1
|
Zeng YL, Gao F, Zhang C, Ren PP, Ma L, Wang X, Wang R, Kang Y, Li K. USF1 modulates transcription and cellular functions by regulating multiple transcription factors in Huh7 cells. Oncol Lett 2023; 26:532. [PMID: 38020298 PMCID: PMC10655063 DOI: 10.3892/ol.2023.14119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 09/28/2023] [Indexed: 12/01/2023] Open
Abstract
Liver cancer, including hepatocellular carcinoma (HCC), is a malignant tumor that has high rates of metastasis and mortality worldwide. Upstream transcription factor 1 (USF1) is a canonical transcription factor (TF) and is associated with the pathogenesis of several cancers, but its biological functions and molecular targets in HCC remain unclear. Huh7 cells that overexpress USF1 were used with whole transcriptome profiling through RNA sequencing and chromatin immunoprecipitation (ChIP) sequencing methods to investigate the downstream targets of USF1. Reverse transcription-quantitative PCR was then used to validate the downstream targets. The results showed that USF1 significantly regulates 350 differentially expressed genes (DEGs). The upregulated DEGs were primarily protein-coding genes enriched in immune and inflammation response pathways, while the downregulated DEGs were mainly coding long non-coding (lnc)RNAs, indicating the regulatory function of USF1. It was also demonstrated that USF1 directly binds to the promoter region of 2,492 genes, which may be involved in the viral progression and cell proliferation pathways. By integrating these two datasets, 16 overlapped genes were detected, including downregulated lncRNA-NEAT1 and upregulated TF-ETV5. The downregulated lncRNA-NEAT1 showed reverse expression pattern and prognosis result compared with that of USF1 in patients with liver cancer, while upregulated TF-ETV5 showed consistent results with USF1. Promoter region motif analysis indicated that ETV5 has more binding motifs and genes than USF1 itself for USF1-regulated DEGs, indicating that USF1 may indirectly modulate gene expression by regulating ETV5 expression in Huh7 cells. The study also validated the direct interaction between USF1 and the promoter of ETV5 using ChIP-qPCR. In summary, the results demonstrated that USF1 binds to the promoter region of thousands of genes and affects a large part of DEGs indirectly. Downstream genes, including lncRNA-NEAT1 and TF-ETV5, may also have potential functions in the regulated network by USF1 and have potential functions in the progression of HCC. The present findings suggested that USF1 and its downstream targets could be potential targets for HCC therapy in the future.
Collapse
Affiliation(s)
- Yan-Li Zeng
- Department of Infectious Diseases, Henan Key Laboratory for Infectious Diseases, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
- Department of Infectious Diseases, Zhengzhou University People's Hospital, Zhengzhou, Henan 450003, P.R. China
- Department of Infectious Diseases, Henan University People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Fei Gao
- Department of Infectious Diseases, Henan Key Laboratory for Infectious Diseases, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Can Zhang
- Department of Infectious Diseases, Henan Key Laboratory for Infectious Diseases, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Pei-Pei Ren
- Department of Infectious Diseases, Henan Key Laboratory for Infectious Diseases, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Li Ma
- Department of Infectious Diseases, Henan Key Laboratory for Infectious Diseases, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Xin Wang
- Department of Infectious Diseases, Henan University, Zhengzhou, Henan 450003, P.R. China
| | - Ruzhen Wang
- Department of Infectious Diseases, Henan Key Laboratory for Infectious Diseases, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Yi Kang
- Department of Infectious Diseases, Henan Key Laboratory for Infectious Diseases, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Ke Li
- Department of Infectious Diseases, Henan Key Laboratory for Infectious Diseases, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
| |
Collapse
|
2
|
Silencing RPL8 inhibits the progression of hepatocellular carcinoma by down-regulating the mTORC1 signalling pathway. Hum Cell 2023; 36:725-737. [PMID: 36577883 DOI: 10.1007/s13577-022-00852-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 12/20/2022] [Indexed: 12/29/2022]
Abstract
This study aimed to explore the role of ribosomal protein L8 (RPL8) in controlling hepatocellular carcinoma (LIHC) development. We measured RPL8 expression, apoptosis, cell viability, proliferation, migration, invasion, glucose uptake, lactate production, and the ATP/ADP ratio of LIHC cells to investigate the effect of RPL8 on LIHC. Bioinformatic analysis was employed to analyse RPL8 expression and its potential mechanism in LIHC. RPL8 was upregulated in LIHC tissues and cells. RPL8 silencing accelerated apoptosis and suppressed viability, growth, and movement of LIHC cells. Additionally, RPL8 silencing inhibited glycolysis in LIHC cells. Bioinformatic analysis revealed that RPL8 is regulated by the upstream transcription factor upstream stimulating factor 1 (USF1) and activates the mTORC1 signalling pathway. USF1 overexpression eliminated the inhibitory effect of RPL8 silencing in LIHC cells. RPL8 overexpression increased cell growth, movement, and glycolysis in LIHC. However, inhibition of the mTORC1 signalling pathway eliminated the effect of RPL8 overexpression on LIHC cells. In conclusion, RPL8 may affect LIHC progression by regulating the mTORC1 signalling pathway.
Collapse
|
3
|
Lebsir N, Zoulim F, Grigorov B. Heparanase-1: From Cancer Biology to a Future Antiviral Target. Viruses 2023; 15:237. [PMID: 36680276 PMCID: PMC9860851 DOI: 10.3390/v15010237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 01/19/2023] Open
Abstract
Heparan sulfate proteoglycans (HSPGs) are a major constituent of the extracellular matrix (ECM) and are found to be implicated in viral infections, where they play a role in both cell entry and release for many viruses. The enzyme heparanase-1 is the only known endo-beta-D-glucuronidase capable of degrading heparan sulphate (HS) chains of HSPGs and is thus important for regulating ECM homeostasis. Heparanase-1 expression is tightly regulated as the uncontrolled cleavage of HS may result in abnormal cell activation and significant tissue damage. The overexpression of heparanase-1 correlates with pathological scenarios and is observed in different human malignancies, such as lymphoma, breast, colon, lung, and hepatocellular carcinomas. Interestingly, heparanase-1 has also been documented to be involved in numerous viral infections, e.g., HSV-1, HPV, DENV. Moreover, very recent reports have demonstrated a role of heparanase-1 in HCV and SARS-CoV-2 infections. Due to the undenied pro-carcinogenic role of heparanase-1, multiple inhibitors have been developed, some reaching phase II and III in clinical studies. However, the use of heparanase inhibitors as antivirals has not yet been proposed. If it can be assumed that heparanase-1 is implicated in numerous viral life cycles, its inhibition by specific heparanase-acting compounds should result in a blockage of viral infection. This review addresses the perspectives of using heparanase inhibitors, not only for cancer treatment, but also as antivirals. Eventually, the development of a novel class antivirals targeting a cellular protein could help to alleviate the resistance problems seen with some current antiretroviral therapies.
