000, Shandong, People's Republic of China
| | - Cuiping Zhang
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, No. 16, Jiangsu Road, Shinan District, Qingdao, 266000, Shandong, People's Republic of China
| | - Yueping Jiang
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, No. 16, Jiangsu Road, Shinan District, Qingdao, 266000, Shandong, People's Republic of China
| | - Qi Zhang
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, No. 16, Jiangsu Road, Shinan District, Qingdao, 266000, Shandong, People's Republic of China
| | - Zibin Tian
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, No. 16, Jiangsu Road, Shinan District, Qingdao, 266000, Shandong, People's Republic of China
| | - Xinyu Wang
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, No. 16, Jiangsu Road, Shinan District, Qingdao, 266000, Shandong, People's Republic of China
| | - Mengyuan Zhang
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, No. 16, Jiangsu Road, Shinan District, Qingdao, 266000, Shandong, People's Republic of China
| | - Xiaoyu Li
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, No. 16, Jiangsu Road, Shinan District, Qingdao, 266000, Shandong, People's Republic of China.
| |
Collapse
|
21
|
He P, Ma J, Liu Y, Deng H, Dong W. Hesperetin Promotes Cisplatin-Induced Apoptosis of Gastric Cancer In Vitro and In Vivo by Upregulating PTEN Expression. Front Pharmacol 2020; 11:1326. [PMID: 32973533 PMCID: PMC7482524 DOI: 10.3389/fphar.2020.01326] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 08/10/2020] [Indexed: 12/24/2022] Open
Abstract
As one of the most common malignant gastrointestinal tumors, gastric cancer (GC) has a high incidence and poor prognosis. Cisplatin (DDP) is often used as chemotherapy for advanced GC; however, the high incidence of drug resistance remains a problem. The use of several anti-tumor drugs as combined chemotherapy is an effective strategy. Hesperetin has anti-tumor ability via its pro-apoptotic effect on various human cancers, both in vitro and in vivo, with no significant toxicity. However, a combination of DDP and hesperetin in GC has not been reported. The present study aimed to investigate the in vitro and in vivo chemosensitization effect and mechanism of hesperetin-augmented DDP-induced apoptosis of GC. The proliferation of GC ty -60cells was inhibited significantly in a time and dose-dependent manner by combined treatment of DDP with hesperetin. Hesperetin markedly increased DDP-induced apoptosis of GC cell lines. In a xenograft tumor mouse model, markedly better tumor suppression was observed after treatment with DDP plus hesperetin compared with that of either agent alone. Additionally, the combination of DDP and hesperetin remarkably increased the expression levels of phosphatase and tensin homolog (PTEN) and Cytochrome C (Cyt C), and significantly decreased the levels of phosphorylated protein kinase B (p-AKT) and CyclinD1. DDP and hesperetin also induced significant increases in apoptosis inducing factor (AIF), BCL2 associated X, apoptosis regulator (BAX), cleaved caspase-9, and cleaved caspase-3, and decreased B-cell lymphoma 2 (BCL2), caspase-9, and caspase-3 levels. Thus, we demonstrated that hesperetin could inhibit the phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)/AKT signaling pathway and induce the mitochondrial pathway via upregulating PTEN expression, thereby significantly enhancing DDP’s anti-tumor effect on GC. Hesperetin is a potential chemotherapeutic agent for GC and merits further clinical investigation.
Collapse
Affiliation(s)
- Pengzhan He
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China.,Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China.,Key Laboratory of Hubei Province for Digestive System Disease, Wuhan, China
| | - Jingjing Ma
- Department of Geriatrics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yinghui Liu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China.,Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China.,Key Laboratory of Hubei Province for Digestive System Disease, Wuhan, China
| | - Huan Deng
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China.,Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China.,Key Laboratory of Hubei Province for Digestive System Disease, Wuhan, China
| | - Weiguo Dong
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
22
|
Liu J, Guo S, Zhang T, Ma X, Wu Z, Jiang K, Zhang X, Guo X, Deng G. MiR-505 as an anti-inflammatory regulator suppresses HMGB1/NF-κB pathway in lipopolysaccharide-mediated endometritis by targeting HMGB1. Int Immunopharmacol 2020; 88:106912. [PMID: 32829092 DOI: 10.1016/j.intimp.2020.106912] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 08/06/2020] [Accepted: 08/15/2020] [Indexed: 12/19/2022]
Abstract
Endometritis is characterized by severe inflammation and tissue damage. It is a common clinical disease that causes infertility due to infectious diseases of the reproductive system. MicroRNAs (miRNAs) are the current focus of research on the regulation of the inflammatory process and play a vital role in various inflammatory diseases. The highly conserved miR-505 regulates the mechanism of lipopolysaccharide (LPS) induced endometritis, but the extent to which pro-inflammatory genes are activated remains unclear. The results of this study showed that the expression of miR-505 was significantly down-regulated in mouse endometritis tissue and LPS-stimulated BEND cells. The study also showed that overexpression of miR-505 significantly suppressed the production of the pro-inflammatory cytokines IL-1β, IL-6 and TNF-α, and this effect was reversed by inhibiting the expression of miR-505. Moreover, miR-505 inhibited the expression of HMGB1 by targeting its 3'-UTR, thereby inhibiting the activation of HMGB1/NF-κB signalling. Taken together, the results of this study further confirmed that miR-505, as an anti-inflammatory agent, regulates the activation of the HMGB1/NF-κB signalling pathway through negative feedback.
Collapse
Affiliation(s)
- Junfeng Liu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, People's Republic of China; College of Animal Science, Tarim University, Alar, Xinjiang 843300, People's Republic of China
| | - Shuai Guo
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| | - Tao Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| | - Xiaofei Ma
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| | - Zhimin Wu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| | - Kangfeng Jiang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| | - Xiuping Zhang
- College of Animal Science, Tarim University, Alar, Xinjiang 843300, People's Republic of China
| | - Xuefeng Guo
- College of Animal Science, Tarim University, Alar, Xinjiang 843300, People's Republic of China.
| | - Ganzhen Deng
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, People's Republic of China.
| |
Collapse
|
23
|
Ashrafizadeh M, Najafi M, Ang HL, Moghadam ER, Mahabady MK, Zabolian A, Jafaripour L, Bejandi AK, Hushmandi K, Saleki H, Zarrabi A, Kumar AP. PTEN, a Barrier for Proliferation and Metastasis of Gastric Cancer Cells: From Molecular Pathways to Targeting and Regulation. Biomedicines 2020; 8:E264. [PMID: 32756305 PMCID: PMC7460532 DOI: 10.3390/biomedicines8080264] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/23/2020] [Accepted: 07/23/2020] [Indexed: 02/06/2023] Open
Abstract
Cancer is one of the life-threatening disorders that, in spite of excellent advances in medicine and technology, there is no effective cure for. Surgery, chemotherapy, and radiotherapy are extensively applied in cancer therapy, but their efficacy in eradication of cancer cells, suppressing metastasis, and improving overall survival of patients is low. This is due to uncontrolled proliferation of cancer cells and their high migratory ability. Finding molecular pathways involved in malignant behavior of cancer cells can pave the road to effective cancer therapy. In the present review, we focus on phosphatase and tensin homolog (PTEN) signaling as a tumor-suppressor molecular pathway in gastric cancer (GC). PTEN inhibits the PI3K/Akt pathway from interfering with the migration and growth of GC cells. Its activation leads to better survival of patients with GC. Different upstream mediators of PTEN in GC have been identified that can regulate PTEN in suppressing growth and invasion of GC cells, such as microRNAs, long non-coding RNAs, and circular RNAs. It seems that antitumor agents enhance the expression of PTEN in overcoming GC. This review focuses on aforementioned topics to provide a new insight into involvement of PTEN and its downstream and upstream mediators in GC. This will direct further studies for evaluation of novel signaling networks and their targeting for suppressing GC progression.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz 5166616471, Iran;
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah 6715847141, Iran;
| | - Hui Li Ang
- Cancer Science Institute of Singapore and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore;
| | - Ebrahim Rahmani Moghadam
- Department of Anatomical Sciences, School of Medicine, Student Research Committee, Shiraz University of Medical Sciences, Shiraz 7134814336, Iran;
- Kazerun Health Technology Incubator, Shiraz University of Medical Sciences, Shiraz 6461665145, Iran
| | - Mahmood Khaksary Mahabady
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan 8715988141, Iran;
| | - Amirhossein Zabolian
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran 1916893813, Iran; (A.Z.); (A.K.B.); (H.S.)
| | - Leila Jafaripour
- Department of Anatomy, School of Medicine, Dezful University of Medical Sciences, Dezful 3419759811, Iran;
| | - Atefe Kazemzade Bejandi
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran 1916893813, Iran; (A.Z.); (A.K.B.); (H.S.)
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology & Zoonoses, Faculty of Veterinary Medicine, University of Tehran, Tehran 1417414418, Iran;
| | - Hossein Saleki
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran 1916893813, Iran; (A.Z.); (A.K.B.); (H.S.)
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla 34956, Istanbul, Turkey
- Center of Excellence for Functional Surfaces and Interfaces (EFSUN), Faculty of Engineering and Natural Sciences, Sabanci University, Tuzla 34956, Istanbul, Turkey
| | - Alan Prem Kumar
- Cancer Science Institute of Singapore and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore;
| |
Collapse
|
24
|
miR193b Promotes Apoptosis of Gastric Cancer Cells via Directly Mediating the Akt Pathway. BIOMED RESEARCH INTERNATIONAL 2020; 2020:2863236. [PMID: 32596290 PMCID: PMC7273449 DOI: 10.1155/2020/2863236] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 04/25/2020] [Indexed: 12/25/2022]
Abstract
Gastric cancer (GC) is one of the most common and fatal malignancies worldwide. MicroRNAs (miRNAs) play a critical role in tumor initiation, proliferation, and metastasis of gastric cancer. miR193b has been identified as a tumor suppressor in a variety of tumor types; however, its role in gastric cancer is yet to be determined. Here, we found a significant downregulation of miR193b expression in both human gastric cancer tissues (p < 0.05) and human gastric cancer cell lines (p < 0.01). Furthermore, the expression level of miR193b correlated with the tumor type, tumor size, and clinical stage (p < 0.05). In vitro, miR193b overexpression inhibited cell survival and induced apoptosis in GC cell lines, indicating that miR193b plays a role in the development of gastric cancer. KRAS was verified as the target of miR193b, and KRAS overexpression attenuated miR193b-induced apoptosis (p < 0.05). Moreover, we found that the Akt pathway negatively regulated miR193b, also affecting apoptosis. Further analyses indicated that PIK3CA mutation and KRAS amplification are two mutually exclusive pathways (p < 0.01), and we hypothesize that both two pathways could result in the carcinogenic overactivation of KRAS. Thus, our results suggest that the Akt-miR193b-KRAS axis may act as a mechanism affecting apoptosis in gastric cancer cells.
