1
|
Li ZB, Li YZ, Sun ZP, Li WX, Xiao Z, Wang F. ZEB2 knockdown inhibits interleukin-1β-induced cartilage degradation and inflammatory response through the Wnt/β-catenin pathway in human chondrocytes. Scand J Rheumatol 2024; 53:409-419. [PMID: 38899454 DOI: 10.1080/03009742.2024.2358594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 05/20/2024] [Indexed: 06/21/2024]
Abstract
OBJECTIVE Osteoarthritis (OA) is a degenerative disease of the joints characterized by inflammation and cartilage degeneration. Zinc finger E-box binding homeobox 2 (ZEB2) contains various function domains that interact with multiple transcription factors involved in various cellular functions. However, the function of ZEB2 in OA has not been clearly illustrated. METHOD Interleukin-1β (IL-1β) was used to establish an OA model in vitro. We quantified the ZEB2 expression in cartilage tissues from OA patients and IL-1β-induced chondrocytes through reverse transcription-quantitative polymerase chain reaction and Western blot. We then used functional assays to explore the function of ZEB2 during OA progression. RESULTS ZEB2 expression was increased in OA cartilage tissues and chondrocytes. The silencing of ZEB2 increased aggrecan and collagen II levels, and reduced the content of matrix metalloproteinase-3 (MMP-3), MMP-9, and MMP-13. ZEB2 knockdown inhibited the effects of IL-1β on the production of nitric oxide and prostaglandin E2, and the expression of inducible nitric oxide synthase and cyclooxygenase-2. ZEB2 inhibition also suppressed the levels of IL-6 and tumour necrosis factor-α, and increased the IL-10 level in IL-1β-treated cells. Mechanically, ZEB2 knockdown blocked the activation of the Wnt/β-catenin pathway in chondrocytes. CONCLUSION Knockdown of ZEB2 alleviated IL-1β-induced cartilage degradation and the inflammatory response through the Wnt/β-catenin pathway in chondrocytes.
Collapse
Affiliation(s)
- Z B Li
- Trauma Ward 2, The Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, PR China
| | - Y Z Li
- Department of Orthopedics, Shangluo Chinese Medicine Hospital, Shangluo, Shaanxi, PR China
| | - Z P Sun
- Trauma Ward 2, The Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, PR China
| | - W X Li
- Trauma Ward 2, The Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, PR China
| | - Z Xiao
- Trauma Ward 2, The Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, PR China
| | - F Wang
- Trauma Ward 2, The Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, PR China
| |
Collapse
|
2
|
Gao J, Lan T, Kostallari E, Guo Y, Lai E, Guillot A, Ding B, Tacke F, Tang C, Shah VH. Angiocrine signaling in sinusoidal homeostasis and liver diseases. J Hepatol 2024; 81:543-561. [PMID: 38763358 PMCID: PMC11906189 DOI: 10.1016/j.jhep.2024.05.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 04/29/2024] [Accepted: 05/10/2024] [Indexed: 05/21/2024]
Abstract
The hepatic sinusoids are composed of liver sinusoidal endothelial cells (LSECs), which are surrounded by hepatic stellate cells (HSCs) and contain liver-resident macrophages called Kupffer cells, and other patrolling immune cells. All these cells communicate with each other and with hepatocytes to maintain sinusoidal homeostasis and a spectrum of hepatic functions under healthy conditions. Sinusoidal homeostasis is disrupted by metabolites, toxins, viruses, and other pathological factors, leading to liver injury, chronic liver diseases, and cirrhosis. Alterations in hepatic sinusoids are linked to fibrosis progression and portal hypertension. LSECs are crucial regulators of cellular crosstalk within their microenvironment via angiocrine signaling. This review discusses the mechanisms by which angiocrine signaling orchestrates sinusoidal homeostasis, as well as the development of liver diseases. Here, we summarise the crosstalk between LSECs, HSCs, hepatocytes, cholangiocytes, and immune cells in health and disease and comment on potential novel therapeutic methods for treating liver diseases.
Collapse
Affiliation(s)
- Jinhang Gao
- Laboratory of Gastroenterology and Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China; Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Tian Lan
- Laboratory of Gastroenterology and Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China; Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China; Department of Hepatology and Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany
| | - Enis Kostallari
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Yangkun Guo
- Laboratory of Gastroenterology and Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China; Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Enjiang Lai
- Laboratory of Gastroenterology and Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China; Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Adrien Guillot
- Department of Hepatology and Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany
| | - Bisen Ding
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany.
| | - Chengwei Tang
- Laboratory of Gastroenterology and Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China; Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China.
| | - Vijay H Shah
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
3
|
Shen J, Wang Z, Liu M, Zhu YJ, Zheng L, Wang LL, Cheng JL, Liu TT, Zhang GD, Yang TY, Wang X, Zhang L. LincRNA-ROR/miR-145/ZEB2 regulates liver fibrosis by modulating HERC5-mediated p53 ISGylation. FASEB J 2023; 37:e22936. [PMID: 37144417 DOI: 10.1096/fj.202201182rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 04/04/2023] [Accepted: 04/12/2023] [Indexed: 05/06/2023]
Abstract
The tumor suppressor p53 has been implicated in the pathogenesis of liver fibrosis. HERC5-mediated posttranslational ISG modification of the p53 protein is critical for controlling its activity. Here, we demonstrated that the expression of HERC5 and ISG15 is highly elevated, whereas p53 is downregulated, in fibrotic liver tissues of mice and transforming growth factor-β1 (TGF-β1)-induced LX2 cells. HERC5 siRNA clearly increased the protein expression of p53, but the mRNA expression of p53 was not obviously changed. The inhibition of lincRNA-ROR (ROR) downregulated HERC5 expression and elevated p53 expression in TGF-β1-stimulated LX-2 cells. Furthermore, the expression of p53 was almost unchanged after TGF-β1-stimulated LX-2 cells were co-transfected with a ROR-expressing plasmid and HERC5 siRNA. We further confirmed that miR-145 is a target gene of ROR. In addition, we also showed that ROR regulates the HERC5-mediated ISGylation of p53 through mir-145/ZEB2. Together, we propose that ROR/miR-145/ZEB2 might be involved in the course of liver fibrosis by regulating ISGylation of the p53 protein.
Collapse
Affiliation(s)
- Jie Shen
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
- The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China
| | - Zhu Wang
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
- The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China
| | - Mei Liu
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
- The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China
| | - Yu-Jie Zhu
- Department of Radiology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ling Zheng
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
- The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China
| | - Li-Li Wang
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
- The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China
| | - Jie-Ling Cheng
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
- The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China
| | - Tong-Tong Liu
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
- The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China
| | - Guo-Dong Zhang
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
- The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China
| | - Tian-Yu Yang
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
- The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China
| | - Xiao Wang
- Department of Radiology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Lei Zhang
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
- The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China
| |
Collapse
|
4
|
Li X, Duan H, Wang S, Lv CX. Umbilical cord mesenchymal stem cell-derived exosomes reverse endometrial fibrosis by the miR-145-5p/ZEB2 axis in intrauterine adhesions. Reprod Biomed Online 2023; 46:234-243. [PMID: 36567149 DOI: 10.1016/j.rbmo.2022.05.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/21/2022] [Accepted: 05/24/2022] [Indexed: 02/07/2023]
Abstract
RESEARCH QUESTION What is the specific mechanism of umbilical cord mesenchymal stem cell-derived exosomes (UCMSC-exos) in regulating endometrial repair and regeneration? DESIGN In this study, UCMSC-exos were harvested by differential ultracentrifugation from umbilical cord mesenchymal stem cell culture supernatant and identified with western blotting, transmission electron microscopy and nanoparticle tracking analysis. Transforming growth factor-β1 (TGFβ1) at different concentrations was used to construct the intrauterine adhesions cell model. The fibrotic markers were assessed by quantitative reverse transcription-polymerase chain reaction and western blotting. The effects of miR-145-5p over-expression on endometrial fibrosis were assessed. Dual luciferase assay was performed to verify the relationship between miR-145-5p and zinc finger E-box binding homeobox 2 (ZEB2). RESULTS The isolated UCMSC-exos had a typical cup-shaped morphology, expressed the specific exosomal markers Alix, CD63 and TSG101, and were approximately 50-150 nm in diameter. TGFβ1 at 10 ng/ml significantly promoted endometrial fibrosis, which was reversed by 20 µg/ml UCMSC-exos. Exosomal miR-145-5p ameliorated TGFβ1-induced endometrial fibrosis. ZEB2 was inversely regulated by exosomal miR-145-5p as a direct target. CONCLUSIONS UCMSC-exos might reverse endometrial stromal cell fibrosis by regulating the miR-145-5p/ZEB2 axis, representing a potential novel strategy to promote endometrial repair.
Collapse
Affiliation(s)
- Xiao Li
- Department of Minimally Invasive Gynecologic Center, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100006, China
| | - Hua Duan
- Department of Minimally Invasive Gynecologic Center, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100006, China.
| | - Sha Wang
- Department of Minimally Invasive Gynecologic Center, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100006, China
| | - Cheng-Xiao Lv
- Department of Minimally Invasive Gynecologic Center, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100006, China
| |
Collapse
|
5
|
Qiu JL, Zhang GF, Chai YN, Han XY, Zheng HT, Li XF, Duan F, Chen LY. Ligustrazine Attenuates Liver Fibrosis by Targeting miR-145 Mediated Transforming Growth Factor- β/Smad Signaling in an Animal Model of Biliary Atresia. J Pharmacol Exp Ther 2022; 381:257-265. [PMID: 35398813 DOI: 10.1124/jpet.121.001020] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 03/28/2022] [Indexed: 01/03/2025] Open
Abstract
To investigate therapeutic target for ligustrazine during liver fibrosis in an ethanol-induced biliary atresia rat model and transforming growth factor-β (TGF-β) induced hepatic stellate cell activation cell model, and the underlying mechanism, a total of 30 rats were randomly assigned into five groups (n = 6 per group): control, sham, ethanol-induced biliary atresia model, model plus pirfenidone, and model plus ligustrazine groups. The liver changes were assessed using H&E and Masson staining and transmission electron microscopy. Expression of miR-145 and mRNA and protein levels of TGF-β/smads pathway-related proteins were detected. HSC-T6 cells were infected with LV-miR or rLV-miR-145 in the presence or absence of SMAD3 inhibitor SIS3 and treated with 2.5 ng/ml TGF-β1 and then with ligustrazine. Collected cells were subjected to detect the expression of miR-145 and mRNA and protein expression levels of TGF-β/smads pathway-related proteins. Ligustrazine rescued liver fibrogenesis and pathology for ethanol-caused bile duct injury, revealed by decreased α-smooth muscle actin and collagen I expression and liver tissue and cell morphology integrity. Further experiments showed that ligustrazine inhibited intrinsic and phosphorylated Smad2/3 protein expression and modification. Similar results were obtained in cells. In addition, ligustrazine altered miR-145 expression in both animal and cell models. Lentivirus mediated miR-145 overexpression and knockdown recombinant virus showed that miR-145 enhanced the TGF-β/Smad pathway, which led to hepatic stellate cell activation, and ligustrazine blocked this activation. This work validated that ligustrazine-regulated miR-145 mediated TGF-β/Smad signaling to inhibit the progression of liver fibrosis in a biliary atresia rat model and provided a new therapeutic strategy for liver fibrosis. SIGNIFICANCE STATEMENT: With an ethanol-induced biliary atresia rat model, ligustrazine was found to rescue liver fibrogenesis and pathology for ethanol caused bile duct injury, revealed by decreased α-smooth muscle actin and collagen I expression and liver tissue and cell morphology integrity. Furthermore, we found ligustrazine upregulated miR-145 expression and inhibited TGF-β/SMAD signaling pathway both in vivo and in vitro. In addition, overexpression and knockdown of miR-145 confirmed that miR-145 is involved in the ligustrazine inhibition of liver fibrosis through the TGF-β/SMAD signaling pathway.