Collapse
Affiliation(s)
- Nadjet Lebsir
- Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, 69434 Lyon, France
- Confluence: Sciences et Humanités (EA 1598), UCLy, 10 Place des Archives, 69002 Lyon, France
| | - Fabien Zoulim
- Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, 69434 Lyon, France
- Hospices Civils de Lyon, 69002 Lyon, France
| | - Boyan Grigorov
- Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, 69434 Lyon, France
| |
Collapse
|
4
|
USF2-mediated upregulation of TXNRD1 contributes to hepatocellular carcinoma progression by activating Akt/mTOR signaling. Cell Death Dis 2022; 13:917. [PMID: 36319631 PMCID: PMC9626593 DOI: 10.1038/s41419-022-05363-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 10/20/2022] [Indexed: 11/05/2022]
Abstract
Thioredoxin reductase 1 (TXNRD1) is one of the major redox regulators in mammalian cells, which has been reported to be involved in tumorigenesis. However, its roles and regulatory mechanism underlying the progression of HCC remains poorly understood. In this study, we demonstrated that TXNRD1 was significantly upregulated in HCC tumor tissues and correlated with poor survival in HCC patients. Functional studies indicated TXNRD1 knockdown substantially suppressed HCC cell proliferation and metastasis both in vitro and in vivo, and its overexpression showed opposite effects. Mechanistically, TXNRD1 attenuated the interaction between Trx1 and PTEN which resulting in acceleration of PTEN degradation, thereby activated Akt/mTOR signaling and its target genes which conferred to elevated HCC cell mobility and metastasis. Moreover, USF2 was identified as a transcriptional suppressor of TXNRD1, which directly interacted with two E-box sites in TXNRD1 promoter. USF2 functioned as tumor suppressor through the downstream repression of TXNRD1. Further clinical data revealed negative co-expression correlations between USF2 and TXNRD1. In conclusion, our findings reveal that USF2-mediated upregulation of TXNRD1 contributes to hepatocellular carcinoma progression by activating Akt/mTOR signaling.
Collapse
|
5
|
Heparanase: A Novel Therapeutic Target for the Treatment of Atherosclerosis. Cells 2022; 11:cells11203198. [PMID: 36291066 PMCID: PMC9599978 DOI: 10.3390/cells11203198] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/07/2022] [Accepted: 10/07/2022] [Indexed: 11/16/2022] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of death and disability worldwide, and its management places a huge burden on healthcare systems through hospitalisation and treatment. Atherosclerosis is a chronic inflammatory disease of the arterial wall resulting in the formation of lipid-rich, fibrotic plaques under the subendothelium and is a key contributor to the development of CVD. As such, a detailed understanding of the mechanisms involved in the development of atherosclerosis is urgently required for more effective disease treatment and prevention strategies. Heparanase is the only mammalian enzyme known to cleave heparan sulfate of heparan sulfate proteoglycans, which is a key component of the extracellular matrix and basement membrane. By cleaving heparan sulfate, heparanase contributes to the regulation of numerous physiological and pathological processes such as wound healing, inflammation, tumour angiogenesis, and cell migration. Recent evidence suggests a multifactorial role for heparanase in atherosclerosis by promoting underlying inflammatory processes giving rise to plaque formation, as well as regulating lesion stability. This review provides an up-to-date overview of the role of heparanase in physiological and pathological processes with a focus on the emerging role of the enzyme in atherosclerosis.
Collapse
|
6
|
Zhao Y, Dong Y, Hong W, Jiang C, Yao K, Cheng C. Computational modeling of chromatin accessibility identified important epigenomic regulators. BMC Genomics 2022; 23:19. [PMID: 34996354 PMCID: PMC8742372 DOI: 10.1186/s12864-021-08234-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 12/03/2021] [Indexed: 11/28/2022] Open
Abstract
Chromatin accessibility is essential for transcriptional activation of genomic regions. It is well established that transcription factors (TFs) and histone modifications (HMs) play critical roles in chromatin accessibility regulation. However, there is a lack of studies that quantify these relationships. Here we constructed a two-layer model to predict chromatin accessibility by integrating DNA sequence, TF binding, and HM signals. By applying the model to two human cell lines (GM12878 and HepG2), we found that DNA sequences had limited power for accessibility prediction, while both TF binding and HM signals predicted chromatin accessibility with high accuracy. According to the HM model, HM features determined chromatin accessibility in a cell line shared manner, with the prediction power attributing to five core HM types. Results from the TF model indicated that chromatin accessibility was determined by a subset of informative TFs including both cell line-specific and generic TFs. The combined model of both TF and HM signals did not further improve the prediction accuracy, indicating that they provide redundant information in terms of chromatin accessibility prediction. The TFs and HM models can also distinguish the chromatin accessibility of proximal versus distal transcription start sites with high accuracy.
Collapse
Affiliation(s)
- Yanding Zhao
- Department of Medicine, Baylor College of Medicine, Room ICTR 100D, One Baylor Plaza, Baylor College of Medicine, Houston, TX, 77030, USA
- The Institute for Clinical and Translational Research, Baylor College of Medicine, Room ICTR 100D, One Baylor Plaza, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yadong Dong
- Department of Medicine, Baylor College of Medicine, Room ICTR 100D, One Baylor Plaza, Baylor College of Medicine, Houston, TX, 77030, USA
- The Institute for Clinical and Translational Research, Baylor College of Medicine, Room ICTR 100D, One Baylor Plaza, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Wei Hong
- Department of Medicine, Baylor College of Medicine, Room ICTR 100D, One Baylor Plaza, Baylor College of Medicine, Houston, TX, 77030, USA
- The Institute for Clinical and Translational Research, Baylor College of Medicine, Room ICTR 100D, One Baylor Plaza, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Chongming Jiang
- Department of Medicine, Baylor College of Medicine, Room ICTR 100D, One Baylor Plaza, Baylor College of Medicine, Houston, TX, 77030, USA
- The Institute for Clinical and Translational Research, Baylor College of Medicine, Room ICTR 100D, One Baylor Plaza, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Kevin Yao
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, TX, 77843, USA
| | - Chao Cheng
- Department of Medicine, Baylor College of Medicine, Room ICTR 100D, One Baylor Plaza, Baylor College of Medicine, Houston, TX, 77030, USA.