Collapse
|
25
|
Rama AR, Perazzoli G, Cabeza L, Mesas C, Quiñonero F, García-Pinel B, Vélez C. Novel MicroRNA Sponges to Specifically Modulate Gene Expression in Colon Cancer Cells. Nucleic Acid Ther 2020; 30:325-334. [PMID: 32429773 DOI: 10.1089/nat.2020.0861] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
MicroRNA (miRNA) sponges allow the selective blockade of a complete family of associated miRNAs, which induce post-transcriptional gene silencing in their target through binding to 3'UTR mRNA. miRNA-365 and miRNA-145 are downregulated in colorectal cancer (CRC) but not in healthy tissues. Based on this, we constructed two vectors by inserting miRNA sponges (one for miRNA-365 and other for miRNA-145), and used enhanced green fluorescent protein (EGFP) as a 3'UTR reporter gene to analyze the ability of each sponge to catch its respective miRNA. Quantitative polymerase chain reaction (qPCR) results corroborated that the expression levels of both miRNAs were lower in CRC cell lines than in normal colon cell lines. Flow cytometry analysis revealed a decrease of the EGFP expression levels in the cell lines transfected with both sponges, being higher on the normal cell line while CRC cell lines presented a minimal decline. Also, this decrease was inversely proportional to the levels of expression of both miRNAs obtained by qPCR. These results were corroborated by fluorescence microscopy, showing a similar decrease in fluorescence. We propose a new vector system to carry in a specific way the expression of genes to CRC cells without affecting healthy cells, preventing damage to healthy tissues.
Collapse
Affiliation(s)
- Ana R Rama
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, Spain.,Institute of Biosanitary Research from Granada (ibs. GRANADA), Granada, Spain.,Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, Spain
| | - Gloria Perazzoli
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, Spain.,Institute of Biosanitary Research from Granada (ibs. GRANADA), Granada, Spain
| | - Laura Cabeza
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, Spain.,Institute of Biosanitary Research from Granada (ibs. GRANADA), Granada, Spain.,Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, Spain
| | - Cristina Mesas
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, Spain.,Institute of Biosanitary Research from Granada (ibs. GRANADA), Granada, Spain.,Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, Spain
| | - Francisco Quiñonero
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, Spain.,Institute of Biosanitary Research from Granada (ibs. GRANADA), Granada, Spain.,Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, Spain
| | - Beatriz García-Pinel
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, Spain.,Institute of Biosanitary Research from Granada (ibs. GRANADA), Granada, Spain.,Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, Spain
| | - Celia Vélez
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, Spain.,Institute of Biosanitary Research from Granada (ibs. GRANADA), Granada, Spain.,Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, Spain
| |
Collapse
|
26
|
Du J, Liang Y, Li J, Zhao JM, Wang ZN, Lin XY. Gastric Cancer Cell-Derived Exosomal microRNA-23a Promotes Angiogenesis by Targeting PTEN. Front Oncol 2020; 10:326. [PMID: 32232005 PMCID: PMC7082307 DOI: 10.3389/fonc.2020.00326] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 02/25/2020] [Indexed: 12/24/2022] Open
Abstract
Hypoxia-exposed lung cancer-released exosomal microRNA-23a (miR-23a) has been shown to enhance angiogenesis as well as vascular permeability, contributing to the close correlation between exosomal miR-23a and tumorigenesis. The current study aimed to investigate whether gastric cancer (GC) cell-derived exosomal miR-23a could induce angiogenesis and to elucidate the potential mechanisms associated with the process. Differentially expressed miRNAs in GC were initially screened from the Gene Expression Omnibus database. Target genes were selected following miRNA-mRNA prediction and subsequently verified by dual luciferase reporter assay. RT-qPCR was conducted to detect miR-23a and PTEN expression in GC tissues, cells and exosomes. Human umbilical venous endothelial cells (HUVECs) were co-cultured with GC cell-derived exosomes to assess the angiogenesis mediated by exosomes in vitro. Additionally, PTEN was overexpressed in HUVECs to analyze the mechanism by which miR-23a regulates angiogenesis. miR-23a was highly expressed in GC tissues and cells and GC cell-derived exosomes. Angiogenesis was promoted by the co-culture of HUVECs and GC cells-derived exosomes, as evidenced by the increased expression of VEGF but decreased expression of TSP-1. PTEN was targeted by miR-23a and was lowly expressed in GC tissues. In a co-culture system, miR-23a carried by GC cells-derived exosomes promoted angiogenesis via the repression of PTEN. Collectively, GC cell-derived exosomal miR-23a could promote angiogenesis and provide blood supply for growth of GC cells. This study contributes to advancement of miRNA-targeted therapeutics.
Collapse
Affiliation(s)
- Jiang Du
- Department of Pathology, The First Affiliated Hospital and College of Basic Medical Science, China Medical University, Shenyang, China
| | - Yuan Liang
- Medical Oncology Department of Thoracic Cancer (2), Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Ji Li
- Department of Pathology, The First Affiliated Hospital and College of Basic Medical Science, China Medical University, Shenyang, China
| | - Jin-Ming Zhao
- Department of Pathology, The First Affiliated Hospital and College of Basic Medical Science, China Medical University, Shenyang, China
| | - Zhen-Ning Wang
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Xu-Yong Lin
- Department of Pathology, The First Affiliated Hospital and College of Basic Medical Science, China Medical University, Shenyang, China
| |
Collapse
|
27
|
Zhou W, Wu J, Zhu Y, Meng Z, Liu X, Liu S, Ni M, Jia S, Zhang J, Guo S. Study on the mechanisms of compound Kushen injection for the treatment of gastric cancer based on network pharmacology. BMC Complement Med Ther 2020; 20:6. [PMID: 32020871 PMCID: PMC7076865 DOI: 10.1186/s12906-019-2787-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 12/04/2019] [Indexed: 12/24/2022] Open
Abstract
Background As an effective prescription for gastric cancer (GC), Compound Kushen Injection (CKI) has been widely used even though few molecular mechanism analyses have been carried out. Methods In this study, we identified 16 active ingredients and 60 GC target proteins. Then, we established a compound-predicted target network and a GC target protein-protein interaction (PPI) network by Cytoscape 3.5.1 and systematically analyzed the potential targets of CKI for the treatment of GC. Finally, molecular docking was applied to verify the key targets. In addition, we analyzed the mechanism of action of the predicted targets by Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) analyses. Results The results showed that the potential targets, including CCND1, PIK3CA, AKT1, MAPK1, ERBB2, and MMP2, are the therapeutic targets of CKI for the treatment of GC. Functional enrichment analysis indicated that CKI has a therapeutic effect on GC by synergistically regulating some biological pathways, such as the cell cycle, pathways in cancer, the PI3K-AKT signaling pathway, the mTOR signaling pathway, and the FoxO signaling pathway. Moreover, molecular docking simulation indicated that the compounds had good binding activity to PIK3CA, AKT1, MAPK1, ERBB2, and MMP2 in vivo. Conclusion This research partially highlighted the molecular mechanism of CKI for the treatment of GC, which has great potential in the identification of the effective compounds in CKI and biomarkers to treat GC.
Collapse
Affiliation(s)
- Wei Zhou
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, No. 11 of North Three-ring East Road, Chao Yang District, Beijing, China
| | - Jiarui Wu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, No. 11 of North Three-ring East Road, Chao Yang District, Beijing, China.
| | - Yingli Zhu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, No. 11 of North Three-ring East Road, Chao Yang District, Beijing, China
| | - Ziqi Meng
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, No. 11 of North Three-ring East Road, Chao Yang District, Beijing, China
| | - Xinkui Liu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, No. 11 of North Three-ring East Road, Chao Yang District, Beijing, China
| | - Shuyu Liu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, No. 11 of North Three-ring East Road, Chao Yang District, Beijing, China
| | - Mengwei Ni
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, No. 11 of North Three-ring East Road, Chao Yang District, Beijing, China
| | - Shanshan Jia
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, No. 11 of North Three-ring East Road, Chao Yang District, Beijing, China
| | - Jingyuan Zhang
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, No. 11 of North Three-ring East Road, Chao Yang District, Beijing, China
| | - Siyu Guo
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, No. 11 of North Three-ring East Road, Chao Yang District, Beijing, China
| |
Collapse
|
28
|
Wei W, Mo X, Yan L, Huang M, Yang Y, Jin Q, Zhong H, Cao W, Wu K, Wu L, Li Z, Wang T, Qin Y, Chen J. Circular RNA Profiling Reveals That circRNA_104433 Regulates Cell Growth by Targeting miR-497-5p in Gastric Cancer. Cancer Manag Res 2020; 12:15-30. [PMID: 32021419 PMCID: PMC6954096 DOI: 10.2147/cmar.s219307] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 11/28/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The role and mechanism of hsa_circRNA_104433 in gastric cancer (GC) are further elucidated. MATERIALS AND METHODS CircRNA_104433 was selected by circRNA microarrays and GEO database. qRT-PCR was used to analyze the expression of circRNA_104433 in GC. The role of circRNA_104433 in GC cells was evaluated based on cell cycle progression, cell proliferation, cell apoptosis, and tumor xenograft experiment assay. To explore the mechanisms of circRNA_104433 in GC TCGA database, STRING version, qRT-PCR and luciferase assay were performed. Furthermore, the prognostic value of CDC25A in GC was determined based on the GEO database. RESULTS The level of circRNA_104433 showed upregulation in GC tissues, and the expression of it showed a positive correlation with the degree of differentiation and the size of the tumor. Knockdown of circRNA_104433 inhibited cell cycle transition, and cell proliferation, while promoted cell apoptosis in GC. CircRNA_104433 directly binds to miR-497-5p, which directly regulates CDC25A. The median survival period of GC patients with high expression levels of CDC25A was 21.3 months, which was shorter than those with low group expression of CDC25A (35.9 months). The cell cycle proteins CDK1, CDK2, CCNB1, PKMYT1, CDC20, CHEK1 and CDC25A were coexpressed with CDC25A. CONCLUSION These findings suggested that knockdown of circRNA_104433 expression suppressed tumor development in GC.