Collapse
Affiliation(s)
- Jian-Li Qiu
- Department of Pediatrics (J.-L.Q., H.-T.Z., X.-F.L.) and Department of Gastroenterology (F.D.), the First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China; Department of Pediatric Surgery (G.-F.Z.) and Department of Pharmacy (Y.-N.C.), the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Henan University of Chinese Medicine, Zhengzhou, China (X.-Y.H.); and Department of Rehabilitation, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China (L.-Y.C.)
| | - Guo-Feng Zhang
- Department of Pediatrics (J.-L.Q., H.-T.Z., X.-F.L.) and Department of Gastroenterology (F.D.), the First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China; Department of Pediatric Surgery (G.-F.Z.) and Department of Pharmacy (Y.-N.C.), the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Henan University of Chinese Medicine, Zhengzhou, China (X.-Y.H.); and Department of Rehabilitation, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China (L.-Y.C.)
| | - Yu-Na Chai
- Department of Pediatrics (J.-L.Q., H.-T.Z., X.-F.L.) and Department of Gastroenterology (F.D.), the First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China; Department of Pediatric Surgery (G.-F.Z.) and Department of Pharmacy (Y.-N.C.), the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Henan University of Chinese Medicine, Zhengzhou, China (X.-Y.H.); and Department of Rehabilitation, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China (L.-Y.C.)
| | - Xiao-Yan Han
- Department of Pediatrics (J.-L.Q., H.-T.Z., X.-F.L.) and Department of Gastroenterology (F.D.), the First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China; Department of Pediatric Surgery (G.-F.Z.) and Department of Pharmacy (Y.-N.C.), the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Henan University of Chinese Medicine, Zhengzhou, China (X.-Y.H.); and Department of Rehabilitation, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China (L.-Y.C.)
| | - Hai-Tao Zheng
- Department of Pediatrics (J.-L.Q., H.-T.Z., X.-F.L.) and Department of Gastroenterology (F.D.), the First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China; Department of Pediatric Surgery (G.-F.Z.) and Department of Pharmacy (Y.-N.C.), the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Henan University of Chinese Medicine, Zhengzhou, China (X.-Y.H.); and Department of Rehabilitation, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China (L.-Y.C.)
| | - Xiang-Feng Li
- Department of Pediatrics (J.-L.Q., H.-T.Z., X.-F.L.) and Department of Gastroenterology (F.D.), the First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China; Department of Pediatric Surgery (G.-F.Z.) and Department of Pharmacy (Y.-N.C.), the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Henan University of Chinese Medicine, Zhengzhou, China (X.-Y.H.); and Department of Rehabilitation, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China (L.-Y.C.)
| | - Fei Duan
- Department of Pediatrics (J.-L.Q., H.-T.Z., X.-F.L.) and Department of Gastroenterology (F.D.), the First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China; Department of Pediatric Surgery (G.-F.Z.) and Department of Pharmacy (Y.-N.C.), the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Henan University of Chinese Medicine, Zhengzhou, China (X.-Y.H.); and Department of Rehabilitation, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China (L.-Y.C.)
| | - Ling-Yan Chen
- Department of Pediatrics (J.-L.Q., H.-T.Z., X.-F.L.) and Department of Gastroenterology (F.D.), the First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China; Department of Pediatric Surgery (G.-F.Z.) and Department of Pharmacy (Y.-N.C.), the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Henan University of Chinese Medicine, Zhengzhou, China (X.-Y.H.); and Department of Rehabilitation, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China (L.-Y.C.)
| |
Collapse
|
6
|
Liu Z, Zhou S, Zhang Y, Zhao M. Rat bone marrow mesenchymal stem cells (BMSCs) inhibit liver fibrosis by activating GSK3β and inhibiting the Wnt3a/β-catenin pathway. Infect Agent Cancer 2022; 17:17. [PMID: 35440002 PMCID: PMC9017036 DOI: 10.1186/s13027-022-00432-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 04/11/2022] [Indexed: 11/20/2022] Open
Abstract
Background Bone marrow mesenchymal stem cells (BMSCs) can effectively alleviate liver fibrosis, which is a pathological injury caused by various chronic liver diseases. This study aimed to investigate the antifibrotic effects of BMSCs and elucidate the underlying mechanism by which BMSCs affect liver fibrosis in vitro and in vivo. Methods After the rat liver fibrosis model was induced by continuous injection of carbon tetrachloride (CCl4), BMSCs were administered for 4 weeks, and histopathological analysis and liver function tests were performed. T6 hepatic stellate cells (HSC-T6 cells) were stimulated by TGF-β1, and the activation and proliferation of cells were analyzed by CCK-8 assays, flow cytometry, real-time PCR, western blotting and enzyme-linked immunosorbent assay (ELISA). Results Our data demonstrated that BMSCs effectively reduced the accumulation of collagen, enhanced liver functionality and ameliorated liver fibrosis in vivo. BMSCs increased the sub-G1 population in HSC-T6 cells. In addition, coculture with BMSCs reduced the expression of α-SMA, collagen I, cyclin-D1, and c-Myc in HSC-T6 cells and activated the phosphorylation of GSK3β. The GSK3β inhibitor SB216763 reversed the effect of BMSCs. The Wnt/β-catenin signalling pathway was involved in BMSC-mediated inhibition of HSC-T6 cell activation. Conclusions Our data suggested that BMSCs exerted antifibrotic effects by activating the expression of GSK3β and inhibiting the Wnt3a/β-catenin signalling pathway.
Collapse
Affiliation(s)
- Zhaoguo Liu
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, China.,Liuzhou Worker's Hospital, Liuzhou, Guangxi Province, China
| | - Song Zhou
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Ya Zhang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Ming Zhao
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, China. .,Department of Organ Transplantation, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510280, Guangdong Province, China.
| |
Collapse
|
7
|
Chen W, Jiang W, Dong J, Wang J, Wang B. MiR-200b-3p induces the formation of insulin-producing cells from umbilical cord mesenchymal stem cells by targeting ZEB2. Crit Rev Eukaryot Gene Expr 2022; 32:33-46. [DOI: 10.1615/critreveukaryotgeneexpr.2022041822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
8
|
Montaldo C, Terri M, Riccioni V, Battistelli C, Bordoni V, D'Offizi G, Prado MG, Trionfetti F, Vescovo T, Tartaglia E, Strippoli R, Agrati C, Tripodi M. Fibrogenic signals persist in DAA-treated HCV patients after sustained virological response. J Hepatol 2021; 75:1301-1311. [PMID: 34271004 DOI: 10.1016/j.jhep.2021.07.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 06/22/2021] [Accepted: 07/01/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS Patients with HCV who achieve a sustained virological response (SVR) on direct-acting antiviral (DAA) therapy still need to be monitored for signs of liver disease progression. To this end, the identification of both disease biomarkers and therapeutic targets is necessary. METHODS Extracellular vesicles (EVs) purified from plasma of 15 healthy donors (HDs), and 16 HCV-infected patients before (T0) and after (T6) DAA treatment were utilized for functional and miRNA cargo analysis. EVs purified from plasma of 17 HDs and 23 HCV-infected patients (T0 and T6) were employed for proteomic and western blot analyses. Functional analysis in LX2 cells measured fibrotic markers (mRNAs and proteins) in response to EVs. Structural analysis was performed by qPCR, label-free liquid chromatography-mass spectrometry and western blot. RESULTS On the basis of observations indicating functional differences (i.e. modulation of FN-1, ACTA2, Smad2/3 phosphorylation, collagen deposition) of plasma-derived EVs from HDs, T0 and T6, we performed structural analysis of EVs. We found consistent differences in terms of both miRNA and protein cargos: (i) antifibrogenic miR204-5p, miR181a-5p, miR143-3p, miR93-5p and miR122-5p were statistically underrepresented in T0 EVs compared to HD EVs, while miR204-5p and miR143-3p were statistically underrepresented in T6 EVs compared to HD EVs (p <0.05); (ii) proteomic analysis highlighted, in both T0 and T6, the modulation of several proteins with respect to HDs; among them, the fibrogenic protein DIAPH1 was upregulated (Log2 fold change of 4.4). CONCLUSIONS Taken together, these results highlight structural EV modifications that are conceivably causal for long-term liver disease progression in patients with HCV despite DAA-mediated SVR. LAY SUMMARY Direct-acting antivirals lead to virological cure in the majority of patients with chronic hepatitis C virus infection. However, the risk of liver disease progression or complications in patients with fibrosis and cirrhosis remains in some patients even after virological cure. Herein, we show that extracellular vesicle modifications could be linked to long-term liver disease progression in patients who have achieved virological cure; these modifications could potentially be used as biomarkers or treatment targets in such patients.
Collapse
Affiliation(s)
- Claudia Montaldo
- National Institute for Infectious Diseases L. Spallanzani, IRCCS, Italy
| | - Michela Terri
- National Institute for Infectious Diseases L. Spallanzani, IRCCS, Italy; Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Veronica Riccioni
- Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Cecilia Battistelli
- Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Veronica Bordoni
- National Institute for Infectious Diseases L. Spallanzani, IRCCS, Italy
| | | | - Maria Giulia Prado
- Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Flavia Trionfetti
- National Institute for Infectious Diseases L. Spallanzani, IRCCS, Italy; Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Tiziana Vescovo
- National Institute for Infectious Diseases L. Spallanzani, IRCCS, Italy
| | | | - Raffaele Strippoli
- National Institute for Infectious Diseases L. Spallanzani, IRCCS, Italy; Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Chiara Agrati
- National Institute for Infectious Diseases L. Spallanzani, IRCCS, Italy
| | - Marco Tripodi
- National Institute for Infectious Diseases L. Spallanzani, IRCCS, Italy; Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
9
|
Abstract
Hepatic fibrosis is a reversible wound healing process following liver injury. Although this process is necessary for maintaining liver integrity, severe excessive extracellular matrix accumulation (ECM) could lead to permanent scar formation and destroy the liver structure. The activation of hepatic stellate cells (HSCs) is a key event in hepatic fibrosis. Previous studies show that most antifibrotic therapies focus on the apoptosis of HSCs and the prevention of HSC activation. Noncoding RNAs (ncRNAs) play a substantial role in HSC activation and are likely to be biomarkers or therapeutic targets for the treatment of hepatic fibrosis. This review summarizes and discusses the previously reported ncRNAs, including the microRNAs, long noncoding RNAs, and circular RNAs, highlighting their regulatory roles and interactions in the signaling pathways that regulate HSC activation in hepatic fibrosis.
Collapse
|
10
|
Li M, Du M, Wang Y, Zhu J, Pan J, Cao Z, He H. CircRNA Lrp6 promotes cementoblast differentiation via miR-145a-5p/Zeb2 axis. J Periodontal Res 2021; 56:1200-1212. [PMID: 34492118 DOI: 10.1111/jre.12933] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 08/26/2021] [Indexed: 01/19/2023]
Abstract
BACKGROUND AND OBJECTIVE Cementum is a part of the periodontium and anchors periodontal ligaments to the alveolar bone. Cementoblasts are responsible for the cementum formation via matrix deposition and subsequently mineralization. Thus, exploring novel mechanisms underlying the function of cementoblast contributes to the treatment of cementum damage. Recently, circRNA Lrp6 (circLRP6) has been of interest due to its active role in cell differentiation, but its potential role in cementoblast differentiation remains unclear. Herein, we attempted to elucidate the role of circLRP6 in cementoblast differentiation and clarify any associated mechanisms. MATERIAL AND METHODS The mRNA expressions of circLRP6, miR-145a-5p, zinc finger E-box binding homeobox 2 (Zeb2), runt-related transcription factor 2 (Runx2), osteopontin (Opn), and bone sialoprotein (Bsp) were evaluated by qRT-PCR. The protein levels of Zeb2 were measured by Western blot. Bioinformatic analysis and dual-luciferase reporter assays were used to test the potential binding targets of miR-145a-5p. The differentiation potentials of the cementoblasts were assessed by Alkaline phosphatase (ALP) staining, ALP activity assay, Alizarin red S (ARS) staining, and quantification. RESULTS In this study, circLRP6 was significantly upregulated in cementoblast differentiation. Furthermore, circLRP6 knockdown inhibited ALP levels, reduced calcium nodule formation and the expression of Runx2, Opn, and Bsp. Mechanically, bioinformatic analysis and dual-luciferase reporter assays confirmed miR-145a-5p was a potential binding target of circLRP6. miR-145a-5p can negatively regulate cementoblast differentiation. Subsequently, bioinformatic analysis and dual-luciferase reporter assays confirmed Zeb2 was a potential miR-145a-5p target. miR-145a-5p overexpression resulted in a downregulation of Zeb2. Furthermore, Zeb2 inhibition partially reversed the effect of circLRP6 during cementoblast differentiation. CONCLUSION Taken together, circLRP6 appears to modulate cementoblast differentiation by antagonizing the function of miR-145a-5p, thereby increasing Zeb2. This study serves as a stepping stone for the potential development of an approach to promote cementum formation.
Collapse
Affiliation(s)
- Mengying Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Mingyuan Du
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yunlong Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Jiaqi Zhu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Jiawen Pan
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zhengguo Cao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Hong He
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China.,Department of Orthodontics, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
11
|
Yang J, Wu M, Fang H, Su Y, Zhang L, Zhou H. Puerarin Prevents Acute Liver Injury via Inhibiting Inflammatory Responses and ZEB2 Expression. Front Pharmacol 2021; 12:727916. [PMID: 34421621 PMCID: PMC8378253 DOI: 10.3389/fphar.2021.727916] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 07/26/2021] [Indexed: 01/02/2023] Open
Abstract
Puerarin, an isoflavone component extracted from herb radix puerariae, is widely used in China in the treatment of immune diseases and inflammation. Previous studies have demonstrated that puerarin prevented acute lung injury by regulating inflammatory responses. However, the effect of puerarin on acute liver injury (ALI) was unclear. The purpose of this study was to explore the beneficial effects of puerarin when applied to ALI. We found that puerarin inhibited liver injury and inflammatory cell infiltration in lipopolysaccharide (LPS)/D-galactose (D-Gal)-induced acute liver failure and the liver pro-inflammatory cytokines interleukin (IL)-1β, IL-6, and tumor necrosis factor-alpha (TNF-α) in liver tissues with ALI and LPS-induced L-02 cells but upregulated the expression level of zinc finger E-box-binding homeobox 2 (ZEB2). Significantly, the results of this study showed that the inhibition of liver pro-inflammatory cytokine (IL-1β, IL-6, and TNF-α) production in LPS-induced L-02 cells was caused by ZEB2 overexpression. However, knocking down ZEB2 promoted LPS-mediated secretion of liver pro-inflammatory cytokines in L-02 cells. Additional experiments showed that puerarin inhibited the activation of the NF-κB signaling pathway by elevating ZEB2 expression in L-02 cells. In summary, puerarin most likely prevented activation of the pro-inflammatory factors and reduced LPS/D-Gal-induced liver injury by enhancing the ZEB2 expression level and, consequently, blocking activation of the NF-κB signaling pathway in the liver.