- The Institute for Clinical and Translational Research, Baylor College of Medicine, Room ICTR 100D, One Baylor Plaza, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
7
|
Dong Y, Tian J, Yan B, Lv K, Li J, Fu D. Liver-Metastasis-Related Genes are Potential Biomarkers for Predicting the Clinical Outcomes of Patients with Pancreatic Adenocarcinoma. Pathol Oncol Res 2021; 27:1609822. [PMID: 34290570 PMCID: PMC8286999 DOI: 10.3389/pore.2021.1609822] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 06/21/2021] [Indexed: 12/26/2022]
Abstract
It is widely acknowledged that metastasis determines the prognosis of pancreatic adenocarcinoma (PAAD), and the liver is the most primary distant metastatic location of PAAD. It is worth exploring the value of liver-metastasis-related genetic prognostic signature (LM-PS) in predicting the clinical outcomes of PAAD patients post R0 resection. We collected 65 tumors and 165 normal pancreatic data from The Cancer Genome Atlas (TCGA) and the Genotype-Tissue Expression project (GTEx), respectively. Differentially expressed genes (DEGs) between primary tumor and normal pancreatic samples were intersected with DEGs between primary tumor samples with liver metastasis and those without new tumor events. The intersected 45 genes were input into univariate Cox regression analysis to identify the prognostic genes. Thirty-three prognostic liver-metastasis-related genes were identified and included in least absolute shrinkage and selection operator (LASSO) analysis to develop a seven-gene LM-PS, which included six risk genes (ANO1, FAM83A, GPR87, ITGB6, KLK10, and SERPINE1) and one protective gene (SMIM32). The PAAD patients were grouped into low- and high-risk groups based on the median value of risk scores. The LM-PS harbored an independent predictive ability to distinguish patients with a high-risk of death and liver metastasis after R0 resection. Moreover, a robust prognostic nomogram based on LM-PS, the number of positive lymph nodes, and histologic grade were established to predict the overall survival of PAAD patients. Besides, a transcription factor‐microRNA coregulatory network was constructed for the seven LM-PS genes, and the immune infiltration and genomic alterations were systematically explored in the TGCA-PAAD cohort.
Collapse
Affiliation(s)
- Yinlei Dong
- Department of Pancreatic Surgery, Pancreatic Disease Institute, Huashan Hospital, Fudan University, Shanghai, China
| | - Junjie Tian
- Department of Urology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Bingqian Yan
- Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Kun Lv
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Ji Li
- Department of Pancreatic Surgery, Pancreatic Disease Institute, Huashan Hospital, Fudan University, Shanghai, China
| | - Deliang Fu
- Department of Pancreatic Surgery, Pancreatic Disease Institute, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
8
|
Yang C, Zhang S, Chang X, Huang Y, Cui D, Liu Z. MicroRNA-219a-2-3p modulates the proliferation of thyroid cancer cells via the HPSE/cyclin D1 pathway. Exp Ther Med 2021; 21:659. [PMID: 33968189 DOI: 10.3892/etm.2021.10091] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 03/25/2021] [Indexed: 12/18/2022] Open
Abstract
Heparanase (HPSE) is an endo-β-D-glucuronidase overexpressed in different types of human cancer, and a predicted target of microRNA (miRNA/miR)-219a-2-3p in thyroid cancer. The present study aimed to investigate the potential role of HPSE and miR-219a-2-3p in thyroid cancer, and the molecular mechanism of miR-219a-2-3p regulating the proliferation of thyroid cancer cells via HPSE was confirmed. Immunohistochemistry analysis was performed to detect HPSE expression in thyroid cancer sections. In addition, reverse transcription-quantitative PCR analysis was performed to detect mRNA and miR-219a-2-3p expression levels in thyroid cancer samples and cell lines. miR-219-2-3p mimic or HPSE plasmid were transfected into B-CPAP and TPC-1 thyroid cancer cells. Furthermore, western blot analysis was performed to detect the protein expression levels of HPSE and cyclin D1. Cell cycle analysis was performed using propidium iodide staining and flow cytometry, and EdU incorporation was performed to detect cell proliferation. The results demonstrated that high HPSE expression was significantly associated with tumor size, extracapsular invasion and lymph node metastasis. Notably, a statistically negative correlation was observed between HPSE mRNA expression and miR-219a-2-3p expression in thyroid cancer tumors, as well as in thyroid cancer cell lines. When exogenously expressed in B-CPAP and TPC-1 cells, miR-219a-2-3p induced cell cycle arrest at the G0/G1 phase and decreased the percentage of proliferating cells. Furthermore, HPSE and cyclin D1 protein expression decreased following transfection with miR-219a-2-3p. Notably, when HPSE was ectopically expressed in miR-219a-2-3p transfected cells, cyclin D1 expression and the number of proliferative cells increased. Taken together, these results suggest that HPSE contributes to the proliferation of thyroid cancer cells. In addition, miR-219a-2-3p was confirmed to target HPSE and inhibit cell proliferation, which was associated with cyclin D1 suppression-mediated cell cycle arrest.