Collapse
Affiliation(s)
- Weiyuan Wei
- Department of Gastrointestinal Surgery, Affiliated Tumor Hospital, Guangxi Medical University, Nanning530021, People’s Republic of China
| | - Xianwei Mo
- Department of Gastrointestinal Surgery, Affiliated Tumor Hospital, Guangxi Medical University, Nanning530021, People’s Republic of China
| | - Linhai Yan
- Department of Gastrointestinal Surgery, Affiliated Tumor Hospital, Guangxi Medical University, Nanning530021, People’s Republic of China
| | - Mingwei Huang
- Department of Gastrointestinal Surgery, Affiliated Tumor Hospital, Guangxi Medical University, Nanning530021, People’s Republic of China
| | - Yang Yang
- Department of Gastrointestinal Surgery, Affiliated Tumor Hospital, Guangxi Medical University, Nanning530021, People’s Republic of China
| | - Qinwen Jin
- Department of Gastrointestinal Surgery, Affiliated Tumor Hospital, Guangxi Medical University, Nanning530021, People’s Republic of China
| | - Huage Zhong
- Department of Gastrointestinal Surgery, Affiliated Tumor Hospital, Guangxi Medical University, Nanning530021, People’s Republic of China
| | - Wenlong Cao
- Departments of Surgery, The First Affiliated Hospital, Guangxi Medical University, Nanning530021, People’s Republic of China
| | - Kun Wu
- Departments of Surgery, The First Affiliated Hospital, Guangxi Medical University, Nanning530021, People’s Republic of China
| | - Liucheng Wu
- Department of Gastrointestinal Surgery, Affiliated Tumor Hospital, Guangxi Medical University, Nanning530021, People’s Republic of China
| | - Zhao Li
- Department of Gastrointestinal Surgery, Affiliated Tumor Hospital, Guangxi Medical University, Nanning530021, People’s Republic of China
| | - Tingan Wang
- Department of Gastrointestinal Surgery, Affiliated Tumor Hospital, Guangxi Medical University, Nanning530021, People’s Republic of China
| | - Yuzhou Qin
- Department of Gastrointestinal Surgery, Affiliated Tumor Hospital, Guangxi Medical University, Nanning530021, People’s Republic of China
| | - Jiansi Chen
- Department of Gastrointestinal Surgery, Affiliated Tumor Hospital, Guangxi Medical University, Nanning530021, People’s Republic of China
| |
Collapse
|
29
|
Hou B, Li W, Li J, Ma J, Xia P, Liu Z, Zeng Q, Zhang X, Chang D. Tumor suppressor LHPP regulates the proliferation of colorectal cancer cells via the PI3K/AKT pathway. Oncol Rep 2019; 43:536-548. [PMID: 31894339 PMCID: PMC6967159 DOI: 10.3892/or.2019.7442] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 11/20/2019] [Indexed: 12/15/2022] Open
Abstract
Colorectal cancer (CRC) is the third most commonly diagnosed cancer and the fourth leading cause of cancer-related mortality worldwide. Thus, identification of the mechanisms involved in the progression of CRC has become a crucial element of facilitating early CRC diagnosis and targeted therapy for patients with advanced CRC. Currently, Phospholysine phosphohistidine inorganic pyrophosphate phosphatase (LHPP), a type of histidine phosphatase protein, has been confirmed as a tumor suppressor in hepatocellular carcinoma (HCC) and cervical cancer. However, the functions and molecular mechanisms underlying LHPP in CRC remain undefined. The present study revealed that dysregulation of LHPP was frequently observed in CRC tissues and was positively correlated with tumor severity and poor prognosis. Functional experiments demonstrated that overexpression of LHPP impeded CRC cell growth and proliferation in vitro, and was associated with a change in p53 expression and PI3K/AKT activity. In contrast, silencing of LHPP significantly promoted cell growth and proliferation by modulating the PI3K/AKT signaling pathway. Notably, the anti-CRC effects of LHPP were also observed in nude mouse in vivo experiments. Overall, the data obtained in the present study suggested that LHPP may be exploited as a diagnostic and prognostic candidate for patients with CRC.
Collapse
Affiliation(s)
- Bin Hou
- Department of Surgical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Wenhan Li
- Department of Surgical Oncology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 10068, P.R. China
| | - Jianhui Li
- Department of Surgical Oncology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 10068, P.R. China
| | - Jia Ma
- Department of Surgical Oncology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 10068, P.R. China
| | - Peng Xia
- Department of Surgical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Zhao Liu
- Department of Surgical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Qingnuo Zeng
- Department of Surgical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Xin Zhang
- Department of Surgical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Dongmin Chang
- Department of Surgical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
30
|
Liu J, Guo S, Jiang K, Zhang T, Zhiming W, Yaping Y, Jing Y, Shaukat A, Deng G. miR-488 mediates negative regulation of the AKT/NF-κB pathway by targeting Rac1 in LPS-induced inflammation. J Cell Physiol 2019; 235:4766-4777. [PMID: 31674024 DOI: 10.1002/jcp.29354] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 10/07/2019] [Indexed: 12/13/2022]
Abstract
Endometritis is an inflammatory change in the structure of the endometrium due to various causes and is a common cause of infertility. Studies have confirmed that microRNAs (miRNAs) play a key regulatory role in various inflammatory diseases. However, the miRNA-mediated mechanism of endometrial inflammation induced by lipopolysaccharides (LPS) remains unclear. In this study, real-time quantitative polymerase chain reaction, Western blot analysis, immunofluorescence and Rac family small GTPase 1 (Rac1) interference were used to reveal the overexpression of miR-488 in the LPS-induced bovine uterus, and the effect of protein kinase B κ-light chain enhancement of the nuclear factor-activated B cells (AKT/NF-κB) pathway in intimal epithelial cells. The results showed that the expression of inflammatory cytokines such as interleukin-1β (IL-1β), IL-6, and tumor necrosis factor-α in the experimental group was significantly lower than that in the control group when miR-488 was overexpressed. Similar results were observed in the expression levels of p-AKT, p-IKK, and p-p65 proteins. In addition, the dual-luciferase reporter system confirmed that miRNA-488 may directly target the 3'-untranslated region of Rac1. In turn, the expression of Rac1 was inhibited. Moreover, the nuclear translocation of NF-κB was inhibited, and meanwhile, the accumulation of reactive oxygen species (ROS) in the cells was reduced. Thus, we provide basic data for the negative regulation of miR-488 in LPS-induced inflammation by inhibiting ROS production and the AKT/NF-kB pathway in intimal epithelial cells.
Collapse
Affiliation(s)
- Junfeng Liu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,College of Animal Science, Tarim University, Alar, Xinjiang, China
| | - Shuai Guo
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Kangfeng Jiang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Tao Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Wu Zhiming
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yang Yaping
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yang Jing
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Aftab Shaukat
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Ganzhen Deng
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
31
|
Liu JC, Granieri L, Shrestha M, Wang DY, Vorobieva I, Rubie EA, Jones R, Ju Y, Pellecchia G, Jiang Z, Palmerini CA, Ben-David Y, Egan SE, Woodgett JR, Bader GD, Datti A, Zacksenhaus E. Identification of CDC25 as a Common Therapeutic Target for Triple-Negative Breast Cancer. Cell Rep 2019; 23:112-126. [PMID: 29617654 PMCID: PMC9357459 DOI: 10.1016/j.celrep.2018.03.039] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 01/31/2018] [Accepted: 03/11/2018] [Indexed: 12/18/2022] Open
Abstract
CDK4/6 inhibitors are effective against cancer cells expressing the tumor suppressor RB1, but not RB1-deficient cells, posing the challenge of how to target RB1 loss. In triple-negative breast cancer (TNBC), RB1 and PTEN are frequently inactivated together with TP53. We performed kinome/phosphatase inhibitor screens on primary mouse Rb/p53-, Pten/p53-, and human RB1/PTEN/TP53-deficient TNBC cell lines and identified CDC25 phosphatase as a common target. Pharmacological or genetic inhibition of CDC25 suppressed growth of RB1-deficient TNBC cells that are resistant to combined CDK4/6 plus CDK2 inhibition. Minimal cooperation was observed in vitro between CDC25 antagonists and CDK1, CDK2, or CDK4/6 inhibitors, but strong synergy with WEE1 inhibition was apparent. In accordance with increased PI3K signaling following long-term CDC25 inhibition, CDC25 and PI3K inhibitors effectively synergized to suppress TNBC growth both in vitro and in xenotransplantation models. These results provide a rationale for the development of CDC25-based therapies for diverse RB1/PTEN/TP53-deficient and -proficient TNBCs. Liu et al. report that inhibition of the protein phosphatase CDC25 kills diverse triple-negative breast cancer (TNBC) cells. Moreover, CDC25 antagonists cooperate with other drugs, such as PI3K inhibitors, to efficiently suppress growth of human TNBC engrafted into mice.