Collapse
Affiliation(s)
- Junfa Yang
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China.,School of Pharmacy, Anhui Medical University, Hefei, China
| | - Maomao Wu
- Department of Pharmacy, Anhui Chest Hospital, Hefei, China
| | - Hui Fang
- Hangzhou Normal University Affiliated Hospital, Hangzhou, China
| | - Yue Su
- Institute of Clinical Trial, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Lingling Zhang
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Huan Zhou
- School of Public Basic, Bengbu Medical College, Bengbu, China
| |
Collapse
|
12
|
Griffin MF, Huber J, Evan FJ, Quarto N, Longaker MT. The role of Wnt signaling in skin fibrosis. Med Res Rev 2021; 42:615-628. [PMID: 34431110 DOI: 10.1002/med.21853] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 05/14/2021] [Accepted: 08/17/2021] [Indexed: 12/20/2022]
Abstract
Skin fibrosis is the excessive deposition of extracellular matrix in the dermis. Cutaneous fibrosis can occur following tissue injury, including burns, trauma, and surgery, resulting in scars that are disfiguring, limit movement and cause significant psychological distress for patients. Many molecular pathways have been implicated in the development of skin fibrosis, yet effective treatments to prevent or reverse scarring are unknown. The Wnt signaling pathways are known to play an important role in skin homeostasis, skin injury, and in the development of fibrotic skin diseases. This review provides a detailed overview of the role of the canonical Wnt signaling pathways in regulating skin scarring. We also discuss how Wnt signaling interacts with other known fibrotic molecular pathways to cause skin fibrosis. We further provide a summary of the different Wnt inhibitor types available for treating skin scarring. Understanding the role of the Wnt pathway in cutaneous fibrosis will accelerate the development of effective Wnt modulators for the treatment of skin fibrosis.
Collapse
Affiliation(s)
- Michelle F Griffin
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford, California, USA
| | - Julika Huber
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford, California, USA
| | - Fahy J Evan
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford, California, USA
| | - Natalina Quarto
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford, California, USA.,Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Michael T Longaker
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford, California, USA.,Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
13
|
Zhao H, Feng YL, Liu T, Wang JJ, Yu J. MicroRNAs in organ fibrosis: From molecular mechanisms to potential therapeutic targets. Pathol Res Pract 2021; 225:153588. [PMID: 34419718 DOI: 10.1016/j.prp.2021.153588] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 08/09/2021] [Accepted: 08/11/2021] [Indexed: 12/20/2022]
Abstract
Fibrosis is caused by chronic tissue injury and characterized by the excessive deposition of extracellular matrix (ECM) that ultimately results in organ failure and death. Owing to lacking of effective treatment against tissue fibrosis, it causes a high morbidity and mortality worldwide. Thus, it is of great importance to find an effective therapy strategy for the treatment of fibrosis. MicroRNAs (miRNAs) play vital roles in many biological processes by targeting downstream genes. Numerous studies demonstrated that miRNAs served as biomarkers of various diseases, suggesting the potential therapeutic targets for diseases. It was recently reported that miRNAs played an important role in the development of organ fibrosis, which showed a promising prospect against fibrosis by targeting intervention. Here, we summarize the roles of miRNAs in the process of organ fibrosis, including liver, lung, heart and kidney, and highlight miRNAs being novel therapeutic targets for organ fibrosis.
Collapse
Affiliation(s)
- Hui Zhao
- Clinical Experimental Center, Xi'an International Medical Center Hospital, No. 777 Xitai Road Xi'an, Shaanxi 710100, China; Xi'an Engineering Technology Research Center for Cardiovascular Active Peptids, No. 777 Xitai Road Xi'an, Shaanxi 710100, China
| | - Ya-Long Feng
- School of Chemistry and Chemical Engineering, Xianyang Normal University, Xianyang, Shaanxi, 712000, China
| | - Tian Liu
- Clinical Experimental Center, Xi'an International Medical Center Hospital, No. 777 Xitai Road Xi'an, Shaanxi 710100, China; Xi'an Engineering Technology Research Center for Cardiovascular Active Peptids, No. 777 Xitai Road Xi'an, Shaanxi 710100, China
| | - Jing-Jing Wang
- Weinan Linwei District Maternal and Child Health Family Planning Service Center, No.144 Dongfeng Road Weinan, Shannxi 714000, China
| | - Jun Yu
- Clinical Experimental Center, Xi'an International Medical Center Hospital, No. 777 Xitai Road Xi'an, Shaanxi 710100, China; Xi'an Engineering Technology Research Center for Cardiovascular Active Peptids, No. 777 Xitai Road Xi'an, Shaanxi 710100, China.
| |
Collapse
|
14
|
de Haan W, Dheedene W, Apelt K, Décombas-Deschamps S, Vinckier S, Verhulst S, Conidi A, Deffieux T, Staring MW, Vandervoort P, Caluwé E, Lox M, Mannaerts I, Takagi T, Jaekers J, Berx G, Haigh J, Topal B, Zwijsen A, Higashi Y, van Grunsven LA, van IJcken WFJ, Mulugeta E, Tanter M, Lebrin FPG, Huylebroeck D, Luttun A. Endothelial Zeb2 preserves the hepatic angioarchitecture and protects against liver fibrosis. Cardiovasc Res 2021; 118:1262-1275. [PMID: 33909875 PMCID: PMC8953454 DOI: 10.1093/cvr/cvab148] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 04/26/2021] [Indexed: 02/06/2023] Open
Abstract
Aims Hepatic capillaries are lined with specialized liver sinusoidal endothelial cells (LSECs) which support macromolecule passage to hepatocytes and prevent fibrosis by keeping hepatic stellate cells (HSCs) quiescent. LSEC specialization is co-determined by transcription factors. The zinc-finger E-box-binding homeobox (Zeb)2 transcription factor is enriched in LSECs. Here, we aimed to elucidate the endothelium-specific role of Zeb2 during maintenance of the liver and in liver fibrosis. Methods and results To study the role of Zeb2 in liver endothelium we generated EC-specific Zeb2 knock-out (ECKO) mice. Sequencing of liver EC RNA revealed that deficiency of Zeb2 results in prominent expression changes in angiogenesis-related genes. Accordingly, the vascular area was expanded and the presence of pillars inside ECKO liver vessels indicated that this was likely due to increased intussusceptive angiogenesis. LSEC marker expression was not profoundly affected and fenestrations were preserved upon Zeb2 deficiency. However, an increase in continuous EC markers suggested that Zeb2-deficient LSECs are more prone to dedifferentiation, a process called ‘capillarization’. Changes in the endothelial expression of ligands that may be involved in HSC quiescence together with significant changes in the expression profile of HSCs showed that Zeb2 regulates LSEC–HSC communication and HSC activation. Accordingly, upon exposure to the hepatotoxin carbon tetrachloride (CCl4), livers of ECKO mice showed increased capillarization, HSC activation, and fibrosis compared to livers from wild-type littermates. The vascular maintenance and anti-fibrotic role of endothelial Zeb2 was confirmed in mice with EC-specific overexpression of Zeb2, as the latter resulted in reduced vascularity and attenuated CCl4-induced liver fibrosis. Conclusion Endothelial Zeb2 preserves liver angioarchitecture and protects against liver fibrosis. Zeb2 and Zeb2-dependent genes in liver ECs may be exploited to design novel therapeutic strategies to attenuate hepatic fibrosis.
Collapse
Affiliation(s)
- Willeke de Haan
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Wouter Dheedene
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Katerina Apelt
- Department of Internal Medicine (Nephrology), Einthoven Laboratory for Experimental Vascular Medicine. Leiden University Medical Center, . Leiden, The Netherlands
| | - Sofiane Décombas-Deschamps
- Physics for Medicine Paris, Inserm, CNRS, ESPCI Paris, Paris Sciences et Lettres University, Paris, France
| | - Stefan Vinckier
- Department of Oncology, Laboratory of Angiogenesis and Vascular Metabolism, KU Leuven, Leuven, Belgium.,Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, Vlaams Instituut voor Biotechnologie (VIB), Leuven, Belgium
| | - Stefaan Verhulst
- Liver Cell Biology research group, Vrije Universiteit Brussel, Brussels, Belgium
| | - Andrea Conidi
- Department of Cell Biology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Thomas Deffieux
- Physics for Medicine Paris, Inserm, CNRS, ESPCI Paris, Paris Sciences et Lettres University, Paris, France
| | - Michael W Staring
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Petra Vandervoort
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Ellen Caluwé
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Marleen Lox
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Inge Mannaerts
- Liver Cell Biology research group, Vrije Universiteit Brussel, Brussels, Belgium
| | - Tsuyoshi Takagi
- Department of Disease Model, Institute of Developmental Research, Aichi Developmental Disability Center, Aichi, Japan
| | | | - Geert Berx
- Molecular and Cellular Oncology Laboratory, Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Jody Haigh
- Department of Pharmacology and Therapeutics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada.,Research Institute in Oncology and Hematology, Cancer Care Manitoba, Winnipeg, Manitoba, Canada
| | - Baki Topal
- Abdominal Surgery, UZ Leuven, Leuven, Belgium
| | - An Zwijsen
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Yujiro Higashi
- Department of Disease Model, Institute of Developmental Research, Aichi Developmental Disability Center, Aichi, Japan
| | - Leo A van Grunsven
- Liver Cell Biology research group, Vrije Universiteit Brussel, Brussels, Belgium
| | - Wilfred F J van IJcken
- Department of Cell Biology, Erasmus University Medical Center, Rotterdam, The Netherlands.,Center for Biomics-Genomics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Eskeatnaf Mulugeta
- Department of Cell Biology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Mickael Tanter
- Physics for Medicine Paris, Inserm, CNRS, ESPCI Paris, Paris Sciences et Lettres University, Paris, France
| | - Franck P G Lebrin
- Department of Internal Medicine (Nephrology), Einthoven Laboratory for Experimental Vascular Medicine. Leiden University Medical Center, . Leiden, The Netherlands.,Physics for Medicine Paris, Inserm, CNRS, ESPCI Paris, Paris Sciences et Lettres University, Paris, France
| | - Danny Huylebroeck
- Department of Cell Biology, Erasmus University Medical Center, Rotterdam, The Netherlands.,Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Aernout Luttun
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
15
|
Zhang P, Gan Z, Tang L, Zhou L, Huang X, Wang J. WITHDRAWN: Exosomes from microRNA-145-5p-modified HUCB-MSCs attenuate CCl4-induced hepatic fibrosis via down-regulating FSCN1 expression. Life Sci 2021:119404. [PMID: 33794251 DOI: 10.1016/j.lfs.2021.119404] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 03/15/2021] [Accepted: 03/23/2021] [Indexed: 02/06/2023]
Abstract
This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at https://www.elsevier.com/about/our-business/policies/article-withdrawal.
Collapse
Affiliation(s)
- Pan Zhang
- Department of Infection, No.3 Hospital of Xiangya, Central South University, Changsha 410013, Hunan, China
| | - Zeying Gan
- Department of Infection, No.3 Hospital of Xiangya, Central South University, Changsha 410013, Hunan, China
| | - Lanyan Tang
- Department of Infection, No.3 Hospital of Xiangya, Central South University, Changsha 410013, Hunan, China
| | - Lizhi Zhou
- Department of Infection, No.3 Hospital of Xiangya, Central South University, Changsha 410013, Hunan, China
| | - Xin Huang
- Department of Infection, No.3 Hospital of Xiangya, Central South University, Changsha 410013, Hunan, China
| | - Jianlong Wang
- Department of Orthopedics, No.3 Hospital of Xiangya, Central South University, Changsha 410013, Hunan, China.
| |
Collapse
|
16
|
Dostal Z, Sebera M, Srovnal J, Staffova K, Modriansky M. Dual Effect of Taxifolin on ZEB2 Cancer Signaling in HepG2 Cells. Molecules 2021; 26:1476. [PMID: 33803107 PMCID: PMC7963166 DOI: 10.3390/molecules26051476] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 03/02/2021] [Accepted: 03/03/2021] [Indexed: 12/16/2022] Open
Abstract
Polyphenols, secondary metabolites of plants, exhibit different anti-cancer and cytoprotective properties such as anti-radical, anti-angiogenic, anti-inflammation, or cardioprotective. Some of these activities could be linked to modulation of miRNAs expression. MiRNAs play an important role in posttranscriptional regulation of their target genes that could be important within cell signalling or preservation of cell homeostasis, e.g., cell survival/apoptosis. We evaluated the influence of a non-toxic concentration of taxifolin and quercetin on the expression of majority human miRNAs via Affymetrix GeneChip™ miRNA 3.0 Array. For the evaluation we used two cell models corresponding to liver tissue, Hep G2 and primary human hepatocytes. The array analysis identified four miRNAs, miR-153, miR-204, miR-211, and miR-377-3p, with reduced expression after taxifolin treatment. All of these miRNAs are linked to modulation of ZEB2 expression in various models. Indeed, ZEB2 protein displayed upregulation after taxifolin treatment in a dose dependent manner. However, the modulation did not lead to epithelial mesenchymal transition. Our data show that taxifolin inhibits Akt phosphorylation, thereby diminishing ZEB2 signalling that could trigger carcinogenesis. We conclude that biological activity of taxifolin may have ambiguous or even contradictory outcomes because of non-specific effect on the cell.