Collapse
Affiliation(s)
- Chuanjia Yang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Siyang Zhang
- Science and Experiment Center, China Medical University, Shenyang, Liaoning 110122, P.R. China
| | - Xiaoying Chang
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Yonglian Huang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Dongxu Cui
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Zhen Liu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| |
Collapse
|
9
|
Chen X, Cheng B, Dai D, Wu Y, Feng Z, Tong C, Wang X, Zhao J. Heparanase induces necroptosis of microvascular endothelial cells to promote the metastasis of hepatocellular carcinoma. Cell Death Discov 2021; 7:33. [PMID: 33597510 PMCID: PMC7889896 DOI: 10.1038/s41420-021-00411-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 09/17/2020] [Accepted: 10/05/2020] [Indexed: 12/20/2022] Open
Abstract
Heparanase (HPSE) is a kind of multifunctional extracellular hydrolase, and related to metastasis of hepatocellular carcinoma (HCC). Endothelial necroptosis promotes the metastasis of cancer cells. It is not clear whether HPSE could mediate necroptosis of microvascular endothelial cells (MVECs) to promote HCC metastasis. Here we found HPSE expression was up-regulated in HCC tissues and its over-expression was correlated with multiple tumor foci, microvascular invasion, and poor outcome of HCC patients. Non-contact co-culture experiments showed high-expressed HPSE in HCC cells mediated the necroptosis of human umbilical vein endothelial cells (HUVECs) and elevated the expression levels of syndecan-1 (SDC-1) and tumor necrosis factor-α (TNF-α) in vitro. As a result of necroptosis, trans-endothelial migration (TEM) of HCC cells was increased. Conversely, both HPSE and SDC-1 knockdowns reversed necroptosis and decreased TNF-α expression level, while HPSE over-expression increased SDC-1 and TNF-α expression and aggravated necroptosis. Animal experiments found that the nude mice, intraperitoneally injected with HPSE high expressing HCC cells, had obvious necroptosis of MVECs and high intrahepatic metastasis rate, which could be relieved by inhibitor of necroptosis. Morever, HPSE elevated the expression levels of p38 mitogen-activated protein kinase (p38 MAPK) rather than nuclear factor kappa B in vitro. Our data suggest that HPSE induces necroptosis of MVECs to promote the metastasis of HCC by activating HPSE/SDC-1/TNF-α axis and p38 MAPK pathway.
Collapse
Affiliation(s)
- Xiaopeng Chen
- First Department of Hepatobiliary Surgery, Affiliated Yijishan Hospital of Wannan Medical College, 241001, Wuhu, China.
| | - Bin Cheng
- First Department of Hepatobiliary Surgery, Affiliated Yijishan Hospital of Wannan Medical College, 241001, Wuhu, China.,Department of Hepatobiliary surgery, Huangshan People's Hospital, 245000, Huangshan, China
| | - Dafei Dai
- First Department of Hepatobiliary Surgery, Affiliated Yijishan Hospital of Wannan Medical College, 241001, Wuhu, China
| | - Yuhai Wu
- First Department of Hepatobiliary Surgery, Affiliated Yijishan Hospital of Wannan Medical College, 241001, Wuhu, China
| | - Zhiwen Feng
- First Department of Hepatobiliary Surgery, Affiliated Yijishan Hospital of Wannan Medical College, 241001, Wuhu, China
| | - Chaogang Tong
- Department of General Surgery, Affiliated Chaohu Hospital, Anhui Medical University, 238000, Hefei, China
| | - Xiangming Wang
- Department of Pathology, Affiliated Yijishan Hospital of Wannan Medical College, 241001, Wuhu, China
| | - Jun Zhao
- Department of Gastrointestinal Surgery, Affiliated Yijishan Hospital of Wannan Medical College, 241001, Wuhu, China
| |
Collapse
|
10
|
Alshehri MA, Alshehri MM, Albalawi NN, Al-Ghamdi MA, Al-Gayyar MMH. Heparan sulfate proteoglycans and their modification as promising anticancer targets in hepatocellular carcinoma. Oncol Lett 2021; 21:173. [PMID: 33552290 PMCID: PMC7798035 DOI: 10.3892/ol.2021.12434] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 12/08/2020] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common types of primary liver cancer. Despite advancements in the treatment strategies of HCC, there is an urgent requirement to identify and develop novel therapeutic drugs that do not lead to resistance. These novel agents should have the potential to influence the primary mechanisms participating in the pathogenesis of HCC. Heparan sulfate proteoglycans (HSPGs) are major elements of the extracellular matrix that perform structural and signaling functions. HSPGs protect against invasion of tumor cells by preventing cell infiltration and intercellular adhesion. Several enzymes, such as heparanase, matrix metalloproteinase-9 and sulfatase-2, have been reported to affect HSPGs, leading to their degradation and thus enhancing tumor invasion. In addition, some compounds that are produced from the degradation of HSPGs, including glypican-3 and syndecan-1, enhance tumor progression. Thus, the identification of enzymes that affect HSPGs or their degradation products in HCC may lead to the development of novel therapeutic targets. The present review discusses the main enzymes and compounds associated with HSPGs, and their involvement with the pathogenicity of HCC.
Collapse
Affiliation(s)
- Mohammed A Alshehri
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Moath M Alshehri
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Naif N Albalawi
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Moshari A Al-Ghamdi
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Mohammed M H Al-Gayyar
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia.,Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt.,Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia
| |
Collapse
|
11
|
Tatsumi Y, Miyake M, Shimada K, Fujii T, Hori S, Morizawa Y, Nakai Y, Anai S, Tanaka N, Konishi N, Fujimoto K. Inhibition of Heparanase Expression Results in Suppression of Invasion, Migration and Adhesion Abilities of Bladder Cancer Cells. Int J Mol Sci 2020; 21:ijms21113789. [PMID: 32471161 PMCID: PMC7313018 DOI: 10.3390/ijms21113789] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/22/2020] [Accepted: 05/26/2020] [Indexed: 01/15/2023] Open
Abstract
Heparan sulfate proteoglycan syndecan-1, CD138, is known to be associated with cell proliferation, adhesion, and migration in malignancies. We previously reported that syndecan-1 (CD138) may contribute to urothelial carcinoma cell survival and progression. We investigated the role of heparanase, an enzyme activated by syndecan-1 in human urothelial carcinoma. Using human urothelial cancer cell lines, MGH-U3 and T24, heparanase expression was reduced with siRNA and RK-682, a heparanase inhibitor, to examine changes in cell proliferation activity, induction of apoptosis, invasion ability of cells, and its relationship to autophagy. A bladder cancer development mouse model was treated with RK-682 and the bladder tissues were examined using immunohistochemical analysis for Ki-67, E-cadherin, LC3, and CD31 expressions. Heparanase inhibition suppressed cellular growth by approximately 40% and induced apoptosis. The heparanase inhibitor decreased cell activity in a concentration-dependent manner and suppressed invasion ability by 40%. Inhibition of heparanase was found to suppress autophagy. In N-butyl-N-(4-hydroxybutyl) nitrosamine (BBN)-induced bladder cancer mice, treatment with heparanase inhibitor suppressed the progression of cancer by 40%, compared to controls. Immunohistochemistry analysis showed that heparanase inhibitor suppressed cell growth, and autophagy. In conclusion, heparanase suppresses apoptosis and promotes invasion and autophagy in urothelial cancer.