Collapse
Affiliation(s)
- Jeff C Liu
- Toronto General Research Institute - University Health Network, 67 College Street, Toronto, ON, Canada M5G 2M1
| | - Letizia Granieri
- Toronto General Research Institute - University Health Network, 67 College Street, Toronto, ON, Canada M5G 2M1; Department of Agriculture, Food, and Environmental Sciences, University of Perugia, Perugia, Italy
| | - Mariusz Shrestha
- Toronto General Research Institute - University Health Network, 67 College Street, Toronto, ON, Canada M5G 2M1; Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Dong-Yu Wang
- Toronto General Research Institute - University Health Network, 67 College Street, Toronto, ON, Canada M5G 2M1
| | - Ioulia Vorobieva
- Toronto General Research Institute - University Health Network, 67 College Street, Toronto, ON, Canada M5G 2M1; Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Elizabeth A Rubie
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, 600 University Avenue, Toronto, ON, Canada
| | - Rob Jones
- Toronto General Research Institute - University Health Network, 67 College Street, Toronto, ON, Canada M5G 2M1
| | - YoungJun Ju
- Toronto General Research Institute - University Health Network, 67 College Street, Toronto, ON, Canada M5G 2M1
| | - Giovanna Pellecchia
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada; The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON, Canada
| | - Zhe Jiang
- Toronto General Research Institute - University Health Network, 67 College Street, Toronto, ON, Canada M5G 2M1
| | - Carlo A Palmerini
- Department of Agriculture, Food, and Environmental Sciences, University of Perugia, Perugia, Italy
| | - Yaacov Ben-David
- The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang, Guizhou 550014, China; State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, Guizhou 550025, China
| | - Sean E Egan
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada; Program in Cell Biology, The Peter Gilgan Center for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
| | - James R Woodgett
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, 600 University Avenue, Toronto, ON, Canada
| | - Gary D Bader
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Alessandro Datti
- Department of Agriculture, Food, and Environmental Sciences, University of Perugia, Perugia, Italy; Network Biology Collaborative Centre, SMART Laboratory for High-Throughput Screening Programs, Mount Sinai Hospital, Toronto, ON, Canada
| | - Eldad Zacksenhaus
- Toronto General Research Institute - University Health Network, 67 College Street, Toronto, ON, Canada M5G 2M1; Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON, Canada; Department of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
32
|
Xie J, Lin J, Wei M, Teng Y, He Q, Yang G, Yang X. Sustained Akt signaling in articular chondrocytes causes osteoarthritis via oxidative stress-induced senescence in mice. Bone Res 2019; 7:23. [PMID: 31646013 PMCID: PMC6804644 DOI: 10.1038/s41413-019-0062-y] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 05/25/2019] [Accepted: 06/13/2019] [Indexed: 12/13/2022] Open
Abstract
Osteoarthritis (OA) is an age-related disorder that is strongly associated with chondrocyte senescence. The causal link between disruptive PTEN/Akt signaling and chondrocyte senescence and the underlying mechanism are unclear. In this study, we found activated Akt signaling in human OA cartilage as well as in a mouse OA model with surgical destabilization of the medial meniscus. Genetic mouse models mimicking sustained Akt signaling in articular chondrocytes via PTEN deficiency driven by either Col2a1-Cre or Col2a1-CreERT2 developed OA, whereas restriction of Akt signaling reversed the OA phenotypes in PTEN-deficient mice. Mechanistically, prolonged activation of Akt signaling caused an accumulation of reactive oxygen species and triggered chondrocyte senescence as well as a senescence-associated secretory phenotype, whereas chronic administration of the antioxidant N-acetylcysteine suppressed chondrocyte senescence and mitigated OA progression in PTEN-deficient mice. Therefore, inhibition of Akt signaling by PTEN is required for the maintenance of articular cartilage. Disrupted Akt signaling in articular chondrocytes triggers oxidative stress-induced chondrocyte senescence and causes OA.
Collapse
Affiliation(s)
- Jing Xie
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206 China
| | - Jingting Lin
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206 China
| | - Min Wei
- 2Department of Orthopaedics, Chinese PLA General Hospital, Beijing, 100853 China
| | - Yan Teng
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206 China
| | - Qi He
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206 China
| | - Guan Yang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206 China
| | - Xiao Yang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206 China
| |
Collapse
|
33
|
Xu G, Li N, Zhang Y, Zhang J, Xu R, Wu Y. MicroRNA-383-5p inhibits the progression of gastric carcinoma via targeting HDAC9 expression. ACTA ACUST UNITED AC 2019; 52:e8341. [PMID: 31365693 PMCID: PMC6668961 DOI: 10.1590/1414-431x20198341] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Accepted: 05/07/2019] [Indexed: 12/15/2022]
Abstract
MicroRNAs (miRNAs), as post-transcriptional regulators, have been reported to be involved in the initiation and progression of various types of cancer, including gastric cancer (GC). The present study aimed to investigate the role of miR-383-5p in gastric carcinogenesis. Cell viability was analyzed using CCK-8 kit. Annexin V-fluorescein isothiocyanate/propidium iodide double staining was used to evaluate cell apoptosis. The expression levels of miR-383-5p and histone deacetylase 9 (HDAC9) mRNA in GC tissues and cell lines were analyzed using RT-qPCR. The protein expression of HDAC9 was detected by western blotting. We found that HDAC9 was up-regulated and miR-383-5p was down-regulated in GC tissues and cell lines. High HDAC9 expression or low miR-383-5p expression was closely related to poor prognosis and metastasis in GC patients. HDAC9 knockout or miR-383-5p mimics led to growth inhibition and increased apoptosis in AGS and SGC-7901 cells. More importantly, we validated that miR-383-5p as a post-transcriptional regulator inhibited HDAC9 expression and was inversely correlated with HDAC9 expression in GC tissues. miR-383-5p had the opposite effects to HDAC9 in gastric carcinogenesis. miR-383-5p played an important role in gastric carcinogenesis, and it is one of the important mechanisms to regulate oncogenic HDAC9 in GC, which might be helpful in the development of novel therapeutic strategies for the treatment of GC.
Collapse
Affiliation(s)
- Gang Xu
- Department of Oncology, Chinese PLA No.148 Hospital, Zibo, Shandong, China
| | - Na Li
- Department of Oncology, Chinese PLA No.148 Hospital, Zibo, Shandong, China
| | - Yan Zhang
- Department of Oncology, Chinese PLA No.148 Hospital, Zibo, Shandong, China
| | - Jinbiao Zhang
- Department of Oncology, Chinese PLA No.148 Hospital, Zibo, Shandong, China
| | - Rui Xu
- Department of Oncology, Chinese PLA No.148 Hospital, Zibo, Shandong, China
| | - Yanling Wu
- Department of Oncology, Chinese PLA No.148 Hospital, Zibo, Shandong, China
| |
Collapse
|
34
|
Li X, Nie C, Tian B, Tan X, Han W, Wang J, Jin Y, Li Y, Guan X, Hong A, Chen X. miR-671-5p Blocks The Progression Of Human Esophageal Squamous Cell Carcinoma By Suppressing FGFR2. Int J Biol Sci 2019; 15:1892-1904. [PMID: 31523191 PMCID: PMC6743296 DOI: 10.7150/ijbs.32429] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 06/02/2019] [Indexed: 12/31/2022] Open
Abstract
Esophageal cancer is the eighth most common malignant tumor worldwide, of which esophageal squamous cell carcinoma (ESCC) is the dominant histological subtype. A drug shortage for ESCC therapy triggered us to explore the roles of fibroblast growth factor receptor 2 (FGFR2) and its upstream regulator miR-671-5p in ESCC progression. We compared the levels of FGFR2 and miR-671-5p between human ESCC tissues and their matched normal esophageal tissues and found an association between higher levels of FGFR2 and lower levels of miR-671-5p in ESCC tissues. High levels of FGFR2 resulted in the activation of the ERK and AKT pathways and a promotion of ESCC progression. High levels of miR-671-5p specifically reduced the expression of FGFR2 and suppressed ESCC progression in both in vitro and in vivo models. Therefore, suppressing FGFR2 and enhancing miR-671-5p expression may be the right approaches for ESCC therapy.