Collapse
Affiliation(s)
- Zdenek Dostal
- Department of Medical Chemistry and Biochemistry, Faculty of Medicine and Dentistry, Palacký University, 77515 Olomouc, Czech Republic;
| | - Martin Sebera
- Faculty of Sport Studies, Masaryk University, 60177 Brno, Czech Republic;
| | - Josef Srovnal
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, 77515 Olomouc, Czech Republic; (J.S.); (K.S.)
| | - Katerina Staffova
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, 77515 Olomouc, Czech Republic; (J.S.); (K.S.)
| | - Martin Modriansky
- Department of Medical Chemistry and Biochemistry, Faculty of Medicine and Dentistry, Palacký University, 77515 Olomouc, Czech Republic;
| |
Collapse
|
17
|
Circ_0001247 functions as a miR-1270 sponge to accelerate cervical cancer progression by up-regulating ZEB2 expression level. Biotechnol Lett 2021; 43:745-755. [PMID: 33386495 DOI: 10.1007/s10529-020-03059-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 12/10/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND There is increasing evidence that circular RNA (circRNA) disorders have an impact on the progression of various malignancies. The expression characteristics, function and underlying mechanism of circ_0001247 in cervical cancer (CC) have not been confirmed. METHODS GSE147483 datasets of circRNAs expression in CC cell line and normal cervical cell line were retrieved from GEO database, and the circRNA with significant difference was selected; circ_0001247, miR-1270, and Zinc finger E-box binding homeobox 2 (ZEB2) expressions in CC tissues and cell lines were analyzed by quantitative real-time polymerase chain reaction (qRT-PCR) assay; cell counting kit-8 (CCK-8) assay and BrdU assay were applied to monitor the proliferative ability of CC cells; Transwell assay was conducted to examine the migration and invasion of CC cells, and flow cytometry was used to evaluate the apoptosis; Western blot assay was adopted to detect ZEB2 protein expressions; dual-luciferase report gene assay was used to verify the targeting relationship between circ_0001247 and miR-1270, and miR-1270 and the 3'UTR of ZEB2. RESULTS Analysis of GSE147483 suggested that circ_0001247 could probably be an oncogenic circRNA in CC. Compared with that in adjacent tissues and normal cervical epithelial cells, circ_0001247 expression in CC tissues and cell lines was significantly increased; knocking down circ_0001247 expression could inhibit the proliferation and metastasis of CC cells, and promote apoptosis, while circ_0001247 overexpression worked oppositely; circ_0001247 sponged miR-1270 in CC cells; miR-1270 diminished the promoting effect of circ_0001247 by inactivating the ZEB2. CONCLUSION Circ_0001247 promotes progression of CC by sponging miR-1270 to upregulate ZEB2 expression level.
Collapse
|
18
|
Hepigenetics: A Review of Epigenetic Modulators and Potential Therapies in Hepatocellular Carcinoma. BIOMED RESEARCH INTERNATIONAL 2020; 2020:9593254. [PMID: 33299889 PMCID: PMC7707949 DOI: 10.1155/2020/9593254] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 10/13/2020] [Accepted: 11/05/2020] [Indexed: 12/13/2022]
Abstract
Hepatocellular carcinoma is the fifth most common cancer worldwide and the second most lethal, following lung cancer. Currently applied therapeutic practices rely on surgical resection, chemotherapy and radiotherapy, or a combination thereof. These treatment options are associated with extreme adversities, and risk/benefit ratios do not always work in patients' favor. Anomalies of the epigenome lie at the epicenter of aberrant molecular mechanisms by which the disease develops and progresses. Modulation of these anomalous events poses a promising prospect for alternative treatment options, with an abundance of felicitous results reported in recent years. Herein, the most recent epigenetic modulators in hepatocellular carcinoma are recapitulated on.
Collapse
|
19
|
Xu X, Zhang M, Xu F, Jiang S. Wnt signaling in breast cancer: biological mechanisms, challenges and opportunities. Mol Cancer 2020; 19:165. [PMID: 33234169 PMCID: PMC7686704 DOI: 10.1186/s12943-020-01276-5] [Citation(s) in RCA: 348] [Impact Index Per Article: 69.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/22/2020] [Indexed: 02/07/2023] Open
Abstract
Wnt signaling is a highly conserved signaling pathway that plays a critical role in controlling embryonic and organ development, as well as cancer progression. Genome-wide sequencing and gene expression profile analyses have demonstrated that Wnt signaling is involved mainly in the processes of breast cancer proliferation and metastasis. The most recent studies have indicated that Wnt signaling is also crucial in breast cancer immune microenvironment regulation, stemness maintenance, therapeutic resistance, phenotype shaping, etc. Wnt/β-Catenin, Wnt-planar cell polarity (PCP), and Wnt-Ca2+ signaling are three well-established Wnt signaling pathways that share overlapping components and play different roles in breast cancer progression. In this review, we summarize the main findings concerning the relationship between Wnt signaling and breast cancer and provide an overview of existing mechanisms, challenges, and potential opportunities for advancing the therapy and diagnosis of breast cancer.
Collapse
Affiliation(s)
- Xiufang Xu
- School of Medical Imaging, Hangzhou Medical College, Hangzhou, 310053 Zhejiang China
| | - Miaofeng Zhang
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009 Zhejiang China
| | - Faying Xu
- School of Medical Imaging, Hangzhou Medical College, Hangzhou, 310053 Zhejiang China
| | - Shaojie Jiang
- School of Medical Imaging, Hangzhou Medical College, Hangzhou, 310053 Zhejiang China
| |
Collapse
|
20
|
Li G, Ma X, Xu L. The roles of zinc finger proteins in non-alcoholic fatty liver disease. LIVER RESEARCH 2020. [DOI: 10.1016/j.livres.2020.01.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
21
|
Li L, Ran J, Li L, Chen G, Zhang S, Wang Y. Gli3 is a novel downstream target of miR‑200a with an anti‑fibrotic role for progression of liver fibrosis in vivo and in vitro. Mol Med Rep 2020; 21:1861-1871. [PMID: 32319630 PMCID: PMC7057771 DOI: 10.3892/mmr.2020.10997] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 07/09/2019] [Indexed: 12/11/2022] Open
Abstract
GLI family zinc finger 3 (Gli3), as the upstream transcriptional activator of hedgehog signaling, has previously been demonstrated to participate in the process of liver fibrosis. The present study aimed to investigate the potential functions of microRNA (miR)‑200a and Gli3 in the progression of liver fibrosis. The expression levels of miR‑200a and Gli3 in cells and tissues were determined by PCR and western blotting; the interaction of Gli3 and miR‑200a was evaluated by bioinformatics analysis and dual‑luciferase reporter assay. miR‑200a was significantly reduced in serum samples from clinical patients, liver tissues of a carbon tetrachloride (CCl4)‑induced rat model and activated LX2 cells. The expression of α‑smooth muscle actin (α‑SMA) and albumin at the mRNA and protein levels was increased and decreased in LX2 cells, respectively. However, the expression levels of α‑SMA and albumin were reversed and Gli3 expression was markedly decreased in LX2 cells when transfected with miR‑200a mimics. In addition, the dual‑-luciferase reporter assay confirmed the target interaction between miR‑200a and Gli3. Finally, following the administration of miR‑200a mimics to CCl4‑induced rats, it was revealed that the alterations of α‑SMA, albumin and Gli3 presented a similar trend to that in LX2 cells with miR‑200a mimics transfection. Taken together, these results indicated that downregulation of miR‑200a might enhance the formation of liver fibrosis, probably by targeting Gli3, and elevated miR‑200a may attenuate the progression of liver fibrosis by suppressing Gli3. These findings suggested that miR‑200a may function as a novel anti‑fibrotic agent in liver fibrosis via inhibition of the expression of Gli3.
Collapse
Affiliation(s)
- Li Li
- Department of Hepatobiliary Surgery, First People's Hospital of Kunming City, Kunming, Yunnan 650034, P.R. China
| | - Jianghua Ran
- Department of Hepatobiliary Surgery, First People's Hospital of Kunming City, Kunming, Yunnan 650034, P.R. China
| | - Lan Li
- Department of Hepatobiliary Surgery, First People's Hospital of Kunming City, Kunming, Yunnan 650034, P.R. China
| | - Gang Chen
- Department of Hepatobiliary Surgery, First People's Hospital of Kunming City, Kunming, Yunnan 650034, P.R. China
| | - Shengning Zhang
- Department of Hepatobiliary Surgery, First People's Hospital of Kunming City, Kunming, Yunnan 650034, P.R. China
| | - Yingjia Wang
- Department of Hepatobiliary Surgery, First People's Hospital of Kunming City, Kunming, Yunnan 650034, P.R. China
| |
Collapse
|
22
|
Yang J, Tao Q, Zhou Y, Chen Q, Li L, Hu S, Liu Y, Zhang Y, Shu J, Zhang X, Zhang L, Zhang L. MicroRNA-708 represses hepatic stellate cells activation and proliferation by targeting ZEB1 through Wnt/β-catenin pathway. Eur J Pharmacol 2020; 871:172927. [PMID: 31962101 DOI: 10.1016/j.ejphar.2020.172927] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 01/12/2020] [Accepted: 01/13/2020] [Indexed: 02/06/2023]
Abstract
Liver fibrosis is caused by a sustained wound healing response to chronic liver injury, and the activation of insubstantial hepatic stellate cells (HSCs) is the key process involved. The progression of liver fibrosis may be attenuated by suppressing activation and proliferation of the HSCs. MicroRNA (miRNA) have emerged as major players in governing fundamental biological processes through multiple mechanisms MiR-708 is known to inhibit the development of hepatocellular carcinoma. However, whether miR-708 can function as a transcriptional regulator in liver fibrosis remains unclear. Our study demonstrated that miR-708 expression was inhibited in fibrotic liver tissues and in activated HSCs, accompanied by an increase of the Zinc finger E-box binding homeobox 1 (ZEB1) level. Besides, overexpression of miR-708 and silencing of ZEB1 inhibited the activation and proliferation of LX-2 cells. While knockdown of miR-708 or overexpression of ZEB1 showed reversed results. Further, dual luciferase reporter assays showed that miR-708 directly targeted ZEB1 in vitro. Interestingly, ZEB1 was found to be involved in HSCs by regulating Wnt/β-catenin signaling pathway. Together, our data showed that miR-708 may be a potential therapeutic target in liver fibrosis therapy.
Collapse
Affiliation(s)
- Junfa Yang
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China; School of Pharmacy, Anhui Medical University, Hefei, 230032, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, 230032, China; Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, Hefei, 230032, China
| | - Qing Tao
- Department of Pathogen Biology, Anhui Medical University, China
| | - Yiwen Zhou
- School of Pharmacy, Anhui Medical University, Hefei, 230032, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, 230032, China; Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, Hefei, 230032, China
| | - Qingfeng Chen
- Clinic Medical College of Anhui Medical University, Hefei, 230032, China
| | - Liangyun Li
- School of Pharmacy, Anhui Medical University, Hefei, 230032, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, 230032, China; Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, Hefei, 230032, China
| | - Shuang Hu
- School of Pharmacy, Anhui Medical University, Hefei, 230032, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, 230032, China; Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, Hefei, 230032, China
| | - Yumin Liu
- School of Pharmacy, Anhui Medical University, Hefei, 230032, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, 230032, China; Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, Hefei, 230032, China
| | - Yu Zhang
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Jinling Shu
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Xianzheng Zhang
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Lei Zhang
- School of Pharmacy, Anhui Medical University, Hefei, 230032, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, 230032, China; Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, Hefei, 230032, China.
| | - Lingling Zhang
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China.
| |
Collapse
|
23
|
Wang Z, Zhang B, Chen Z, He Y, Ru F, Liu P, Chen X. The long noncoding RNA myocardial infarction-associated transcript modulates the epithelial-mesenchymal transition in renal interstitial fibrosis. Life Sci 2020; 241:117187. [PMID: 31863776 DOI: 10.1016/j.lfs.2019.117187] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 12/13/2019] [Accepted: 12/15/2019] [Indexed: 01/12/2023]
Abstract
AIMS Renal interstitial fibrosis (RIF) is marked by the epithelial-mesenchymal transition (EMT) and excessive extracellular matrix deposition. The long noncoding RNA myocardial infarction-associated transcript (MIAT) facilitates RIF; however, the molecular mechanism of MIAT in RIF remains unclear. Here, we explored the possible underlying mechanisms through which MIAT modulates RIF. MATERIALS AND METHODS MIAT expression in human renal fibrotic tissues and unilateral ureteral obstruction (UUO) model mice was detected by qPCR. Transforming growth factor β1 (TGF-β1) was introduced to stimulate the EMT in human renal proximal tubular epithelial (HK-2) cells. CCK8, EdU, transwell and wound healing assays were employed to measure cell viability, proliferation, and migration respectively. RNA immunoprecipitation (RIP) assays and dual luciferase reporter assays were applied to determine the relationships among MIAT, miR-145, and EIF5A2. KEY FINDINGS MIAT was upregulated in human renal fibrotic tissues and UUO model mice compared with normal tissue adjacent to renal tumors and sham operation mice, respectively. MIAT knockdown reduced cell viability, proliferation, migration, and the EMT in HK-2 cells. Additionally, MIAT served as an endogenous sponge for miR-145 in the TGF-β1-induced-EMT in HK-2 cells, as demonstrated by dual luciferase reporter assays and RIP assays. EIF5A2 was confirmed as a target of miR-145, and MIAT knockdown suppressed EIF5A2 expression by sponging miR-145. Downregulation of EIF5A2 partly reversed induction of the EMT by miR-145 inhibitor transfection. SIGNIFICANCE MIAT promoted cell viability, proliferation, migration, and the EMT via regulation of the miR-145/EIF5A2 axis. These data established a potential therapy for RIF.