Collapse
Affiliation(s)
- Yoshihiro Tatsumi
- Department of Urology, Nara Medical University, 840 Shijo-cho, Nara 634-8522, Japan; (Y.T.); (M.M.); (S.H.); (Y.M.); (Y.N.); (S.A.); (N.T.)
- Department of Pathology, Nara Medical University, 840 Shijo-cho, Nara 634-8522, Japan; (K.S.); (T.F.); (N.K.)
| | - Makito Miyake
- Department of Urology, Nara Medical University, 840 Shijo-cho, Nara 634-8522, Japan; (Y.T.); (M.M.); (S.H.); (Y.M.); (Y.N.); (S.A.); (N.T.)
| | - Keiji Shimada
- Department of Pathology, Nara Medical University, 840 Shijo-cho, Nara 634-8522, Japan; (K.S.); (T.F.); (N.K.)
| | - Tomomi Fujii
- Department of Pathology, Nara Medical University, 840 Shijo-cho, Nara 634-8522, Japan; (K.S.); (T.F.); (N.K.)
| | - Shunta Hori
- Department of Urology, Nara Medical University, 840 Shijo-cho, Nara 634-8522, Japan; (Y.T.); (M.M.); (S.H.); (Y.M.); (Y.N.); (S.A.); (N.T.)
| | - Yosuke Morizawa
- Department of Urology, Nara Medical University, 840 Shijo-cho, Nara 634-8522, Japan; (Y.T.); (M.M.); (S.H.); (Y.M.); (Y.N.); (S.A.); (N.T.)
| | - Yasushi Nakai
- Department of Urology, Nara Medical University, 840 Shijo-cho, Nara 634-8522, Japan; (Y.T.); (M.M.); (S.H.); (Y.M.); (Y.N.); (S.A.); (N.T.)
| | - Satoshi Anai
- Department of Urology, Nara Medical University, 840 Shijo-cho, Nara 634-8522, Japan; (Y.T.); (M.M.); (S.H.); (Y.M.); (Y.N.); (S.A.); (N.T.)
| | - Nobumichi Tanaka
- Department of Urology, Nara Medical University, 840 Shijo-cho, Nara 634-8522, Japan; (Y.T.); (M.M.); (S.H.); (Y.M.); (Y.N.); (S.A.); (N.T.)
| | - Noboru Konishi
- Department of Pathology, Nara Medical University, 840 Shijo-cho, Nara 634-8522, Japan; (K.S.); (T.F.); (N.K.)
| | - Kiyohide Fujimoto
- Department of Urology, Nara Medical University, 840 Shijo-cho, Nara 634-8522, Japan; (Y.T.); (M.M.); (S.H.); (Y.M.); (Y.N.); (S.A.); (N.T.)
- Correspondence: ; Tel.: +81-744-22-3051 (ext. 2338)
| |
Collapse
|
12
|
Zhou X, Hu M, Ge Z. Tumor‑suppressive miR‑299‑3p inhibits gastric cancer cell invasion by targeting heparanase. Mol Med Rep 2019; 20:2151-2158. [PMID: 31257534 PMCID: PMC6691259 DOI: 10.3892/mmr.2019.10436] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Accepted: 05/29/2019] [Indexed: 01/10/2023] Open
Abstract
Gastric cancer (GC) remains a leading cause of cancer‑associated mortality globally. Emerging evidence suggests that microRNAs (miRs) function as oncogenes or tumor suppressors, contributing to various aspects of cancer progression, including invasion and metastasis. In the present study, the specific role of miR‑299‑3p in the invasion of GC cells was investigated. The expression level of miR‑299‑3p was measured using reverse transcription‑quantitative PCR and in situ hybridization in human GC tissues. Effects of miR‑299‑3p on GC cell invasion were determined by Transwell assay. Bioinformatics and luciferase reporter assays were performed to identify and verify the downstream effectors of miR‑299‑3p. miR‑299‑3p expression analysis in clinical GC samples revealed a significant downregulation of miR‑299‑3p compared with non‑tumor tissues. Inhibition of miR‑299‑3p promoted the invasive abilities of GC cells, whereas its overexpression significantly suppressed cell invasion. Bioinformatics and luciferase reporter assays identified heparanase (HPSE) as a direct target of miR‑299‑3p, the ectopic expression of which reversed the impairment in cell invasion induced by miR‑299‑3p upregulation. Furthermore, HPSE expression was negatively associated with miR‑299‑3p levels in human GC tissues. Overall, the present study indicated that miR‑299‑3p functions as a tumor suppressor by directly targeting HPSE, highlighting its potential as a target for the treatment of GC.
Collapse
Affiliation(s)
- Xiangjun Zhou
- Department of Gastroenterology, Danyang People's Hospital Affiliated to Nantong University, Danyang, Jiangsu 212300, P.R. China
| | - Mengmou Hu
- Department of Clinical Laboratory, Danyang People's Hospital Affiliated to Nantong University, Danyang, Jiangsu 212300, P.R. China
| | - Zhenghui Ge
- Department of Gastroenterology, Danyang People's Hospital Affiliated to Nantong University, Danyang, Jiangsu 212300, P.R. China
| |
Collapse
|
13
|
Chi TF, Horbach T, Götz C, Kietzmann T, Dimova EY. Cyclin-Dependent Kinase 5 (CDK5)-Mediated Phosphorylation of Upstream Stimulatory Factor 2 (USF2) Contributes to Carcinogenesis. Cancers (Basel) 2019; 11:cancers11040523. [PMID: 31013770 PMCID: PMC6521020 DOI: 10.3390/cancers11040523] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Revised: 03/30/2019] [Accepted: 04/08/2019] [Indexed: 12/12/2022] Open
Abstract
The transcription factor USF2 is supposed to have an important role in tumor development. However, the regulatory mechanisms contributing to the function of USF2 are largely unknown. Cyclin-dependent kinase 5 (CDK5) seems to be of importance since high levels of CDK5 were found in different cancers associated with high USF2 expression. Here, we identified USF2 as a phosphorylation target of CDK5. USF2 is phosphorylated by CDK5 at two serine residues, serine 155 and serine 222. Further, phosphorylation of USF2 at these residues was shown to stabilize the protein and to regulate cellular growth and migration. Altogether, these results delineate the importance of the CDK5-USF2 interplay in cancer cells.