Collapse
Affiliation(s)
- Xiaoyan Li
- Institute of Biomedicine & Department of cell Biology, Jinan University, Guangzhou, Guangdong, 510632, P. R. China.,National Engineering Research Center of Genetic Medicine, Guangzhou, Guangdong, 510632, P. R. China.,Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou, Guangdong, 510632, P. R. China
| | - Changjun Nie
- Institute of Biomedicine & Department of cell Biology, Jinan University, Guangzhou, Guangdong, 510632, P. R. China.,National Engineering Research Center of Genetic Medicine, Guangzhou, Guangdong, 510632, P. R. China.,Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou, Guangdong, 510632, P. R. China
| | - Baoqing Tian
- Institute of Biomedicine & Department of cell Biology, Jinan University, Guangzhou, Guangdong, 510632, P. R. China.,National Engineering Research Center of Genetic Medicine, Guangzhou, Guangdong, 510632, P. R. China.,Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou, Guangdong, 510632, P. R. China
| | - Xuan Tan
- Institute of Biomedicine & Department of cell Biology, Jinan University, Guangzhou, Guangdong, 510632, P. R. China.,National Engineering Research Center of Genetic Medicine, Guangzhou, Guangdong, 510632, P. R. China.,Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou, Guangdong, 510632, P. R. China
| | - Wei Han
- Institute of Biomedicine & Department of cell Biology, Jinan University, Guangzhou, Guangdong, 510632, P. R. China.,National Engineering Research Center of Genetic Medicine, Guangzhou, Guangdong, 510632, P. R. China.,Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou, Guangdong, 510632, P. R. China
| | - Jiakang Wang
- Cancer Center of Guangzhou Medical University, Guangzhou, Guangdong, 510090, P. R. China
| | - Yuan Jin
- Institute of Biomedicine & Department of cell Biology, Jinan University, Guangzhou, Guangdong, 510632, P. R. China.,National Engineering Research Center of Genetic Medicine, Guangzhou, Guangdong, 510632, P. R. China.,Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou, Guangdong, 510632, P. R. China
| | - Yadan Li
- Institute of Biomedicine & Department of cell Biology, Jinan University, Guangzhou, Guangdong, 510632, P. R. China.,National Engineering Research Center of Genetic Medicine, Guangzhou, Guangdong, 510632, P. R. China.,Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou, Guangdong, 510632, P. R. China
| | - Xinyuan Guan
- Department of Clinical Oncology, University of Hong Kong, Hong Kong, China
| | - An Hong
- Institute of Biomedicine & Department of cell Biology, Jinan University, Guangzhou, Guangdong, 510632, P. R. China.,National Engineering Research Center of Genetic Medicine, Guangzhou, Guangdong, 510632, P. R. China.,Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou, Guangdong, 510632, P. R. China
| | - Xiaojia Chen
- Institute of Biomedicine & Department of cell Biology, Jinan University, Guangzhou, Guangdong, 510632, P. R. China.,National Engineering Research Center of Genetic Medicine, Guangzhou, Guangdong, 510632, P. R. China.,Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou, Guangdong, 510632, P. R. China
| |
Collapse
|
35
|
Han Q, Cheng P, Yang H, Liang H, Lin F. Altered expression of microRNA-365 is related to the occurrence and development of non-small-cell lung cancer by inhibiting TRIM25 expression. J Cell Physiol 2019; 234:22321-22330. [PMID: 31099423 DOI: 10.1002/jcp.28798] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 03/01/2019] [Accepted: 03/06/2019] [Indexed: 12/11/2022]
Abstract
The purpose of this current study is to elucidate whether altered microRNA-365 (miR-365) has an association with the initiation and development of non-small-cell lung cancer (NSCLC) by targeting TRIM25 expression. The expression of miR-365 and TRIM25 in NSCLC tissues, adjacent normal tissues, and NSCLC cell lines were detected. The relationship between miR-365 expression and TRIM25 with the clinicopathological characteristics of NSCLC was analyzed. The putative binding site between miR-365 and TRIM25 was determined by luciferase activity assay. miR-365 inhibitors and miR-365 mimics were transfected to human NSCLC A549 cells, and the cell viability was detected by cell counting kit-8 assay; flow cytometry was carried out to determine cell cycle and apoptosis rate. Poorly expressed miR-365 and overexpressed TRIM25 was found in NSCLC tissues. TRIM25 was determined as a target gene of miR-365. The miR-365 and TRIM25 expression were related to the clinicopathological features of NSCLC, such as pathological classification, differentiation degree, TNM stage as well as lymph node metastasis. miR-365 suppressed the expression of TRIM25 and elevated the expression of the proapoptotic protein in NSCLC cells. Our study demonstrates that altered expression of miR-365 has a close association with the occurrence and development of NSCLC by inhibiting TRIM25 expression.
Collapse
Affiliation(s)
- Qian Han
- Department of Radiotherapy, Henan Provincial People's Hospital, Zhengzhou, Henan, People's Republic of China
| | - Peng Cheng
- Department of Radiotherapy, Henan Provincial People's Hospital, Zhengzhou, Henan, People's Republic of China
| | - Hongjie Yang
- Department of Radiotherapy, Henan Provincial People's Hospital, Zhengzhou, Henan, People's Republic of China
| | - Hengpo Liang
- Department of Radiotherapy, Henan Provincial People's Hospital, Zhengzhou, Henan, People's Republic of China
| | - Fengchun Lin
- Department of Radiotherapy, Henan Provincial People's Hospital, Zhengzhou, Henan, People's Republic of China
| |
Collapse
|
36
|
MicroRNA Dysregulation in Cutaneous Squamous Cell Carcinoma. Int J Mol Sci 2019; 20:ijms20092181. [PMID: 31052530 PMCID: PMC6540078 DOI: 10.3390/ijms20092181] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Revised: 04/15/2019] [Accepted: 04/29/2019] [Indexed: 02/07/2023] Open
Abstract
Cutaneous squamous cell carcinoma (CSCC) is the second most frequent cancer in humans and it can be locally invasive and metastatic to distant sites. MicroRNAs (miRNAs or miRs) are endogenous, small, non-coding RNAs of 19–25 nucleotides in length, that are involved in regulating gene expression at a post-transcriptional level. MicroRNAs have been implicated in diverse biological functions and diseases. In cancer, miRNAs can proceed either as oncogenic miRNAs (onco-miRs) or as tumor suppressor miRNAs (oncosuppressor-miRs), depending on the pathway in which they are involved. Dysregulation of miRNA expression has been shown in most of the tumors evaluated. MiRNA dysregulation is known to be involved in the development of cutaneous squamous cell carcinoma (CSCC). In this review, we focus on the recent evidence about the role of miRNAs in the development of CSCC and in the prognosis of this form of skin cancer.
Collapse
|
37
|
Hernández R, Sánchez-Jiménez E, Melguizo C, Prados J, Rama AR. Downregulated microRNAs in the colorectal cancer: diagnostic and therapeutic perspectives. BMB Rep 2019. [PMID: 30158023 PMCID: PMC6283029 DOI: 10.5483/bmbrep.2018.51.11.116] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Colorectal cancer (CRC), the third most common cancer in the world, has no specific biomarkers that facilitate its diagnosis and subsequent treatment. The miRNAs, small single-stranded RNAs that repress the mRNA translation and trigger the mRNA degradation, show aberrant levels in the CRC, by which these molecules have been related with the initiation, progression, and drug-resistance of this cancer type. Numerous studies show the microRNAs influence the cellular mechanisms related to the cell cycle, differentiation, apoptosis, and migration of the cancer cells through the post-transcriptionally regulated gene expression. Specific patterns of the upregulated and down-regulated miRNA have been associated with the CRC diagnosis, prognosis, and therapeutic response. Concretely, the downregulated miRNAs represent attractive candidates, not only for the CRC diagnosis, but for the targeted therapies via the tumor-suppressing microRNA replacement. This review shows a general overview of the potential uses of the miRNAs in the CRC diagnosis, prognosis, and treatment with a special focus on the downregulated ones. [BMB Reports 2018; 51(11): 563-571].
Collapse
Affiliation(s)
- Rosa Hernández
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada 18100; Biosanitary Institute of Granada (ibs. GRANADA), SAS-Universidad de Granada, Granada 18100; Department of Human Anatomy and Embryology, School of Medicine, University of Granada, Granada 18100, Spain
| | - Ester Sánchez-Jiménez
- Proteomics Laboratory CSIC/UAB, Institute of Biomedical Research, Barcelona 08036, Spain
| | - Consolación Melguizo
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada 18100; Biosanitary Institute of Granada (ibs. GRANADA), SAS-Universidad de Granada, Granada 18100; Department of Human Anatomy and Embryology, School of Medicine, University of Granada, Granada 18100, Spain
| | - Jose Prados
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada 18100; Biosanitary Institute of Granada (ibs. GRANADA), SAS-Universidad de Granada, Granada 18100; Department of Human Anatomy and Embryology, School of Medicine, University of Granada, Granada 18100, Spain
| | - Ana Rosa Rama
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada 18100; Department of Health Science, University of Jaén, Jaén 23071, Spain
| |
Collapse
|
38
|
Shang W, Xie Z, Lu F, Fang D, Tang T, Bi R, Chen L, Jiang L. Increased Thioredoxin-1 Expression Promotes Cancer Progression and Predicts Poor Prognosis in Patients with Gastric Cancer. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:9291683. [PMID: 30911354 PMCID: PMC6398115 DOI: 10.1155/2019/9291683] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 10/02/2018] [Accepted: 10/22/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND Thioredoxin-1 (Trx-1) is a small redox protein, which plays an important role in many biological processes. Although increased expression of Trx-1 in various solid tumors has been reported, the prognostic significance and function of Trx-1 in human gastric cancer (GC) are still unclear. Here, we investigated the clinical and prognostic significance of Trx-1 expression and the function and mechanism of Trx-1 in human GC. METHODS We analyzed Trx-1 mRNA expression from the GEO database and Trx-1 protein expression in 144 GC tissues using immunohistochemistry. Effects of Trx-1 on GC cell were assessed in vitro and in vivo through Trx-1 knockdown or overexpression. The antitumor effects of the Trx-1 inhibitor, PX-12, on GC cells were investigated. PTEN and p-AKT expressions were evaluated by Western blotting. RESULTS Increased Trx-1 expression was found in GC tissues and associated with poor prognosis and aggressive clinicopathological characteristics in patients with GC. High Trx-1 expression predicted poor prognosis, and its expression was an independent prognostic factor for overall survival of GC patients. Knockdown of Trx-1 expression inhibited GC cell growth, migration, and invasion in vitro and tumor growth and lung metastasis in vivo. Conversely, overexpression of Trx-1 promoted GC cell growth, migration, and invasion. We also found that PX-12 inhibited GC cell growth, migration, and invasion. Overexpression of Trx-1 caused a decrease in PTEN and increase in p-AKT levels whereas silencing Trx-1 caused an increase in PTEN and decrease in p-AKT levels in GC cells. Inhibition of AKT signaling pathway by MK2206 also inhibited GC cell growth, migration, and invasion. CONCLUSION Our results indicate that Trx-1 may be a promising prognostic indicator and therapeutic target for GC patients.