Collapse
Affiliation(s)
- Zhaohui Wang
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Bo Zhang
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Zhi Chen
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Yao He
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Feng Ru
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Peihua Liu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, PR China.
| | - Xiang Chen
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, PR China.
| |
Collapse
|
24
|
Li LY, Yang CC, Yang JF, Li HD, Zhang BY, Zhou H, Hu S, Wang K, Huang C, Meng XM, Zhou H, Zhang L, Li J, Xu T. ZEB1 regulates the activation of hepatic stellate cells through Wnt/β-catenin signaling pathway. Eur J Pharmacol 2019; 865:172787. [DOI: 10.1016/j.ejphar.2019.172787] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 10/31/2019] [Accepted: 11/07/2019] [Indexed: 12/31/2022]
|
25
|
Wu J, Huang Q, Li P, Wang Y, Zheng C, Lei X, Li S, Gong W, Yin B, Luo C, Xiao J, Zhou W, Xu Z, Chen Y, Peng F, Long H. MicroRNA-145 promotes the epithelial-mesenchymal transition in peritoneal dialysis-associated fibrosis by suppressing fibroblast growth factor 10. J Biol Chem 2019; 294:15052-15067. [PMID: 31431501 PMCID: PMC6791318 DOI: 10.1074/jbc.ra119.007404] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 08/14/2019] [Indexed: 12/11/2022] Open
Abstract
Peritoneal fibrosis is a common complication of long-term peritoneal dialysis (PD) and the principal cause of ultrafiltration failure during PD. The initial and reversible step in PD-associated peritoneal fibrosis is the epithelial-mesenchymal transition (EMT). Although the mechanisms in the EMT have been the focus of many studies, only limited information is currently available concerning microRNA (miRNA) regulation in peritoneal fibrosis. In this study, we aimed to characterize the roles of microRNA-145 (miR-145) and fibroblast growth factor 10 (FGF10) in peritoneal fibrosis. After inducing EMT with transforming growth factor-β1 (TGF-β1) in vitro, we found that miR-145 is significantly up-regulated, whereas FGF10 is markedly down-regulated, suggesting a close link between miR-145 and FGF10 in peritoneal fibrosis, further confirmed in luciferase reporter experiments. Furthermore, in human peritoneal mesothelial cells (i.e. HMrSV5 cells), miR-145 mimics induced EMT, whereas miR-145 inhibition suppressed EMT, and we also observed that miR-145 suppressed FGF10 expression. In vivo, we found that the exogenous delivery of an miR-145 expression plasmid both blocked FGF10 and intensified the EMT, whereas miR-145 inhibition promoted the expression of FGF10 and reversed the EMT. In conclusion, miR-145 promotes the EMT during the development of peritoneal fibrosis by suppressing FGF10 activity, suggesting that miR-145 represents a potential therapeutic target for managing peritoneal fibrosis.
Collapse
Affiliation(s)
- Jiayu Wu
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Qianyin Huang
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Peilin Li
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yuxian Wang
- Department of Gerontology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Chenghao Zheng
- Second Clinical Medical School, Southern Medical University, Guangzhou 510280, China
| | - Xianghong Lei
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Shuting Li
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Wangqiu Gong
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Bohui Yin
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Congwei Luo
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Jing Xiao
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Weidong Zhou
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Zhaozhong Xu
- Department of Emergency, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yihua Chen
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Fenfen Peng
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Haibo Long
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| |
Collapse
|
26
|
Shan L, Liu Z, Ci L, Shuai C, Lv X, Li J. Research progress on the anti-hepatic fibrosis action and mechanism of natural products. Int Immunopharmacol 2019; 75:105765. [PMID: 31336335 DOI: 10.1016/j.intimp.2019.105765] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Accepted: 07/15/2019] [Indexed: 12/15/2022]
Abstract
Hepatic fibrosis is the most common pathological feature of most chronic liver diseases, and its continuous deterioration gradually develops into liver cirrhosis and eventually leads to liver cancer. At present, there are many kinds of drugs used to treat liver fibrosis. However, Western drugs tend to only target single genes/proteins and induce many adverse reactions. Most of the mechanisms and active ingredients of traditional Chinese medicine (TCM) are not clear, and there is a lack of unified diagnosis and treatment standards. Natural products, which are characterized by structural diversity, low toxicity, and origination from a wide range of sources, have unique advantages and great potential in anti-liver fibrosis. This article summarizes the work done over the previous decade, on the active ingredients in natural products that are reported to have anti-hepatic fibrosis effects. The effective anti-hepatic fibrosis ingredients identified can be generally divided into flavonoids, saponins, polysaccharides and alkaloids. Mechanisms of anti-liver fibrosis include inhibition of liver inflammation, anti-lipid peroxidation injury, inhibition of the activation and proliferation of hepatic stellate cells (HSCs), modulation of the synthesis and secretion of pro-fibrosis factors, and regulation of the synthesis and degradation of the extracellular matrix (ECM). This review provides suggestions for the development of anti-hepatic fibrosis drugs.
Collapse
Affiliation(s)
- Liang Shan
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China; The Key laboratory of Anti-inflammatory and Immune medicines, Ministry of Education Hefei, China; Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Zhenni Liu
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China; The Key laboratory of Anti-inflammatory and Immune medicines, Ministry of Education Hefei, China; Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Leilei Ci
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China; The Key laboratory of Anti-inflammatory and Immune medicines, Ministry of Education Hefei, China; Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Chen Shuai
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China; The Key laboratory of Anti-inflammatory and Immune medicines, Ministry of Education Hefei, China; Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Xiongwen Lv
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China; The Key laboratory of Anti-inflammatory and Immune medicines, Ministry of Education Hefei, China; Institute for Liver Diseases of Anhui Medical University, Hefei, China.
| | - Jun Li
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China; The Key laboratory of Anti-inflammatory and Immune medicines, Ministry of Education Hefei, China; Institute for Liver Diseases of Anhui Medical University, Hefei, China
| |
Collapse
|
27
|
Cui J, Pan G, He Q, Yin L, Guo R, Bi H. MicroRNA-545 targets ZEB2 to inhibit the development of non-small cell lung cancer by inactivating Wnt/β-catenin pathway. Oncol Lett 2019; 18:2931-2938. [PMID: 31452774 PMCID: PMC6676444 DOI: 10.3892/ol.2019.10619] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 06/10/2019] [Indexed: 02/06/2023] Open
Abstract
The specific function of microRNA-545 (miR-545) has been reported to regulate the development of human cancers. However, the effect of miR-545 is still unclear in non-small cell lung cancer (NSCLC). Hence, this study explored the molecular mechanism of miR-545 in NSCLC. The expression levels of miR-545 and ZEB2 were measured through reverse transcription-quantitative polymerase chain reaction (RT-qPCR) assay. The protein expression was detected by western blotting. Dual luciferase assay was applied to evaluate the relationship between miR-545 and zinc finger E-box-binding homeobox 2 (ZEB2). MTT and Transwell assays were used to investigate the function of miR-545 in NSCLC. The expression of miR-545 was decreased in NSCLC tissues. The overexpression of miR-545 suppressed the migration, invasion and proliferation of NSCLC cells. Furthermore, ZEB2 was a direct target gene of miR-545. The knockout of ZEB2 suppressed the development of NSCLC. miR-545 inhibited the progression of NSCLC through targeting ZEB2. Moreover, miR-545 repressed the development of NSCLC via blocking EMT and Wnt/β-catenin pathway. In conclusion, miR-545 inhibited the progression of NSCLC through targeting ZEB2 and blocking EMT and Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Jianying Cui
- Department of Respiratory Medicine, Anqiu People's Hospital, Weifang, Shandong 262100, P.R. China
| | - Guodong Pan
- Department of Thoracic Surgery, Traditional Chinese Medical Hospital of Huangdao District, Qingdao, Shandong 266500, P.R. China
| | - Qingjuan He
- Department of Respiratory Medicine, The People's Hospital of Zhangqiu Area, Jinan, Shandong 250200, P.R. China
| | - Lizhi Yin
- Health Management Center, The People's Hospital of Zhangqiu Area, Jinan, Shandong 250200, P.R. China
| | - Rui Guo
- Outpatient Department, The People's Hospital of Zhangqiu Area, Jinan, Shandong 250200, P.R. China
| | - Hongmei Bi
- Department of Respiratory Medicine, The Third People's Hospital of Qingdao, Qingdao, Shandong 266041, P.R. China
| |
Collapse
|
28
|
Liu Q, Chen J, Wang B, Zheng Y, Wan Y, Wang Y, Zhou L, Liu S, Li G, Yan Y. Retracted: miR-145 modulates epithelial-mesenchymal transition and invasion by targeting ZEB2 in non-small cell lung cancer cell lines. J Cell Biochem 2019; 120:8409-8418. [PMID: 30525209 DOI: 10.1002/jcb.28126] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 10/31/2018] [Indexed: 01/24/2023]
Abstract
Lung cancer is the leading cause of cancer-related deaths worldwide. Epithelial-mesenchymal transition (EMT) is a major event that drives cancer progression. Here we aim to investigate the role of microRNA, miR-145, in regulating EMT of the highly invasive non-small cell lung cancer (NSCLC). Quantitative real-time polymerase chain reaction analysis indicated that miR-145 was downregulated in cancer tissue compared with that in adjacent normal tissue. NSCLC cell lines, namely H1299, PC7, and SPCA-1, also demonstrated miR-145 downregulation, which is correlated well with their invasive ability, assessed by the Matrigel invasion assay. miR-145 overexpression resulted in downregulation of N-cadherin, and downregulation of vimentin and E-cadherin, suggesting a decreased EMT activity. TargetScan analysis predicted that a binding site exists between miR-145 and an oncogene, ZEB2, which was verified using the dual-luciferase assay. Alteration of miR-145 expression also induced inverse effects on ZEB2 expression, and a negative correlation exists between ZEB2 and miR-145 in human tissues. ZEB2 and miR-145 also exerted antagonizing effects on the invasion of NSCLC cells. Therefore, miR-145 is an important molecule in NSCLC that regulates cancer EMT through targeting ZEB2.
Collapse
Affiliation(s)
- Qun Liu
- Medical Ward 20, Lianshui County People's Hospital, Huai'an, Jiangsu, China
| | - Jianhui Chen
- Tongji University School of Medicine, Shanghai, China
- Department of Respiratory and Critical Care Medicine, Huai'an Second People's Hospital and The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu, China
| | - Baolan Wang
- Department of Respiratory Medicine, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, China
| | - Yulong Zheng
- Department of Respiratory and Critical Care Medicine, Huai'an Second People's Hospital and The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu, China
| | - Yufeng Wan
- Department of Respiratory and Critical Care Medicine, Huai'an Second People's Hospital and The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu, China
| | - Yi Wang
- Department of Respiratory and Critical Care Medicine, Huai'an Second People's Hospital and The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu, China
| | - Liyang Zhou
- Department of Respiratory and Critical Care Medicine, Huai'an Second People's Hospital and The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu, China
| | - Shu Liu
- Department of Respiratory and Critical Care Medicine, Huai'an Second People's Hospital and The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu, China
| | - Gang Li
- Department of Respiratory and Critical Care Medicine, Huai'an Second People's Hospital and The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu, China
| | - Yi Yan
- Department of Respiratory Medicine, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
29
|
Wang YZ, Zhang W, Wang YH, Fu XL, Xue CQ. Repression of liver cirrhosis achieved by inhibitory effect of miR-454 on hepatic stellate cells activation and proliferation via Wnt10a. J Biochem 2019; 165:361-367. [PMID: 30535384 DOI: 10.1093/jb/mvy111] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 12/06/2018] [Indexed: 12/30/2022] Open
Abstract
As is known, hepatic stellate cells (HSCs) activation contributes to liver cirrhosis. This study aims to find out the acting mechanisms of miR-454 inhibiting the activation and proliferation of hepatic stellate cells. The expression of Col1A1, α-smooth muscle actin (α-SMA) and Wnt10a were determined by western blot, and the miR-454 level was determined by quantitative real-time PCR in this study. We took two objects as experiment subjects, one was liver cirrhosis rats, and the other was transforming growth factor (TGF)-β1-stimulated HSC-T6 cells. After activated with TGF-β1 and transfected with microRNA-454 mimic, separately or successively, the changes on the Col1A1 and α-SMA expression, HSC proliferation, miR-454 level and Wnt10a expression were examined in HSC-T6 cells, respectively. Interaction between miR-454 and Wnt10a was evaluated with dual luciferase reporter assay. MiR-454 expression was down-regulated in tissues of liver cirrhosis rats. TGF-β1 caused the down-regulation of the miR-454 in HSC-T6 cells. MiR-454 inhibited the activation and proliferation of HSC-T6 cells. Wnt10a had a targeting relationship with miR-454. TGF-β1 promoted HSC-T6 activation and proliferation via down-regulating miR-454 expression, which further up-regulated Wnt10a expression. MiR-454 mimic inhibited cirrhosis progression in liver cirrhosis rats. MiR-454 can inhibit the activation and proliferation of HSCs via suppressing the expression of Wnt10a, to restrain liver cirrhosis.