Collapse
Affiliation(s)
- Tabughang Franklin Chi
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90014 Oulu, Finland; (T.F.C.); (T.K.)
| | - Tina Horbach
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90014 Oulu, Finland; (T.F.C.); (T.K.)
| | - Claudia Götz
- Medical Biochemistry and Molecular Biology, Saarland University, 66421 Homburg, Germany;
| | - Thomas Kietzmann
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90014 Oulu, Finland; (T.F.C.); (T.K.)
| | - Elitsa Y. Dimova
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90014 Oulu, Finland; (T.F.C.); (T.K.)
- Correspondence: ; Tel.: +358-0-294-485-785; Fax: +358-8-553-114
| |
Collapse
|
14
|
Kim KC, Yun J, Son DJ, Kim JY, Jung JK, Choi JS, Kim YR, Song JK, Kim SY, Kang SK, Shin DH, Roh YS, Han SB, Hong JT. Suppression of metastasis through inhibition of chitinase 3-like 1 expression by miR-125a-3p-mediated up-regulation of USF1. Am J Cancer Res 2018; 8:4409-4428. [PMID: 30214629 PMCID: PMC6134921 DOI: 10.7150/thno.26467] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 07/15/2018] [Indexed: 12/30/2022] Open
Abstract
Rationale: Chitinase 3-like 1 (Chi3L1) protein is up-regulated in various diseases including solid cancers. According to Genome-Wide Association Study (GWAS)/Online Mendelian Inheritance in Man (OMIM)/Differentially Expressed Gene (DEG) analyses, Chi3L1 is associated with 38 cancers, and more highly associated with cancer compared to other oncogenes such as EGFR, TNFα, etc. However, the mechanisms and pathways by which Chi3L1 is associated with cancer are not clear. In current study, we investigated the role of Chi3L1 in lung metastasis. Methods: We performed the differentially expressed gene analysis to explore the genes which are associated with Chi3L1 using the web-based platform from Biomart. We investigated the metastases in lung tissues of C57BL/6 mice injected with B16F10 melanoma following treatment with Ad-shChi3L1. We also investigated the expression of USF1 and Chi3L1 in Chi3L1 KD mice lung tissues by Western blotting and IHC. We also analyzed lung cancer cells metastases induced by Chi3L1 using migration and cell proliferation assay in human lung cancer cell lines. The involvement of miR-125a-3p in Chi3L1 regulation was determined by miRNA qPCR and luciferase reporter assay. Results: We showed that melanoma metastasis in lung tissues was significantly reduced in Chi3L1 knock-down mice, accompanied by down-regulation of MMP-9, MMP-13, VEGF, and PCNA in Chi3L1 knock-down mice lung tissue, as well as in human lung cancer cell lines. We also found that USF1 was conversely expressed against Chi3L1. USF1 was increased by knock-down of Chi3L1 in mice lung tissues, as well as in human lung cancer cell lines. In addition, knock-down of USF1 increased Chi3L1 levels in addition to augmenting metastasis cell migration and proliferation in mice model, as well as in human cancer cell lines. Moreover, in human lung tumor tissues, the expression of Chi3L1 was increased but USF1 was decreased in a stage-dependent manner. Finally, Chi3L1 expression was strongly regulated by the indirect translational suppressing activity of USF1 through induction of miR-125a-3p, a target of Chi3L1. Conclusion: Metastases in mice lung tissues and human lung cancer cell lines were decreased by KD of Chi3L1. USF1 bound to the Chi3L1 promoter, however, Chi3L1 expression was decreased by USF1, despite USF1 enhancing the transcriptional activity of Chi3L1. We found that USF1 induced miR-125a-3p levels which suppressed Chi3L1 expression. Ultimately, our results suggest that lung metastasis is suppressed by knock-down of Chi3L1 through miR-125a-3p-mediated up-regulation of USF1.
Collapse
|
15
|
Knockdown of Heparanase Suppresses Invasion of Human Trophoblasts by Activating p38 MAPK Signaling Pathway. DISEASE MARKERS 2018; 2018:7413027. [PMID: 29849826 PMCID: PMC5932509 DOI: 10.1155/2018/7413027] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Revised: 03/12/2018] [Accepted: 03/25/2018] [Indexed: 12/11/2022]
Abstract
Preeclampsia is a pregnancy-related disease with increasing maternal and perinatal morbidity and mortality worldwide. Defective trophoblast invasion is considered to be a major factor in the pathophysiological mechanism of preeclampsia. Heparanase, the only endo-β-glucuronidase in mammalian cells, has been shown to be abnormally expressed in the placenta of preeclampsia patients in our previous study. The biological role and potential mechanism of heparanase in trophoblasts remain unclear. In the present study, stably transfected HTR8/SVneo cell lines with heparanase overexpression or knockdown were constructed. The effect of heparanase on cellular proliferation, apoptosis, invasion, tube formation, and potential pathways in trophoblasts was explored. Our results showed that overexpression of heparanase promoted proliferation and invasion. Knockdown of heparanase suppressed proliferation, invasion, and tube formation but induced apoptosis. These findings reveal that downregulation of heparanase may contribute to defective placentation and plays a crucial role in the pathogenesis of preeclampsia. Furthermore, increased activation of p38 MAPK in heparanase-knockdown HTR8/SVneo cell was shown by MAPK pathway phosphorylation array and Western blotting assay. After pretreatment with 3 specific p38 MAPK inhibitors (BMS582949, SB203580, or BIRB796), inadequate invasion in heparanase-knockdown HTR8/SVneo cell was rescued. That indicates that knockdown of heparanase decreases HTR8/SVneo cell invasion through excessive activation of the p38 MAPK signaling pathway. Our study suggests that heparanase can be a potential predictive biomarker for preeclampsia at an early stage of pregnancy and represents a promising therapeutic target for the treatment of preeclampsia.