Collapse
Affiliation(s)
- Wenjing Shang
- Central Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Zhongdong Xie
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Fengying Lu
- Central Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Daoquan Fang
- Central Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Tianbin Tang
- Central Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Ruichun Bi
- Central Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Lingli Chen
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Lei Jiang
- Central Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| |
Collapse
|
39
|
Pang X, Zhou Z, Yu Z, Han L, Lin Z, Ao X, Liu C, He Y, Ponnusamy M, Li P, Wang J. Foxo3a-dependent miR-633 regulates chemotherapeutic sensitivity in gastric cancer by targeting Fas-associated death domain. RNA Biol 2019; 16:233-248. [PMID: 30628514 DOI: 10.1080/15476286.2019.1565665] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The development of chemotherapeutic drugs resistance such as doxorubicin (DOX) and cisplatin (DDP) is the major barrier in gastric cancer therapy. Emerging evidences reveal that microRNAs (miRNAs) contribute to chemosensitivity. In this study, we investigated the role of miR-633, an oncogenic miRNA, in gastric cancer chemoresistance. In gastric cancer tissue and cell lines, miR-633 expression was highly increased and correlated with down regulation of Fas-associated protein with death domain (FADD). Inhibition of miR-633 significantly increased FADD protein level and enhanced DOX/DDP induced apoptosis in vitro. MiR-633 antagomir administration remarkably decreased tumor growth in combination with DOX in vivo, suggesting that miR-633 targets FADD to block gastric cancer cell death. We found that the promoter region of miR-633 contained putative binding sites for forkhead box O 3 (Foxo3a), which can directly repress miR-633 transcription. In addition, we observed that DOX-induced nuclear accumulation of Foxo3a leaded to the suppression of miR-633 transcription. Together, our study revealed that miR-633/FADD axis played a significant role in the chemoresistance and Foxo3a regulated this pathway in gastric cancer. Thus, miR-633 antagomir resensitized gastric cancer cells to chemotherapy drug and had potentially therapeutic implication.
Collapse
Affiliation(s)
- Xin Pang
- a Center for Regenerative Medicine, Institute for Translational Medicine , Qingdao University , Qingdao , Shandong Province , China
| | - Zhixia Zhou
- a Center for Regenerative Medicine, Institute for Translational Medicine , Qingdao University , Qingdao , Shandong Province , China
| | - Zhuang Yu
- b Department of Oncology , Affiliated Hospital of Qingdao University , Qingdao , Shandong Province , China
| | - Lichun Han
- b Department of Oncology , Affiliated Hospital of Qingdao University , Qingdao , Shandong Province , China
| | - Zhijuan Lin
- a Center for Regenerative Medicine, Institute for Translational Medicine , Qingdao University , Qingdao , Shandong Province , China.,c Key Lab for Immunology in Universities of Shandong Province, School of Clinical Medicine , Weifang Medical University , Weifang , Shandong Province , China
| | - Xiang Ao
- a Center for Regenerative Medicine, Institute for Translational Medicine , Qingdao University , Qingdao , Shandong Province , China
| | - Chang Liu
- d Department of Oncology , PLA Army General Hospital , Beijin , China
| | - Yuqi He
- e Department of Gastroenterology , PLA Army General Hospital , Beijin , China
| | - Murugavel Ponnusamy
- a Center for Regenerative Medicine, Institute for Translational Medicine , Qingdao University , Qingdao , Shandong Province , China
| | - Peifeng Li
- a Center for Regenerative Medicine, Institute for Translational Medicine , Qingdao University , Qingdao , Shandong Province , China
| | - Jianxun Wang
- a Center for Regenerative Medicine, Institute for Translational Medicine , Qingdao University , Qingdao , Shandong Province , China
| |
Collapse
|
40
|
Identification of a nine-miRNA signature for the prognosis of Uveal Melanoma. Exp Eye Res 2019; 180:242-249. [PMID: 30615885 DOI: 10.1016/j.exer.2019.01.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Revised: 12/24/2018] [Accepted: 01/03/2019] [Indexed: 02/05/2023]
Abstract
The present study aims to construct a miRNA-based predictive signature of Uveal melanoma (UM) based on the database of the cancer genome atlas (TCGA). We obtained miRNA expression profiles and clinical information of 80 UM patients from TCGA, and randomly divided them into a training and a testing set. After data processing and forward screening, a total of 204 miRNAs with prognostic value were then examined by the Cox proportional hazard regression model in the training set. Receiver operating curve (ROC) analysis was applied to validate the accuracy of the signature. The biological relevance of putative miRNA target genes was also analyzed using the bioinformatics method. As a result, a linear prognostic model consisting of 9 miRNAs (miR-195, miR-224, miR-365a, miR-365b, miR-452, miR-4709, miR-7702, miR-513c, miR-873) was developed to divide UM patients into a high- and a low-risk group. Patients assigned to the high-risk group had significantly shorter overall survival than those in the low-risk group, which was further confirmed by the Area under curve (AUC) value of 0.858 at 5 year obtained from ROC. Gene Ontology (GO) analysis indicated that predicted target genes of these miRNAs are primarily associated with the modulation of protein expression and function, such as the activity of ubiquitin protein ligase and protein kinase. Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis revealed that these genes were involved in multiple signaling pathways linked to carcinogenesis. The tumor specific 9-miRNA signature was also verified in the testing and entire set. In summary, based on UM data of TCGA, we identified and validated a 9-miRNA-based prognostic signature.
Collapse
|
41
|
Tang Y, Yang G, Zhang J, Li X, Zhang C, Wang Y, Xu J, Chen Y, Teng Y, Yang X. E-cadherin is Required for the Homeostasis of Lgr5 + Gastric Antral Stem Cells. Int J Biol Sci 2019; 15:34-43. [PMID: 30662345 PMCID: PMC6329931 DOI: 10.7150/ijbs.28879] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 09/28/2018] [Indexed: 01/02/2023] Open
Abstract
Lgr5-expressing stem cells contribute to the epithelial turnover of the gastric antrum. However, the mechanism controlling the homeostasis of Lgr5+ antral stem cells is not fully understood. Here, we demonstrate the key role of E-cadherin in the homeostasis of Lgr5+ gastric antral stem cells. The deletion of E-cadherin in these cells results in their apoptosis, thereby leading to a marked decrease in their number. A reduced Lgr5+ stem cell pool caused by the loss of E-cadherin impairs gastric antral epithelial homeostasis in vivo and organoid growth in vitro. Furthermore, p53 contributes to the apoptosis of Lgr5+ stem cells following E-cadherin loss, while the simultaneous deletion of p53 rescues the phenotype in E-cadherin mutants. Our study reveals the critical pro-survival function of E-cadherin in Lgr5+ gastric antral stem cells and the key role of the Lgr5+ stem cell pool in the maintenance of gastric epithelial homeostasis.
Collapse
Affiliation(s)
- Yuling Tang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of Lifeomics, Beijing 102206, China
| | - Guan Yang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of Lifeomics, Beijing 102206, China
| | - Jinliang Zhang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of Lifeomics, Beijing 102206, China
| | - Xiubin Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of Lifeomics, Beijing 102206, China
| | - Chong Zhang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of Lifeomics, Beijing 102206, China
| | - Yanxiao Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of Lifeomics, Beijing 102206, China
| | - Jiaqian Xu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of Lifeomics, Beijing 102206, China
| | - Yeguang Chen
- State Key Laboratory of Membrane Biology, College of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yan Teng
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of Lifeomics, Beijing 102206, China
| | - Xiao Yang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of Lifeomics, Beijing 102206, China
| |
Collapse
|
42
|
Exosomes impact survival to radiation exposure in cell line models of nervous system cancer. Oncotarget 2018; 9:36083-36101. [PMID: 30546829 PMCID: PMC6281426 DOI: 10.18632/oncotarget.26300] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 10/21/2018] [Indexed: 12/24/2022] Open
Abstract
Radiation is utilized in the therapy of more than 50% of cancer patients. Unfortunately, many malignancies become resistant to radiation over time. We investigated the hypothesis that one method of a cancer cell's ability to survive radiation occurs through cellular communication via exosomes. Exosomes are cell-derived vesicles containing DNA, RNA, and protein. Three properties were analyzed: 1) exosome function, 2) exosome profile and 3) exosome uptake/blockade. To analyze exosome function, we show radiation-derived exosomes increased proliferation and enabled recipient cancer cells to survive radiation in vitro. Furthermore, radiation-derived exosomes increased tumor burden and decreased survival in an in vivo model. To address the mechanism underlying the alterations by exosomes in recipient cells, we obtained a profile of radiation-derived exosomes that showed expression changes favoring a resistant/proliferative profile. Radiation-derived exosomes contain elevated oncogenic miR-889, oncogenic mRNAs, and proteins of the proteasome pathway, Notch, Jak-STAT, and cell cycle pathways. Radiation-derived exosomes contain decreased levels of tumor-suppressive miR-516, miR-365, and multiple tumor-suppressive mRNAs. Ingenuity pathway analysis revealed the most represented networks included cell cycle, growth/survival. Upregulation of DNM2 correlated with increased exosome uptake. To analyze the property of exosome blockade, heparin and simvastatin were used to inhibit uptake of exosomes in recipient cells resulting in inhibited induction of proliferation and cellular survival. Because these agents have shown some success as cancer therapies, our data suggest their mechanism of action could be limiting exosome communication between cells. The results of our study identify a novel exosome-based mechanism that may underlie a cancer cell's ability to survive radiation.
Collapse
|
43
|
Wang C, Su K, Zhang Y, Zhang W, Chu D, Zhao Q, Guo R. MicroRNA-365 targets multiple oncogenes to inhibit proliferation, invasion, and self-renewal of aggressive endometrial cancer cells. Cancer Manag Res 2018; 10:5171-5185. [PMID: 30464615 PMCID: PMC6215916 DOI: 10.2147/cmar.s174889] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Background MicroRNA-365 (miR-365) has been reported to be a tumor suppressor miRNA. However, the role of miR-365 in progression of endometrial cancer (EC) has not been explored, in this study, we have found that re-expression of miRNA-365 inhibits cell proliferation, causes apoptosis and senescence. Materials and methods Overexpression of miR-365 attenuated cell migration and invasion, inhibited sphere-forming capacity, and enhanced the chemosensitivity to paclitaxel. In silico prediction tools identified the potential targets of miR-365. Results We identified EZH2 and FOS as targets of miR-365 and found that downregulating these genes imitated the tumor suppressive effect of miR-365. The outcomes of the study suggested that a reverse correlation existed between low miR-365 and overexpression of FOS and EZH2 in EC tissue specimens. Conclusion The study concludes that miR-365 acts as an important tumor suppressor and contributes by suppressing cell invasiveness, proliferation, and self-renewal in cancer cell lines by regulating multiple oncogenes. We establish that miR-365-EZH2/FOS pathway is an important target for treating EC.