Collapse
Affiliation(s)
- Yong-Zhen Wang
- Department of Interventional Radiology and Vascular Surgery, Nanjing Second Hospital, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wei Zhang
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan
| | - Yan-Hua Wang
- Department of Ultrasound, Nanjing Second Hospital, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xi-Lin Fu
- Department of Hepatopathy, Nanjing Second Hospital, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chen-Qi Xue
- Department of Interventional Radiology and Vascular Surgery, Nanjing Second Hospital, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
30
|
Perugorria MJ, Olaizola P, Labiano I, Esparza-Baquer A, Marzioni M, Marin JJG, Bujanda L, Banales JM. Wnt-β-catenin signalling in liver development, health and disease. Nat Rev Gastroenterol Hepatol 2019; 16:121-136. [PMID: 30451972 DOI: 10.1038/s41575-018-0075-9] [Citation(s) in RCA: 390] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The canonical Wnt-β-catenin pathway is a complex, evolutionarily conserved signalling mechanism that regulates fundamental physiological and pathological processes. Wnt-β-catenin signalling tightly controls embryogenesis, including hepatobiliary development, maturation and zonation. In the mature healthy liver, the Wnt-β-catenin pathway is mostly inactive but can become re-activated during cell renewal and/or regenerative processes, as well as in certain pathological conditions, diseases, pre-malignant conditions and cancer. In hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA), the two most prevalent primary liver tumours in adults, Wnt-β-catenin signalling is frequently hyperactivated and promotes tumour growth and dissemination. A substantial proportion of liver tumours (mainly HCC and, to a lesser extent, CCA) have mutations in genes encoding key components of the Wnt-β-catenin signalling pathway. Likewise, hepatoblastoma, the most common paediatric liver cancer, is characterized by Wnt-β-catenin activation, mostly as a result of β-catenin mutations. In this Review, we discuss the most relevant molecular mechanisms of action and regulation of Wnt-β-catenin signalling in liver development and pathophysiology. Moreover, we highlight important preclinical and clinical studies and future directions in basic and clinical research.
Collapse
Affiliation(s)
- Maria J Perugorria
- Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute - Donostia University Hospital - University of the Basque Country (UPV/EHU), San Sebastian, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Institute of Health (ISCIII), Madrid, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Paula Olaizola
- Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute - Donostia University Hospital - University of the Basque Country (UPV/EHU), San Sebastian, Spain
| | - Ibone Labiano
- Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute - Donostia University Hospital - University of the Basque Country (UPV/EHU), San Sebastian, Spain
| | - Aitor Esparza-Baquer
- Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute - Donostia University Hospital - University of the Basque Country (UPV/EHU), San Sebastian, Spain
| | - Marco Marzioni
- Department of Gastroenterology, Università Politecnica delle Marche, Ancona, Italy
| | - Jose J G Marin
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Institute of Health (ISCIII), Madrid, Spain
- Experimental Hepatology and Drug Targeting (HEVEFARM), Biomedical Research Institute of Salamanca (IBSAL), University of Salamanca, Salamanca, Spain
| | - Luis Bujanda
- Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute - Donostia University Hospital - University of the Basque Country (UPV/EHU), San Sebastian, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Institute of Health (ISCIII), Madrid, Spain
| | - Jesus M Banales
- Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute - Donostia University Hospital - University of the Basque Country (UPV/EHU), San Sebastian, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Institute of Health (ISCIII), Madrid, Spain.
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain.
| |
Collapse
|
31
|
Verma A, Bennett J, Örme AM, Polycarpou E, Rooney B. Cocaine addicted to cytoskeletal change and a fibrosis high. Cytoskeleton (Hoboken) 2019; 76:177-185. [PMID: 30623590 DOI: 10.1002/cm.21510] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 12/05/2018] [Accepted: 12/18/2018] [Indexed: 12/17/2022]
Abstract
Cocaine is one of the most widely abused illicit drugs due to its euphoric and addictive properties. Cocaine-mediated cognitive impairments are the result of dynamic cytoskeletal rearrangements involved in mediating structural and behavioural plasticity. Cytoskeletal changes initiated following cocaine abuse are regulated by the Rho family of GTPases with significant downstream activity in key actin binding proteins. Moreover, signalling via the endoplasmic reticulum chaperone protein, sigma-1 receptor has highlighted the possibility of cocaine regulated pathology in other organ systems. However, the question of whether upstream stimulation of such a high affinity binding receptor is directly involved in cocaine-mediated cytoskeletal changes at present remains unknown. In this review, we describe the functional role of key cytoskeletal regulators in response to cocaine-induced signalling cues. In addition, we ascertain the extent of whether global cytoskeletal modulators involved in cocaine-induced neurological stimulation can be used as a platform for future studies into elucidating its fibrotic potential within the hepatic microenvironment. A focus on aspects still poorly understood relating to the nonneuronal pathological impact of cocaine is discussed in the sphere of hepatic dysregulation. Lastly, we suggest that cocaine may mediate its pathological capacity via the sigma1 receptor in regulating hepatoxicity, hepatic stellate cells activity, cytoskeletal dynamics, and the transcriptional regulation of key hepato-fibrogenic modulators.
Collapse
Affiliation(s)
- Avnish Verma
- Kingston University, Department of Applied and Human Sciences, School of Life Sciences, Pharmacy and Chemistry, Surrey, United Kingdom
| | - Jason Bennett
- Centre for Cell Signalling and Inflammation, Department of Medicine, Imperial College London, London, United Kingdom
| | - Ayşe Merve Örme
- Kingston University, Department of Applied and Human Sciences, School of Life Sciences, Pharmacy and Chemistry, Surrey, United Kingdom
| | - Elena Polycarpou
- Kingston University, Department of Applied and Human Sciences, School of Life Sciences, Pharmacy and Chemistry, Surrey, United Kingdom
| | - Brian Rooney
- Kingston University, Department of Applied and Human Sciences, School of Life Sciences, Pharmacy and Chemistry, Surrey, United Kingdom
| |
Collapse
|
32
|
Wang F, Zhu W, Yang R, Xie W, Wang D. LncRNA ZEB2-AS1 contributes to the tumorigenesis of gastric cancer via activating the Wnt/β-catenin pathway. Mol Cell Biochem 2019; 456:73-83. [PMID: 30635820 DOI: 10.1007/s11010-018-03491-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Accepted: 12/22/2018] [Indexed: 12/29/2022]
Abstract
Studies have shown that long noncoding RNA Zinc finger E-box-binding homeobox 2 antisense RNA 1 (ZEB2-AS1) is involved in the progression of lung cancer, bladder cancer, and hepatocellular carcinoma. However, its role in the pathogenesis of gastric cancer remains unknown. The Wnt/β-catenin pathway contributes to the development of gastric cancer. ZEB2-AS1 expression was firstly detected in the gastric carcinoma tissue samples as well as in gastric cancer cells. Knockdown of ZEB2-AS1 was performed by ZEB2-AS1-shRNA, and the viability, migration, invasion, and apoptosis of gastric cancer cells were determined by CCK-8, scratch assay, transwell, and flow cytometry, respectively. Furthermore, levels of Ki-67, PCNA, VEGF, MMP9, epithelial-mesenchymal transition (EMT) markers (E-cadherin, Vimentin and ZEB2), cleaved caspase 3/8/9 and PARP, active β-catenin, c-Myc, cyclinD1, and AXIN2 were assayed by Western blot or real-time PCR. Additionally, the role and mechanism of ZEB2-AS1 were confirmed in a xenograft nude mouse model. We found ZEB2-AS1 expression was increased in gastric carcinoma samples, and it was correlated with tumor progression. Also, its expression was elevated in gastric cancer cells. Knockdown of ZEB2-AS1 reduced the proliferation, migration, invasion, and EMT, but increased the apoptosis of gastric carcinoma cells. Furthermore, ZEB2-AS1 downregulation remarkably suppressed the expression of Ki-67, PCNA, VEGF and MMP9, and the activation of Wnt/β-catenin signaling, whereas elevated the levels of cleaved caspase 3/8/9 and PARP in gastric cancer cells. And ZEB2 overexpression reversed the effects of ZEB2-AS1 downregulation on the proliferation, EMT and inactivation of Wnt/β-catenin signaling. Additionally, ZEB2-AS1 knockdown inhibited tumor growth, Ki-67 staining, and the expression of VEGF, MMP9, active β-catenin, c-Myc, cyclinD1, and AXIN2 in mice. In conclusion, ZEB2-AS1 promotes the tumorigenesis of gastric carcinoma that is related to the upregulation of ZEB2 and the activation of the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Feixia Wang
- Department of Clinical Laboratory, Chang'an Hospital, Xi'an, 710016, Shaanxi, People's Republic of China
| | - Wu Zhu
- Department of Clinical Laboratory, Ankang Municipality of Traditional Chinese Medicine Hospital, No. 47, East Bashan Road, Ankang, 725000, Shaanxi, People's Republic of China
| | - Ruili Yang
- Department of Clinical Laboratory, Chang'an Hospital, Xi'an, 710016, Shaanxi, People's Republic of China
| | - Wanhua Xie
- Department of Clinical Laboratory, Ankang Municipality of Traditional Chinese Medicine Hospital, No. 47, East Bashan Road, Ankang, 725000, Shaanxi, People's Republic of China
| | - Daojun Wang
- Department of Clinical Laboratory, Ankang Municipality of Traditional Chinese Medicine Hospital, No. 47, East Bashan Road, Ankang, 725000, Shaanxi, People's Republic of China.
| |
Collapse
|
33
|
Zhang X, Xu X, Ge G, Zang X, Shao M, Zou S, Zhang Y, Mao Z, Zhang J, Mao F, Qian H, Xu W. miR‑498 inhibits the growth and metastasis of liver cancer by targeting ZEB2. Oncol Rep 2018; 41:1638-1648. [PMID: 30592286 PMCID: PMC6365765 DOI: 10.3892/or.2018.6948] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 12/05/2018] [Indexed: 01/27/2023] Open
Abstract
MicroRNAs (miRNAs) play critical roles in the growth, metastasis and therapeutic resistance of liver cancer. Accumulating evidence suggests that miR-498 is aberrantly expressed in several human malignancies. However, the role and underlying mechanism of miR-498 in liver cancer remain unclear. In the present study, we investigated the potential roles and clinical value of miR-498 in liver cancer. We found that the miR-498 expression level was significantly lower in liver cancer patient tissues than that in healthy control tissues. The expression of miR-498 was also decreased in liver cancer cell lines compared to that noted in a normal human normal liver cell line. miR-498 overexpression markedly inhibited liver cancer cell proliferation, migration and invasion. miR-498 overexpression induced cell cycle arrest and apoptosis while it suppressed epithelial-mesenchymal transition (EMT) in liver cancer cells. Bioinformatic analysis and luciferase reporter assay further identified zinc finger E-box binding homeobox 2 (ZEB2) as a novel target of miR-498. Furthermore, ZEB2 knockdown recapitulated the inhibitory effects of miR-498 overexpression in liver cancer cells. ZEB2 overexpression rescued the inhibition of liver cancer cell proliferation, migration, and invasion by miR-498, indicating that ZEB2 acts as a downstream effector of miR-498 in liver cancer cells. Thus, we demonstrated that miR-498 suppresses the growth and metastasis of liver cancer cells, partly at least, by directly targeting ZEB2, suggesting that miR-498 may serve as a potential biomarker for the diagnosis and therapy of liver cancer.