Collapse
|
16
|
Sheng N, Zhang L, Yang S. MicroRNA-429 decreases the invasion ability of gastric cancer cell line BGC-823 by downregulating the expression of heparanase. Exp Ther Med 2017; 15:1927-1933. [PMID: 29434786 PMCID: PMC5776652 DOI: 10.3892/etm.2017.5608] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 05/15/2017] [Indexed: 12/17/2022] Open
Abstract
The present study aimed to measure the expression of microRNA (miRNA/miR) −429 in gastric cancer and investigate the associated mechanism of action. A total of 30 patients with gastric cancer who received radical or palliative resection at Jining No. 1 People's Hospital between January-October 2016 were included in the present study. Resected gastric cancer and tumor-adjacent tissues were resected. Gastric cancer BGC-823 cells were transfected with miR-429 mimics to induce the overexpression of miR-429, or transfected with small interfering (si)RNA of heparanase (HPSE) for the silencing of HPSE. Reverse transcription-quantitative polymerase chain reaction was used to measure the expression of miR-429 or HPSE mRNA. Western blotting was employed to determine the protein expression of HPSE. Cell-Counting Kit-8 assay was carried out to test cell proliferation and a Transwell assay was used to determine cell invasion ability. Expression of HPSE mRNA and protein in gastric cancer tissues was increased compared with tumor-adjacent tissues. Reduced expression of miR-429 in gastric cancer tissues may be associated with the targeting of HPSE mRNA by miR-429. Overexpression of miR-429 inhibited the transcription and translation of the HPSE gene. However, overexpression of miR-429 did not affect the proliferation of gastric cancer cells. Notably, overexpression of miR-429 reduced the invasion ability of gastric cancer cells. Transfection with HPSE siRNA decreased the expression of the HPSE protein in BGC-823 cells and inhibited the occurrence and development of gastric cancer by reducing the invasion ability of the cells. The present study demonstrated that expression of miR-429 in gastric cancer tissues was significantly reduced compared with tumor-adjacent tissues. As a tumor-suppressor gene, miR-429 decreases the invasion ability of gastric cancer cells by downregulating the expression of HSPE.
Collapse
Affiliation(s)
- Nan Sheng
- Department of Gastroenterology, Jining No. 1 People's Hospital, Jining, Shandong 272011, P.R. China
| | - Lin Zhang
- Department of Oncology, Jining No. 1 People's Hospital, Jining, Shandong 272011, P.R. China
| | - Shaofeng Yang
- Department of Gastroenterology, Jining No. 1 People's Hospital, Jining, Shandong 272011, P.R. China
| |
Collapse
|
17
|
Chen X, Jiang W, Yue C, Zhang W, Tong C, Dai D, Cheng B, Huang C, Lu L. Heparanase Contributes To Trans-Endothelial Migration of Hepatocellular Carcinoma Cells. J Cancer 2017; 8:3309-3317. [PMID: 29158804 PMCID: PMC5665048 DOI: 10.7150/jca.20159] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 08/30/2017] [Indexed: 12/12/2022] Open
Abstract
The overall outcome of patients with hepatocellular carcinoma (HCC) is still very poor due to its high metastasis and recurrence rate. During metastasis, trans-endothelial migration (TEM) of HCC cells is a key step. Heparanase (HPSE) is an endo-beta-glucuronidase and exerts prometastatic properties for normal and tumor-derived cells. However, it is remains unclear that HPSE contributes to TEM of HCC cells. In this study, human umbilical vein endothelial cells-C (HUVEC-C) was used to simulate vascular endothelial cells (VECs), and the HCCLM3 cells with high HPSE expression were chosen and used for in vitro TEM assay and in vivo experiment. As results, we found that HCCLM3 cells showed higher TEM rate compared with other HCC cells. Downregulation or inhibition of HPSE activity resulted in suppression of TEM of HCC cells both in vitro and in vivo. Our findings suggest that HPSE contributes to TEM of HCC cells, which may be a new biological function of HPSE.
Collapse
Affiliation(s)
- Xiaopeng Chen
- Department of Hepatobiliary Surgery, Yijishan Hospital of Wannan Medical College, Wuhu 241001, China
| | - Wen Jiang
- Department of General Surgery, Maanshan People's Hospital, Maanshan 243000, China
| | - Chaofu Yue
- Department of Hepatobiliary Surgery, Yijishan Hospital of Wannan Medical College, Wuhu 241001, China
| | - Wenjun Zhang
- Department of Hepatobiliary Surgery, Yijishan Hospital of Wannan Medical College, Wuhu 241001, China
| | - Chaogang Tong
- Department of General Surgery, Affiliated Chaohu Hospital, Anhui Medical University, Hefei 238000, China
| | - Dafei Dai
- Department of Hepatobiliary Surgery, Yijishan Hospital of Wannan Medical College, Wuhu 241001, China
| | - Bin Cheng
- Department of Hepatobiliary Surgery, Yijishan Hospital of Wannan Medical College, Wuhu 241001, China
| | - Chen Huang
- Department of Hepatobiliary Surgery, Yijishan Hospital of Wannan Medical College, Wuhu 241001, China
| | - Linming Lu
- Department of Pathology, Yijishan Hospital of Wannan Medical College, Wuhu 241001, China
| |
Collapse
|
18
|
Heparanase-1-induced shedding of heparan sulfate from syndecan-1 in hepatocarcinoma cell facilitates lymphatic endothelial cell proliferation via VEGF-C/ERK pathway. Biochem Biophys Res Commun 2017; 485:432-439. [DOI: 10.1016/j.bbrc.2017.02.060] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Accepted: 02/10/2017] [Indexed: 12/11/2022]
|
19
|
Intramyocardial Injection of siRNAs Can Efficiently Establish Myocardial Tissue-Specific Renalase Knockdown Mouse Model. BIOMED RESEARCH INTERNATIONAL 2016; 2016:1267570. [PMID: 27868059 PMCID: PMC5102703 DOI: 10.1155/2016/1267570] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 09/22/2016] [Indexed: 01/03/2023]
Abstract
Ischaemia/reperfusion (I/R) injury will cause additional death of cardiomyocytes in ischaemic heart disease. Recent studies revealed that renalase was involved in the I/R injury. So, the myocardial tissue-specific knockdown mouse models were needed for the investigations of renalase. To establish the mouse models, intramyocardial injection of siRNAs targeting renalase was performed in mice. The wild distribution and high transfection efficiency of the siRNAs were approved. And the renalase expression was efficiently suppressed in myocardial tissue. Compared with the high cost, time consumption, and genetic compensation risk of the Cre/loxP technology, RNA interference (RNAi) technology is much cheaper and less time-consuming. Among the RNAi technologies, injection of siRNAs is safer than virus. And considering the properties of the I/R injury mouse models, the efficiency and durability of injection with siRNAs are acceptable for the studies. Altogether, intramyocardial injection of siRNAs targeting renalase is an economical, safe, and efficient method to establish myocardial tissue-specific renalase knockdown mouse models.