Collapse
Affiliation(s)
- Chunfang Wang
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China,
| | - Ke Su
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China,
| | - Yanyan Zhang
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China,
| | - Weiwei Zhang
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China,
| | - Danxia Chu
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China,
| | - Qian Zhao
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China,
| | - Ruixia Guo
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China,
| |
Collapse
|
44
|
Jiang ZB, Ma BQ, Liu SG, Li J, Yang GM, Hou YB, Si RH, Gao P, Yan HT. miR-365 regulates liver cancer stem cells via RAC1 pathway. Mol Carcinog 2018; 58:55-65. [PMID: 30182377 PMCID: PMC6585981 DOI: 10.1002/mc.22906] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 08/03/2018] [Accepted: 08/31/2018] [Indexed: 12/11/2022]
Abstract
Liver cancer stem cells (CSCs) were involved in tumorigenesis, progression, recurrence, and drug resistance of hepatocellular carcinoma (HCC). miR-365 was downregulated in hepatocellular carcinoma and inhibited HCC cell proliferation and invasion. However, the role of miR-365 in liver cancer stem cells was unknown. Herein, we observed a remarkable decrease of miR-365 expression in CD133 or EpCAM-positive liver CSCs as well as in CSC-enriched hepatoma spheres. Up-regulated miR-365 suppressed liver CSC expansion by inhibiting the dedifferentiation of hepatoma cells and decreasing the self-renewal ability of liver CSCs. Mechanistically, bioinformatic and luciferase reporter analysis identified Ras-related C3 botulinum toxin substrate 1 (RAC1) as a direct target of miR-365. Overexpression of miR-365 in hepatoma cells downregulated the RAC1 mRNA and protein expression. RAC1 also could promote the expansion of liver CSCs. The special RAC1 inhibitor EHop-106 or RAC1 overexpression abolished the discrepancy in liver CSC proportion and the self-renewal capacity between miR-365 overexpression hepatoma cells and control cells, which further confirmed that RAC1 was required in miR-365-suppressed liver CSCs expansion. miR-365 was downregulated in liver CSCs and could inhibit HCC cells dedifferentiation and liver CSCs expansion by targeting RAC1 signaling.
Collapse
Affiliation(s)
- Ze-Bin Jiang
- Department of General Surgery, Gansu Provincial Hospital, Gansu, China
| | - Bing-Qiang Ma
- Department of General Surgery, Gansu Provincial Hospital, Gansu, China
| | - Shao-Guang Liu
- Department of Emergency Surgery, Gansu Provincial Hospital, Gansu, China
| | - Jing Li
- Department of General Surgery, Gansu Provincial Hospital, Gansu, China
| | - Guang-Ming Yang
- Department of General Surgery, Gansu Provincial Hospital, Gansu, China
| | - Ya-Bo Hou
- Department of General Surgery, Gansu Provincial Hospital, Gansu, China
| | - Ruo-Huang Si
- Department of General Surgery, Gansu Provincial Hospital, Gansu, China
| | - Peng Gao
- Department of General Surgery, Gansu Provincial Hospital, Gansu, China
| | - Hui-Ting Yan
- Department of Nursing Department, Gansu Provincial Hospital, Gansu, China
| |
Collapse
|
45
|
Zheng K, Wang N, Shen Y, Zhang Z, Gu Q, Xu X, Qin Q, Liu Y. Pro-apoptotic effects of micro-ribonucleic acid-365 on retinal neurons by targeting insulin-like growth factor-1 in diabetic rats: An in vivo and in vitro study. J Diabetes Investig 2018; 9:1041-1051. [PMID: 29427460 PMCID: PMC6123048 DOI: 10.1111/jdi.12815] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 01/17/2018] [Accepted: 02/04/2018] [Indexed: 12/22/2022] Open
Abstract
AIMS/OBJECTIVE The present study aimed to explore the effects of micro-ribonucleic acid-365 (miR-365) on apoptosis of retinal neurons by targeting insulin-like growth factor-1 (IGF-1) in diabetes mellitus rats. MATERIALS AND METHODS High glucose-induced retinal neurons were assigned into the blank (with no plasmid transfection), negative control (with plasmid transfection), anti-miR-365 (transfected miR-365 antagomir), transfected IGF-1 short hairpin RNA plasmid (sh-IGF-1) and transfected miR-365 antagomir and IGF-1 shRNA plasmid (anti-miR-365 + sh-IGF-1) groups. Proliferation and apoptosis of retinal neurons were detected by 5-ethynyl-2'-deoxyuridine assay and Hoechst 33342 staining, respectively. Expressions of miR-365, IGF-1, Bcl-2-associated X protein (Bax) and Bcl-2 were determined by reverse transcription quantitative polymerase chain reaction and western blotting. A control group contained 10 healthy rats. Terminal deoxynucleotidyl transferase dUTP nick-end labeling staining was used to evaluate apoptosis of retinal neurons in rats. RESULTS In the anti-miR-365 group, the apoptosis rate and Bax expression were reduced in comparison with the negative control and blank groups, whereas the sh-IGF-1 and anti-miR-365 + sh-IGF-1 groups presented an opposite trend. Compared with the normal group, expressions of miR-365 and Bax were increased, and expressions of IGF-1 and Bcl-2 were decreased, with more apoptotic cells in diabetes mellitus rat models. The sh-IGF-1 group had lower Bax expression, and higher expressions of IGF-1 and Bcl-2 with fewer apoptotic cells. Additionally, Bax expression was upregulated, expressions of IGF-1 and Bcl-2 were downregulated, and apoptotic cells were higher in the anti-miR-365 + sh-IGF-1 groups than the anti-miR-365 group. CONCLUSION The results of the present study suggest that suppressed miR-365 increases the IGF-1 expression, leading to anti-apoptotic effects on retinal neurons in diabetic rats.
Collapse
Affiliation(s)
- Kairong Zheng
- Department of OphthalmologyShanghai General HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Ning Wang
- Department of OphthalmologyShanghai General HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Yinchen Shen
- Department of OphthalmologyShanghai General HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Zhihua Zhang
- Department of OphthalmologyShanghai General HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Qing Gu
- Department of OphthalmologyShanghai General HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Xu Xu
- Department of OphthalmologyShanghai General HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Qing Qin
- Department of OphthalmologyShanghai General HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Yinchen Liu
- Department of OphthalmologyShanghai General HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| |
Collapse
|
46
|
Liu S, Tian Y, Zhu C, Yang X, Sun Q. High miR-718 Suppresses Phosphatase and Tensin Homolog (PTEN) Expression and Correlates to Unfavorable Prognosis in Gastric Cancer. Med Sci Monit 2018; 24:5840-5850. [PMID: 30131483 PMCID: PMC6116637 DOI: 10.12659/msm.909527] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Phosphatase and tensin homolog (PTEN) is a kind of phosphatase which has been demonstrated to suppress progression of gastric cancer. Many micro-RNAs (miRNAs), such as miR-106b, miR-93, and miR-200c, could inhibit expression of PTEN in cell lines; and many miRNAs including miR-21, miR-22, miR-18a, and miR-222 are related to the progression and prognosis of gastric cancer. However, among these miRNAs, the clinical significance of miR-718 has not yet been elucidated. MATERIAL AND METHODS The expression of PTEN and miR-718 in 141 gastric cancer tissues were detected by immunohistochemistry and quantitative real-time PCR respectively. The correlation between PTEN, miR-718, and the clinicopathological factors was analyzed by χ² test. The prognostic significance of PTEN and miR-718 was evaluated by univariate and multivariate analysis. Luciferase reporter assay was performed to evaluate the regulation of PTEN by miR-718. The effect of miR-718 on gastric cancer proliferation and invasion was investigated by MTT assay and Transwell assay. RESULTS Low expression of PTEN and high expression of miR-718 were both significantly associated with unfavorable prognosis, and both were identified as biomarkers predicting poorer prognosis of patients with gastric cancer. Increased miR-718 expression could decrease PTEN expression, thus enhancing phosphatidylinositide 3-kinases/protein kinase B (PI3K/Akt) signaling. Moreover, the abilities of proliferation and invasion of gastric cells transfected with miR-718 were promoted significantly compared with those transfected with control miRNA. CONCLUSIONS Low expression of PTEN and increased expression of miR-718 in gastric cancer tissues were both independent unfavorable prognostic factors of gastric cancer. Upregulation of miR-718 could increase PI3K/Akt signaling by directly downregulating PTEN, thus promoting the proliferation and invasion of gastric cancer cells.
Collapse
Affiliation(s)
- Shufang Liu
- Department of Clinical Laboratory, Linyi Central Hospital, Linyi, Shandong, China (mainland)
| | - Ying Tian
- Department of Clinical Laboratory, Linyi Central Hospital, Linyi, Shandong, China (mainland)
| | - Chanchan Zhu
- Department of Pathology, Medical School of Shandong University, Jinan, Shandong, China (mainland)
| | - Xiaoqing Yang
- Department of Pathology, Qianfoshan Hospital, Jinan, Shandong, China (mainland)
| | - Qing Sun
- Department of Pathology, Qianfoshan Hospital, Jinan, Shandong, China (mainland)
| |
Collapse
|
47
|
Zheng J, Dai X, Chen H, Fang C, Chen J, Sun L. Down-regulation of LHPP in cervical cancer influences cell proliferation, metastasis and apoptosis by modulating AKT. Biochem Biophys Res Commun 2018; 503:1108-1114. [PMID: 29944886 DOI: 10.1016/j.bbrc.2018.06.127] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Accepted: 06/22/2018] [Indexed: 12/22/2022]
Abstract
Cervical cancer is a leading severe malignancy throughout the world. Though various pathologies associated with cervical cancer progression have been demonstrated, further study is still necessary to reveal the tumorigenesis of cervical cancer. The protein histidine phosphatase LHPP is reported as a tumor suppressor. Histidine phosphorylation, also known as hidden phosphoproteome, is a poorly characterized post-translational modification of proteins. LHPP is evolutionarily conserved from worm to human. In the present study, we discovered that LHPP expression levels were lower in human cervical cancer tumors than that in adjacent normal tissue samples. LHPP expression levels were also reduced in several cervical cancer cell lines. Further, LHPP over-expression reduced the cell proliferation, migration and invasion, associated with the change of p53 and metastasis signaling pathways. Moreover, over-expressing LHPP markedly induced apoptosis in human cervical cancer cells via promoting the cleaved Caspse-3 and PARP. Importantly, we found that LHPP over-expression blocked AKT activation. Elevating AKT activity could abolish the role of LHPP over-expression in reducing cell proliferation and metastasis, as well as in inducing apoptotic response. Moreover, suppressing p53 expression with its inhibitor of PFTα abrogated the activity of LHPP to impede cell proliferation and metastasis, and to trigger apoptosis. AKT phosphorylation also restrained p53 expression levels in cervical cancer cells. In vivo, the anti-cervical cancer effects of LJPP were verified, which were also via the repression of cell proliferation and metastasis, and the induction of apoptosis. Therefore, LHPP could be considered as an effective candidate to develop effective therapeutic strategy against cervical cancer development.