Collapse
Affiliation(s)
- Xu Zhang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Xueying Xu
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Guohong Ge
- Liver Disease and Cancer Institute, The Affiliated Zhenjiang Third Hospital of Jiangsu University, Zhenjiang, Jiangsu 212021, P.R. China
| | - Xueyan Zang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Meng Shao
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Shengqiang Zou
- Liver Disease and Cancer Institute, The Affiliated Zhenjiang Third Hospital of Jiangsu University, Zhenjiang, Jiangsu 212021, P.R. China
| | - Yu Zhang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Zheying Mao
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Jiayin Zhang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Fei Mao
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Hui Qian
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Wenrong Xu
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| |
Collapse
|
34
|
Yang J, Lu Y, Yang P, Chen Q, Wang Y, Ding Q, Xu T, Li X, Li C, Huang C, Meng X, Li J, Zhang L, Wang X. MicroRNA-145 induces the senescence of activated hepatic stellate cells through the activation of p53 pathway by ZEB2. J Cell Physiol 2018; 234:7587-7599. [PMID: 30479019 DOI: 10.1002/jcp.27521] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 09/10/2018] [Indexed: 12/11/2022]
Abstract
Activation of quiescent hepatic stellate cells (HSCs) is the major event in liver fibrosis, along with enhancement of cell proliferation and overproduction of extracellular matrix. Recent findings suggest that senescence of activated HSCs might limit the development of liver fibrosis. The p53, a guardian of the genome is associated with liver fibrosis, has been shown to regulate HSCs senescence. In this study, we report that microRNA-145 (miR-145) and p53 were downregulated in vivo and in vitro, concomitant with the enhanced expression of zinc finger E-box binding homeobox 2 (ZEB2). In addition, overexpression of miR-145 and p53 led to upregulation of the number of senescence-associated β-galactosidase-positive HSCs and the expression of senescence markers p16 and p21, along with the reduced abundance of HSC activation markers α-smooth muscle actin and type I collagen in activated HSCs. Furthermore, silencing of ZEB2 promoted senescence of activated HSCs. Moreover, we also demonstrated that miR-145 specifically targeted the 3'-untranslated regions of ZEB2. In vitro promoter regulation studies show that ZEB2 could bind to the E-box of the p53 promoter as well as inhibit its promoter activity and thus suppress the expression of p53, which in turn repressed activated HSCs senescence. Taken together, our results describe a novel miR-145-ZEB2-p53 regulatory line might participate in the senescence of activated HSCs and might carry potential therapeutic targets for restraining liver fibrosis.
Collapse
Affiliation(s)
- Junfa Yang
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, Hefei, China
| | - Yuchen Lu
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, Hefei, China
| | - Peipei Yang
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, Hefei, China
| | - Qingfeng Chen
- Department of Clinical Medicine, Clinic Medical College of Anhui Medical University, Hefei, China
| | - Yang Wang
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, Hefei, China
| | - Qi Ding
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, Hefei, China
| | - Tao Xu
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, Hefei, China
| | - Xiaofeng Li
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, Hefei, China
| | - Changyao Li
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, Hefei, China
| | - Cheng Huang
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, Hefei, China
| | - Xiaoming Meng
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, Hefei, China
| | - Jun Li
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, Hefei, China
| | - Lei Zhang
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, Hefei, China
| | - Xiao Wang
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
35
|
Octreotide attenuates hepatic fibrosis and hepatic stellate cells proliferation and activation by inhibiting Wnt/β-catenin signaling pathway, c-Myc and cyclin D1. Int Immunopharmacol 2018; 63:183-190. [DOI: 10.1016/j.intimp.2018.08.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 08/01/2018] [Accepted: 08/06/2018] [Indexed: 12/20/2022]
|
36
|
Zhu D, Yang C, Shen P, Chen L, Chen J, Sun X, Duan L, Zhang L, Zhu J, Duan Y. rSjP40 suppresses hepatic stellate cell activation by promoting microRNA-155 expression and inhibiting STAT5 and FOXO3a expression. J Cell Mol Med 2018; 22:5486-5493. [PMID: 30091834 PMCID: PMC6201359 DOI: 10.1111/jcmm.13819] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Revised: 06/12/2018] [Accepted: 06/29/2018] [Indexed: 01/09/2023] Open
Abstract
Activation of hepatic stellate cells (HSCs) is the central event of the evolution of hepatic fibrosis. Schistosomiasis is one of the pathogenic factors which could induce hepatic fibrosis. Previous studies have shown that recombinant Schistosoma japonicum egg antigen P40 (rSjP40) can inhibit the activation and proliferation of HSCs. MicroRNA‐155 is one of the multifunctional noncoding RNA, which is involved in a series of important biological processes including cell development, proliferation, differentiation and apoptosis. Here, we try to observe the role of microRNA‐155 in rSjP40‐inhibited HSC activation and explore its potential mechanisms. We found that microRNA‐155 was raised in rSjP40‐treated HSCs, and further studies have shown that rSjP40 enhanced microRNA‐155 expression by inhibiting STAT5 transcription. Up‐regulated microRNA‐155 can down‐regulate the expression of FOXO3a and then participate in rSjP40‐inhibited expression of α‐smooth muscle actin (α‐SMA) and collagen I. Furthermore, we observed microRNA‐155 inhibitor could partially restore the down‐regulation of FOXO3a, α‐SMA and collagen I expression in LX‐2 cells induced by rSjP40. Therefore, our research provides further insight into the mechanism by which rSjP40 could inhibit HSC activation via miR‐155.
Collapse
Affiliation(s)
- Dandan Zhu
- Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, China
| | - Chunzhao Yang
- Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, China
| | - Pei Shen
- Laboratory Medicine Center, Affiliated Hospital of Nantong University, Nantong, China
| | - Liuting Chen
- Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, China
| | - Jinling Chen
- Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, China
| | - Xiaolei Sun
- Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, China
| | - Lian Duan
- Department of Medical Informatics, School of Medicine, Nantong University, Nantong, China
| | - Li Zhang
- Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, China
| | - Jinhua Zhu
- Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, China
| | - Yinong Duan
- Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, China
| |
Collapse
|
37
|
Zhu D, Hu B, Zhou Y, Sun X, Chen J, Chen L, Ji Z, Zhu J, Duan Y. microRNA-146a is involved in rSjP40-inhibited activation of LX-2 cells by targeting Smad4 expression. J Cell Biochem 2018; 119:9249-9253. [PMID: 29953648 DOI: 10.1002/jcb.27193] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 05/24/2018] [Indexed: 12/31/2022]
Abstract
Previous studies have demonstrated that the recombinant Schistosoma japonicum protein P40 (rSjP40) could inhibit activation of hepatic stellate cells (HSCs) through the TGF-β1/Smads signaling pathway. Since multiple microRNAs could play essential roles in HSC activation and in the process of hepatic fibrosis through targeting Smads, we attempted to seek the potential microRNAs that could be involved in rSjP40-induced inhibition of HSC activation. Using the method of quantitative real-time PCR, we found that rSjP40 could induce miR-146a expression in LX-2 cells. The down-regulated expression levels of Smad4 and α-SMA in LX-2 cells induced by rSjP40 were partially restored by an miR-146a inhibitor. miR-146a can be involved in rSjP40-induced inhibition of HSC activation through targeting Smad4. These findings provide us a new idea to explore the potential mechanisms by which rSjP40 could regulate the process of hepatic fibrosis.
Collapse
Affiliation(s)
- Dandan Zhu
- Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, Jiangsu, China
| | - Bin Hu
- Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, Jiangsu, China
| | - Yonghua Zhou
- Key Laboratory of National Health and Family Planning Commission on Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi, Jiangsu, China
| | - Xiaolei Sun
- Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, Jiangsu, China
| | - Jinling Chen
- Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, Jiangsu, China
| | - Liuting Chen
- Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, Jiangsu, China
| | - Zhaodong Ji
- Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, Jiangsu, China
| | - Jinhua Zhu
- Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, Jiangsu, China
| | - Yinong Duan
- Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
38
|
Feng L, Shi L, Lu YF, Wang B, Tang T, Fu WM, He W, Li G, Zhang JF. Linc-ROR Promotes Osteogenic Differentiation of Mesenchymal Stem Cells by Functioning as a Competing Endogenous RNA for miR-138 and miR-145. MOLECULAR THERAPY-NUCLEIC ACIDS 2018; 11:345-353. [PMID: 29858070 PMCID: PMC5992460 DOI: 10.1016/j.omtn.2018.03.004] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 03/07/2018] [Accepted: 03/08/2018] [Indexed: 12/16/2022]
Abstract
Long noncoding RNAs (lncRNAs), which serve as important and powerful regulators of various biological activities, have gained widespread attention in recent years. Emerging evidence has shown that some lncRNAs play important regulatory roles in osteoblast differentiation of mesenchymal stem cells (MSCs), suggesting a potential therapeutic strategy for bone fracture. As a recently identified lncRNA, linc-ROR was reported to mediate the reprogramming ability of differentiated cells into induced pluripotent stem cells (iPSCs) and human embryonic stem cells (ESCs) self-renewal. However, other functions of linc-ROR remain elusive. In this study, linc-ROR was found to be upregulated during osteogenesis of human bone-marrow-derived MSCs. Ectopic expression of linc-ROR significantly accelerated, whereas knockdown of linc-ROR suppressed, osteoblast differentiation. Using bioinformatic prediction and luciferase reporter assays, we demonstrated that linc-ROR functioned as a microRNA (miRNA) sponge for miR-138 and miR-145, both of which were negative regulators of osteogenesis. Further investigations revealed that linc-ROR antagonized the functions of these two miRNAs and led to the de-repression of their shared target ZEB2, which eventually activated Wnt/β-catenin pathway and hence potentiated osteogenesis. Taken together, linc-ROR modulated osteoblast differentiation by acting as a competing endogenous RNA (ceRNA), which may shed light on the functional characterization of lncRNAs in coordinating osteogenesis.
Collapse
Affiliation(s)
- Lu Feng
- Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences and Lui Che Woo Institute of Innovative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China
| | - Liu Shi
- Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences and Lui Che Woo Institute of Innovative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China
| | - Ying-Fei Lu
- Central Laboratory, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu 211100, China
| | - Bin Wang
- Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences and Lui Che Woo Institute of Innovative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China
| | - Tao Tang
- Department of Obstetrics & Gynaecology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Wei-Ming Fu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Wei He
- Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Gang Li
- Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences and Lui Che Woo Institute of Innovative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China.
| | - Jin-Fang Zhang
- Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China; Laboratory of Orthopaedics & Traumatology, Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
39
|
Sathyanarayanan A, Chandrasekaran KS, Karunagaran D. microRNA-145 downregulates SIP1-expression but differentially regulates proliferation, migration, invasion and Wnt signaling in SW480 and SW620 cells. J Cell Biochem 2018; 119:2022-2035. [PMID: 28833449 DOI: 10.1002/jcb.26365] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 08/16/2017] [Indexed: 01/03/2025]
Abstract
microRNA-145 (miR-145) has been shown to act as a tumor suppressor in colorectal cancer but its role in the regulation of epithelial-mesenchymal transition (EMT) is unclear. Ectopic expression of miR-145 suppressed the proliferation, migration and invasion in SW480 but surprisingly enhanced these traits in its metastatic counterpart, SW620 cells, while, antimiR-145 reversed the effects of miR-145 in both of these human colorectal cancer cells. In SW480 and SW620 cells, SMAD-interacting protein 1 (SIP1), was identified as a target of miR-145, and its expression was suppressed both at mRNA and protein levels, and siRNA-SIP1 mimicked the effects of miR-145. Further, re-introduction of SIP1 alone or its co-expression with miR-145, rescued SW480 and SW620 cells from the effects of miR-145, indicating that the distinct functions of miR-145 might be mediated, in part, through SIP1. Since Wnt signaling plays an essential role in EMT in CRC progression, the effects of miR-145 on the expression of Wnt signaling intermediates and EMT markers were studied. Re-expression of miR-145 was found to downregulate the expression of CTNNB1, TCF4, CCND1, VIM, and SNAI, but, upregulated CDH1 expression in SW480 cells. On the other hand, miR-145 exhibited an oncogenic potential in SW620 cells by actuating Wnt signaling and the expression of EMT-relevant markers. These results strongly hint that the paradoxical functions of miR-145 in the regulation of proliferation, migration and invasion might be mediated through downregulation of SIP1, and differential tuning of Wnt signaling and EMT-mediators.
Collapse
Affiliation(s)
- Anusha Sathyanarayanan
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, Tamil Nadu, India
| | - Karthik Subramanian Chandrasekaran
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, Tamil Nadu, India
| | - Devarajan Karunagaran
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, Tamil Nadu, India
| |
Collapse
|
40
|
Yang J, Liu Q, Cao S, Xu T, Li X, Zhou D, Pan L, Li C, Huang C, Meng X, Zhang L, Wang X. MicroRNA-145 Increases the Apoptosis of Activated Hepatic Stellate Cells Induced by TRAIL through NF-κB Signaling Pathway. Front Pharmacol 2018; 8:980. [PMID: 29375381 PMCID: PMC5770373 DOI: 10.3389/fphar.2017.00980] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 12/21/2017] [Indexed: 12/18/2022] Open
Abstract
During the liver fibrosis recovery stage tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) can effectively induce apoptosis of activated hepatic stellate cells (HSCs). Normal hepatic stellate cells are resistant to TRAIL cytotoxicity. Therefore, enhancing the sensitivity of TRAIL-induced apoptosis of HSCs may be useful to treat hepatic fibrogenesis. Here, we demonstrated that miR-145 and TRAIL were down-regulated in both liver fibrosis tissue samples and transforming growth factor-β1 induced HSCs, concomitant with increased the expression of ZEB2. In addition, we found that mimics-mediated over-expression of miR-145 led to resistance to the ZEB2 expression and up-regulation of the TRAIL-induced apoptosis after treatment of LX-2 cells with TRAIL. Furthermore, ZEB2-siRNA transfected LX-2 cells showed the increased sensitivity to TRAIL-induced apoptosis. Whereas, opposite results were obtained in miR-145-inhibitor group or ZEB2 plasmid group. Moreover, miR-145 regulated ZEB2 gene expression by specifically interacting with the 3′-UTR of ZEB2 mRNA to inhibit the expression of ZEB2. Further studies showed that the over-expression of ZEB2 could inhibit TRAIL-induced apoptosis via inhibiting nuclear factor-κB (NF-κB) signaling pathway in LX-2 cells. Collectively, our data suggest that up-regulation of miR-145 can down-regulate ZEB2 expression, consequently promoting TRAIL-induced apoptosis in LX-2 cells through NF-κB signaling pathway, which facilitates the resolution of liver fibrosis.