Collapse
|
20
|
Downregulation of Heparanase Expression Results in Suppression of Invasion, Migration, and Adhesion Abilities of Hepatocellular Carcinoma Cells. BIOMED RESEARCH INTERNATIONAL 2015; 2015:241983. [PMID: 26839882 PMCID: PMC4709605 DOI: 10.1155/2015/241983] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 10/31/2015] [Accepted: 11/03/2015] [Indexed: 11/17/2022]
Abstract
Objective. Heparanase (HPSE) is high-expressed in most malignant tumors including hepatocellular carcinoma (HCC) and promotes cancer cell invasion and migration. The aim of the study is to explore whether HPSE enhances adhesion in metastasis of HCC cells. Methods. HPSE expressions in human HCC cells were measured with real-time RT-PCR and Western blot analysis. Four recombinant miRNA vectors pcDNATM6.2-GW/EmGFP-miR-HPSE (pmiR-HPSE) were transfected into HCCLM3 cell. HPSE expression in transfected cell was measured. The cell invasion, migration, and adhesion abilities were detected, respectively. Results. Both HPSE mRNA and protein relative expression levels were higher in HepG2, BEL-7402, and HCCLM3 cells than those in normal hepatocyte (P < 0.05). HPSE showed highest expression level in HCCLM3 cell (P < 0.05). Transfection efficiencies of four miRNA vectors were 75%–85%. The recombinant vectors significantly decreased HPSE expression in transfected HCCLM3 cells (P < 0.01), and pmiR-HPSE-1 showed best interference effect (P < 0.05). pmiR-HPSE-1 significantly decreased the penetrated and migrating cells numbers and adherence rate of HCCLM3 cells (P < 0.05). Conclusion. HPSE is a potentiator of cell adhesion in metastasis of HCC.
Collapse
|
21
|
Qing Q, Zhang S, Chen Y, Li R, Mao H, Chen Q. High glucose-induced intestinal epithelial barrier damage is aggravated by syndecan-1 destruction and heparanase overexpression. J Cell Mol Med 2015; 19:1366-74. [PMID: 25702768 PMCID: PMC4459850 DOI: 10.1111/jcmm.12523] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 11/27/2014] [Indexed: 02/06/2023] Open
Abstract
Syndecan-1 (Sdc1) and its endo-beta-d-glucuronidase heparanase (HPSE) are implicated in maintenance of intestinal epithelial barrier (IEB), but their alterations and roles in high-glucose/hyperglycaemia (HG) conditions have not been fully investigated. This study aimed to determine the expression pattern, the possible regulation mechanism of Sdc1 and HPSE in HG conditions, and their potential effects on IEB. Therefore, diabetic mice/cell models were developed, and tissue/serum samples, cell lysate and culture supernatants were harvested. The expression of Sdc1 and HPSE in control, HG and designated interventions groups were detected. Phosphorylations of mitogen-activated protein kinase signalling pathway (MAPK), the expressions of Occludin and ZO-1, and the levels of transepithelial electrical resistance (TEER) were measured and monitored. The results showed that in HG conditions, intestinal tissue and cellular Sdc1 were significantly decreased, but the expression of HPSE, and soluble Sdc1 in serum and culture supernatants were remarkably increased. Such alterations of Sdc1 and HPSE were associated with solely p38 MAPK activation, and were correlated with the reductions of Occludin, ZO-1 and TEER. Heparin (Sdc1 analogue) and SB203580 (a p38 MAPK inhibitor), instead of insulin, alleviated Sdc1 destruction and HPSE overexpression, and effectively prevented against the reductions of tight junctions and the abnormality of intestinal permeability in HG conditions. In conclusion, we confirm the unique alterations of Sdc1 and HPSE in HG conditions, and found their interactions with p38 MAPK activation and IEB. These indicate that Sdc1/HPSE modulation can be viewed as an important complementary treatment for relieving HG-induced gastrointestinal damage.
Collapse
Affiliation(s)
- Qing Qing
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Department of Gastroenterology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shaoheng Zhang
- Department of Gastroenterology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Ye Chen
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Gastroenterology, Guangzhou, China
| | - Runhua Li
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Gastroenterology, Guangzhou, China
| | - Hua Mao
- Department of Gastroenterology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Qikui Chen
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
22
|
Gao B, Ning SF, Tang YP, Liu HZ, Li JL, Zhang LT. Differential mRNA expression profiles between hepatocellular carcinoma and adjacent normal liver tissue. Shijie Huaren Xiaohua Zazhi 2014; 22:4734-4744. [DOI: 10.11569/wcjd.v22.i31.4734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To identify differentially expressed genes between hepatocellular carcinoma and normal liver tissues and to carry out bioinformatics analysis.
METHODS: Agilent 8×60 K microarray was used to detect the changes of gene expression between hepatocellular carcinoma and adjacent normal liver tissues. Bioinformatics methods were used to identify differentially expressed genes and perform GO pathway analysis. Real-time PCR was applied to verify microarray data.
RESULTS: Microarray analysis screened a total of up-regulated 924 mRNAs and 1770 down-regulated mRNAs in hepatocellular carcinoma tissues compared with the normal tissues. GO pathway analysis demonstrated that these mRNAs are involved in transcription, redox, signal transduction, ion transport, immune response, cell adhesion, and binding functions. The results of real-time PCR were in high concordance with microarray results.
CONCLUSION: Differentially expressed genes identified in this study may be involved in signal transduction, immune response and other key biological processes. These genes may provide new targets for targeted therapy.
Collapse
|