Collapse
Affiliation(s)
- Jiangli Zheng
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
| | - Xue Dai
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
| | - Hailin Chen
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
| | - Chunli Fang
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
| | - Jian Chen
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
| | - Lizhou Sun
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China.
| |
Collapse
|
48
|
Binenbaum Y, Fridman E, Yaari Z, Milman N, Schroeder A, Ben David G, Shlomi T, Gil Z. Transfer of miRNA in Macrophage-Derived Exosomes Induces Drug Resistance in Pancreatic Adenocarcinoma. Cancer Res 2018; 78:5287-5299. [PMID: 30042153 DOI: 10.1158/0008-5472.can-18-0124] [Citation(s) in RCA: 263] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Revised: 05/17/2018] [Accepted: 07/13/2018] [Indexed: 01/01/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is known for its resistance to gemcitabine, which acts to inhibit cell growth by termination of DNA replication. Tumor-associated macrophages (TAM) were recently shown to contribute to gemcitabine resistance; however, the exact mechanism of this process is still unclear. Using a genetic mouse model of PDAC and electron microscopy analysis, we show that TAM communicate with the tumor microenvironment via secretion of approximately 90 nm vesicles, which are selectively internalized by cancer cells. Transfection of artificial dsDNA (barcode fragment) to murine peritoneal macrophages and injection to mice bearing PDAC tumors revealed a 4-log higher concentration of the barcode fragment in primary tumors and in liver metastasis than in normal tissue. These macrophage-derived exosomes (MDE) significantly decreased the sensitivity of PDAC cells to gemcitabine, in vitro and in vivo This effect was mediated by the transfer of miR-365 in MDE. miR-365 impaired activation of gemcitabine by upregulation of the triphospho-nucleotide pool in cancer cells and the induction of the enzyme cytidine deaminase; the latter inactivates gemcitabine. Adoptive transfer of miR-365 in TAM induced gemcitabine resistance in PDAC-bearing mice, whereas immune transfer of the miR-365 antagonist recovered the sensitivity to gemcitabine. Mice deficient of Rab27 a/b genes, which lack exosomal secretion, responded significantly better to gemcitabine than did wildtype. These results identify MDE as key regulators of gemcitabine resistance in PDAC and demonstrate that blocking miR-365 can potentiate gemcitabine response.Significance: Harnessing macrophage-derived exosomes as conveyers of antagomiRs augments the effect of chemotherapy against cancer, opening new therapeutic options against malignancies where resistance to nucleotide analogs remains an obstacle to overcome. Cancer Res; 78(18); 5287-99. ©2018 AACR.
Collapse
Affiliation(s)
- Yoav Binenbaum
- The Laboratory for Applied Cancer Research, Department of Otolaryngology Head and Neck Surgery, Clinical Research Institute at Rambam Healthcare Campus, Haifa, Israel
| | - Eran Fridman
- The Laboratory for Applied Cancer Research, Department of Otolaryngology Head and Neck Surgery, Clinical Research Institute at Rambam Healthcare Campus, Haifa, Israel
| | - Zvi Yaari
- Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Technion, Israel Institute of Technology, Haifa, Israel
| | - Neta Milman
- The Laboratory for Applied Cancer Research, Department of Otolaryngology Head and Neck Surgery, Clinical Research Institute at Rambam Healthcare Campus, Haifa, Israel
| | - Avi Schroeder
- Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Technion, Israel Institute of Technology, Haifa, Israel
| | - Gil Ben David
- The Laboratory for Applied Cancer Research, Department of Otolaryngology Head and Neck Surgery, Clinical Research Institute at Rambam Healthcare Campus, Haifa, Israel
| | - Tomer Shlomi
- Departments of Computer Science and Biology, Technion, Israel Institute of Technology, Haifa, Israel
| | - Ziv Gil
- The Laboratory for Applied Cancer Research, Department of Otolaryngology Head and Neck Surgery, Clinical Research Institute at Rambam Healthcare Campus, Haifa, Israel. .,Technion Integrated Cancer Center, Rappaport Institute of Medicine and Research, Technion, Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
49
|
Jamali L, Tofigh R, Tutunchi S, Panahi G, Borhani F, Akhavan S, Nourmohammadi P, Ghaderian SM, Rasouli M, Mirzaei H. Circulating microRNAs as diagnostic and therapeutic biomarkers in gastric and esophageal cancers. J Cell Physiol 2018; 233:8538-8550. [DOI: 10.1002/jcp.26850] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Accepted: 05/10/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Leila Jamali
- Department of Medical Genetics School of Medicine, Shahid Beheshti University of Medical Sciences Tehran Iran
| | | | - Sara Tutunchi
- Department of Medical Genetics Shahid Sadoughi University of Medical Sciences Yazd Iran
| | - Ghodratollah Panahi
- Department of Biochemistry Faculty of Medicine, Tehran University of Medical Sciences Tehran Iran
| | - Fatemeh Borhani
- Department of Basic Sciences Faculty of Medicine, Gonabad University of Medical Sciences Gonabad Iran
- Department of Basic Sciences Faculty of Medicine, Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Saeedeh Akhavan
- Department of Biology School of Basic Sciences, Science and Research Branch, Islamic Azad University Tehran Iran
| | - Parisa Nourmohammadi
- Department of Medical Genetics Shahid Sadoughi University of Medical Sciences Yazd Iran
| | - Sayyed M.H. Ghaderian
- Urogenital Stem Cell Research Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Milad Rasouli
- Department of Immunology Faculty of Medical Sciences, Tarbiat Modares University Tehran Iran
| | - Hamed Mirzaei
- Department of Medical Biotechnology School of Medicine Mashhad University of Medical Sciences Mashhad Iran
| |
Collapse
|
50
|
Lin Z, Zhou Z, Guo H, He Y, Pang X, Zhang X, Liu Y, Ao X, Li P, Wang J. Long noncoding RNA gastric cancer-related lncRNA1 mediates gastric malignancy through miRNA-885-3p and cyclin-dependent kinase 4. Cell Death Dis 2018; 9:607. [PMID: 29789536 PMCID: PMC5964145 DOI: 10.1038/s41419-018-0643-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 04/04/2018] [Accepted: 04/12/2018] [Indexed: 01/17/2023]
Abstract
Gastric cancer (GC) is one of the most common malignancy and the third leading cancer-related death in China. Long noncoding RNAs (lncRNAs) have been implicated in numerous tumors, including GC, however, the mechanism of many functional lncRNAs is still unclear. In this study, we identified the abundantly expressed lncRNA, RP11-290F20.3, in GC cells and patient tumor tissues. We named this lncRNA as GC-related lncRNA1 (GCRL1), which could regulate gastric cell proliferation and metastasis, both in vitro and in vivo. Mechanistically, miRNA-885-3p (miR-885-3p) could inhibit the cell proliferation and metastasis in GC by negatively regulating the expression of cyclin-dependent kinase 4 (CDK4) at the post-transcriptional level. Further, GCRL1 promoted the cell proliferation and metastasis by sponging miR-885-3p and hence, positively regulating CDK4 in GC cells. Taken together, our results demonstrate a novel regulatory axis of malignant cell proliferation and invasion in GC, comprising GCRL1, miR-885-3p, and CDK4, which may serve as a potential therapeutic target in GC.
Collapse
Affiliation(s)
- Zhijuan Lin
- Center for Tumor Molecular Biology, Institute for Translational Medicine, Qingdao University, Qingdao, 266021, China
- Key Lab for Immunology in Universities of Shandong Province, School of Clinical Medicine, Weifang Medical University, Weifang, 261053, China
| | - Zhixia Zhou
- Center for Tumor Molecular Biology, Institute for Translational Medicine, Qingdao University, Qingdao, 266021, China
| | - Hang Guo
- Department of Anesthesiology, PLA Army General Hospital, Beijing, 100700, China
| | - Yuqi He
- Department of Gastroenterology, PLA Army General Hospital, Beijing, 100700, China
| | - Xin Pang
- Center for Tumor Molecular Biology, Institute for Translational Medicine, Qingdao University, Qingdao, 266021, China
| | - Xumei Zhang
- Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang, 261041, China
| | - Ying Liu
- Center for Tumor Molecular Biology, Institute for Translational Medicine, Qingdao University, Qingdao, 266021, China
| | - Xiang Ao
- Center for Tumor Molecular Biology, Institute for Translational Medicine, Qingdao University, Qingdao, 266021, China
| | - Peifeng Li
- Center for Tumor Molecular Biology, Institute for Translational Medicine, Qingdao University, Qingdao, 266021, China.
| | - Jianxun Wang
- Center for Tumor Molecular Biology, Institute for Translational Medicine, Qingdao University, Qingdao, 266021, China.
| |
Collapse
|