Collapse
Affiliation(s)
- Junfa Yang
- School of Pharmacy, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, China
| | - Qingxue Liu
- School of Pharmacy, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, China
| | - Shiyang Cao
- School of Pharmacy, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, China
| | - Tao Xu
- School of Pharmacy, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, China
| | - Xiaofeng Li
- School of Pharmacy, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, China
| | - Dandan Zhou
- School of Pharmacy, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, China
| | - Linxin Pan
- School of Life Sciences, Anhui Medical University, Hefei, China
| | - Changyao Li
- School of Pharmacy, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, China
| | - Cheng Huang
- School of Pharmacy, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, China
| | - Xiaoming Meng
- School of Pharmacy, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, China
| | - Lei Zhang
- School of Pharmacy, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, China
| | - Xiao Wang
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
41
|
Hepatic stellate cells as key target in liver fibrosis. Adv Drug Deliv Rev 2017; 121:27-42. [PMID: 28506744 DOI: 10.1016/j.addr.2017.05.007] [Citation(s) in RCA: 1026] [Impact Index Per Article: 128.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 03/21/2017] [Accepted: 05/09/2017] [Indexed: 02/06/2023]
Abstract
Progressive liver fibrosis, induced by chronic viral and metabolic disorders, leads to more than one million deaths annually via development of cirrhosis, although no antifibrotic therapy has been approved to date. Transdifferentiation (or "activation") of hepatic stellate cells is the major cellular source of matrix protein-secreting myofibroblasts, the major driver of liver fibrogenesis. Paracrine signals from injured epithelial cells, fibrotic tissue microenvironment, immune and systemic metabolic dysregulation, enteric dysbiosis, and hepatitis viral products can directly or indirectly induce stellate cell activation. Dysregulated intracellular signaling, epigenetic changes, and cellular stress response represent candidate targets to deactivate stellate cells by inducing reversion to inactivated state, cellular senescence, apoptosis, and/or clearance by immune cells. Cell type- and target-specific pharmacological intervention to therapeutically induce the deactivation will enable more effective and less toxic precision antifibrotic therapies.
Collapse
|
42
|
Du Y, Li J, Xu T, Zhou DD, Zhang L, Wang X. MicroRNA-145 induces apoptosis of glioma cells by targeting BNIP3 and Notch signaling. Oncotarget 2017; 8:61510-61527. [PMID: 28977881 PMCID: PMC5617441 DOI: 10.18632/oncotarget.18604] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 05/22/2017] [Indexed: 01/21/2023] Open
Abstract
MicroRNAs (miRNAs) are involved in the pathogenesis of various human cancers. Here we show that miR-145 expression is decreased in human glioma samples, rat glioma tissues, and glioma cell lines, while expression of BNIP3 is increased. Over-expression of miR-145 or suppression of BNIP3 induced glioma cell apoptosis. BNIP3 is localized in the nucleus in glioma cells, and miR-145 inhibits BNIP3 expression by binding to the 3’ untranslated region of its mRNA. Interestingly, miR-145 and BNIP3 regulate glioma cell apoptosis by modulating Notch signaling. These results indicate that miR-145 increases glioma cell apoptosis by inhibiting BNIP3 and Notch signaling, and suggest that miR-145 may serve as a novel therapeutic target for malignant glioma.
Collapse
Affiliation(s)
- Yan Du
- School of Pharmacy, Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei 230032, China.,Institute for Liver Disease of Anhui Medical University, Anhui Medical University, Hefei 230032, China
| | - Juan Li
- Anhui Provincial Hospital, Hefei 230032, China
| | - Tao Xu
- School of Pharmacy, Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei 230032, China.,Institute for Liver Disease of Anhui Medical University, Anhui Medical University, Hefei 230032, China
| | - Dan-Dan Zhou
- School of Pharmacy, Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei 230032, China.,Institute for Liver Disease of Anhui Medical University, Anhui Medical University, Hefei 230032, China
| | - Lei Zhang
- School of Pharmacy, Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei 230032, China.,Institute for Liver Disease of Anhui Medical University, Anhui Medical University, Hefei 230032, China
| | - Xiao Wang
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| |
Collapse
|
43
|
Abstract
Studies have shown that transforming growth factor-β (TGF-β) is one of the most important factors to promote hepatic fibrosis (HF), and the TGF-β/Smad pathway is a major signaling pathway involved in HF. Abnormal expression of microRNAs (miRNAs) has a key role in the development of HF. In recent years, studies suggest that regulating miRNAs may affect the TGF-β/Smad pathway. This paper discusses the TGF-β/Smad pathway and the related miRNAs that are associated with HF.
Collapse
|
44
|
Men R, Wen M, Zhao M, Dan X, Yang Z, Wu W, Wang MH, Liu X, Yang L. MircoRNA-145 promotes activation of hepatic stellate cells via targeting krüppel-like factor 4. Sci Rep 2017; 7:40468. [PMID: 28091538 PMCID: PMC5238405 DOI: 10.1038/srep40468] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 12/06/2016] [Indexed: 02/05/2023] Open
Abstract
Krüppel-like Factor 4 (KLF4), a target gene of miR-145, can negatively regulate lung fibrosis. However, the potential role of KLF4 and miR-145 in hepatic stellate cells (HSCs) activation or in hepatic fibrosis keeps unclear. This study aims to characterize miR-145 and KLF4 in activated HSCs and liver cirrhotic, and the underlying molecular basis. miR-145 was significantly up-regulated, while KLF4 was dramatically down-regulated during the activation of rat primary HSCs and TGF-βtreated HSCs. Furthermore, miR-145 mimics induced and inhibition of miR-145 reduced α-SMA and COL-I expression in primary HSCs. Additionally, the mRNA and protein levels of KLF4 in the liver of cirrhotic patients and rats were significantly down-regulated. α-SMA and COL-I were increased after inhibition of KLF4 by specific shRNA in primary HSCs. Forced KLF4 expression led to a reduction of α-SMA and COL-I expression in HSCs. miR-145 promotes HSC activation and liver fibrosis by targeting KLF4.
Collapse
Affiliation(s)
- Ruoting Men
- Division of Gastroenterology & Hepatology, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Biostatistics, JC school of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, China
| | - Maoyao Wen
- Division of Gastroenterology & Hepatology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Mingyue Zhao
- Laboratory of Cardiovascular Diseases, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xuelian Dan
- Division of Gastroenterology & Hepatology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zongze Yang
- Creation and Management of a Tumour Bank, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Wenchao Wu
- Laboratory of Cardiovascular Diseases, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Maggie Haitian Wang
- Department of Biostatistics, JC school of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, China
| | - Xiaojing Liu
- Laboratory of Cardiovascular Diseases, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Li Yang
- Division of Gastroenterology & Hepatology, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
45
|
Ye Y, Li Z, Feng Q, Chen Z, Wu Z, Wang J, Ye X, Zhang D, Liu L, Gao W, Zhang L, Wang B. Downregulation of microRNA-145 may contribute to liver fibrosis in biliary atresia by targeting ADD3. PLoS One 2017; 12:e0180896. [PMID: 28902846 PMCID: PMC5597134 DOI: 10.1371/journal.pone.0180896] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 06/22/2017] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Biliary atresia (BA) is a pediatric liver disease characterized by fibro-obliteration and obstruction of the extrahepatic biliary system, that invariably leads to cirrhosis and even death, if left untreated for extended time. However, its pathology and etiology still remained unknown. In this study, we tested the expression of adducin 3 (ADD3), the gene identified as a susceptibility gene in BA by GWAS, and uncovered its upstream regulatory microRNA in the pathogenesis of BA. METHODS In this study, 14 infants with BA and 14 infants with choledochal cyst (CC) were enrolled as experimental group and control group, respectively. ADD3 and microRNA-145 (miR-145) expression profiles in liver tissues of BA and CC were determined using qPCR. Luciferase reporter assay was performed to verify the direct interaction between miR-145-5p and ADD3 3' Untranslated Regions (3'UTR). The Lentiviral vectors containing miR-145, miR-145-3p inhibitor, miR-145-5p inhibitor, empty vector were transfected into human hepatic stellate cell line (LX-2) to determine the functional effect of miR-145 on ADD3 expression at both mRNA and protein level. RESULTS MiR-145 was shown to be down-regulated in liver tissues of infants with BA compared to CC (p = 0.0267). ADD3, verified as a target of miR-145-5p, was shown to be overexpressed in infants with BA at the mRNA level (p = 0.0118). Transfection of lentiviruses containing miR-145 into LX-2 cells decreased the expression of ADD3 at both mRNA and protein level compared to negative control group, and suppressed the expression of p-Akt at protein level. CONCLUSIONS Our study has shown that overexpressed ADD3 and downregulated miR-145 were detected in BA liver tissues. MiR-145-5p was confirmed to target ADD3 by luciferase reporter assay. The downregulation of miR-145 may contribute to liver fibrosis in BA by upregulating the expression of ADD3.
Collapse
Affiliation(s)
- Yongqin Ye
- Shantou University Medical College, Shantou, Guangdong, China
- Department of General Surgery, Shenzhen Children’s Hospital, Shenzhen, Guangdong, China
| | - Zhihan Li
- Shantou University Medical College, Shantou, Guangdong, China
- Department of General Surgery, Shenzhen Children’s Hospital, Shenzhen, Guangdong, China
| | - Qi Feng
- Department of General Surgery, Shenzhen Children’s Hospital, Shenzhen, Guangdong, China
| | - Zimin Chen
- Department of General Surgery, Shenzhen Children’s Hospital, Shenzhen, Guangdong, China
| | - Zhouguang Wu
- Department of General Surgery, Shenzhen Children’s Hospital, Shenzhen, Guangdong, China
| | - Jianyao Wang
- Department of General Surgery, Shenzhen Children’s Hospital, Shenzhen, Guangdong, China
| | - Xiaoshuo Ye
- Department of General Surgery, Shenzhen Children’s Hospital, Shenzhen, Guangdong, China
| | - Dahao Zhang
- Shantou University Medical College, Shantou, Guangdong, China
- Department of General Surgery, Shenzhen Children’s Hospital, Shenzhen, Guangdong, China
| | - Lei Liu
- Shenzhen Key Laboratory of Hepatobiliary Disease, Shenzhen Third People’s Hospital, Shenzhen, Guangdong, China
| | - Wei Gao
- Department of Organ Transplatation, Tianjin First Center Hospital, Tianjin, China
- * E-mail: (BW); (WG); (LZ)
| | - Lihui Zhang
- Department of Traditional Chinese Medicine, Shenzhen Children’s Hospital, Shenzhen, Guangdong, China
- * E-mail: (BW); (WG); (LZ)
| | - Bin Wang
- Department of General Surgery, Shenzhen Children’s Hospital, Shenzhen, Guangdong, China
- * E-mail: (BW); (WG); (LZ)
| |
Collapse
|
46
|
microRNA-145 modulates epithelial-mesenchymal transition and suppresses proliferation, migration and invasion by targeting SIP1 in human cervical cancer cells. Cell Oncol (Dordr) 2016; 40:119-131. [PMID: 27933466 DOI: 10.1007/s13402-016-0307-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/22/2016] [Indexed: 10/20/2022] Open
Abstract
PURPOSE Previously, it has been reported that microRNA-145 (miR-145) is lowly expressed in human cervical cancers and that its putative tumour suppressive role may be attributed to epithelial-mesenchymal transition (EMT) regulation. Here, we aimed to assess whether miR-145 may affect EMT-associated markers/genes and suppress cervical cancer growth and motility, and to provide a mechanistic basis for these phenomena. METHODS The identification of the SMAD-interacting protein 1 (SIP1) mRNA as putative miR-145 target was investigated using a 3' untranslated region (3'UTR) luciferase assay and Western blotting, respectively. The functional effects of exogenous miR-145 expression, miR-145 suppression or siRNA-mediated SIP1 expression down-regulation in cervical cancer-derived C33A and SiHa cells were analysed using Western blotting, BrdU incorporation (proliferation), transwell migration and invasion assays. In addition, the expression levels of miR-145 and SIP1 were determined in primary human cervical cancer and non-cancer tissue samples using qRT-PCR. RESULTS We found that miR-145 binds to the wild-type 3'UTR of SIP1, but not to its mutant counterpart, and that, through this binding, miR-145 can effectively down-regulate SIP1 expression. In addition, we found that exogenous miR-145 expression or siRNA-mediated down-regulation of SIP1 expression attenuates the proliferation, migration and invasion of C33A and SiHa cells and alters the expression of the EMT-associated markers CDH1, VIM and SNAI1, whereas inhibition of endogenous miR-145 expression elicited the opposite effects. The expression of miR-145 in cervical cancer tissue samples was found to be low, while that of SIP1 was found to be high compared to non-cancerous cervical tissues. An inverse expression correlation between the two was substantiated through the anlaysis of data deposited in the TCGA database. CONCLUSION Our data indicate that low miR-145 expression levels in conjunction with elevated SIP1 expression levels may contribute to cervical cancer development. MiR-145-mediated regulation of SIP1 provides a novel mechanistic basis for its tumour suppressive mode of action in human cervical cancer cells.
Collapse
|