1
|
Sendi H, Yazdimamaghani M, Hu M, Sultanpuram N, Wang J, Moody AS, McCabe E, Zhang J, Graboski A, Li L, Rojas JD, Dayton PA, Huang L, Wang AZ. Nanoparticle delivery of miR-122 inhibits colorectal cancer liver metastasis. Cancer Res 2021; 82:105-113. [PMID: 34753773 DOI: 10.1158/0008-5472.can-21-2269] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/13/2021] [Accepted: 11/05/2021] [Indexed: 11/16/2022]
Abstract
Liver metastasis is a leading cause of cancer morbidity and mortality. Thus, there has been strong interest in the development of therapeutics that can effectively prevent liver metastasis. One potential strategy is to utilize molecules that have broad effects on the liver microenvironment, such as microRNA-122 (miR-122), a liver-specific microRNA (miRNA) that is a key regulator of diverse hepatic functions. Here we report the development of a nanoformulation miR-122 as a therapeutic agent for preventing liver metastasis. We engineered a galactose-targeted lipid calcium phosphate (Gal-LCP) nanoformulation of miR-122. This nanotherapeutic elicited no significant toxicity and delivered miR-122 into hepatocytes with specificity and high efficiency. Across multiple colorectal cancer (CRC) liver metastasis models, treatment with Gal-LCP miR-122 treatment effectively prevented CRC liver metastasis and prolonged survival. Mechanistic studies revealed that delivery of miR-122 was associated with downregulation of key genes in involved in metastatic and cancer inflammation pathways, including several pro-inflammatory factors, matrix metalloproteinases, and other extracellular matrix degradation enzymes. Moreover, Gal-LCP miR-122 treatment was associated with an increased CD8+/CD4+ T-cell ratio and decreased immunosuppressive cell infiltration, which makes the liver more conducive to anti-tumor immune response. Collectively, this work presents a strategy to improve cancer prevention and treatment with nanomedicine-based delivery of miRNA.
Collapse
Affiliation(s)
- Hossein Sendi
- Department of Radiation Oncology, UNC School of Medicine, Chapel Hill, North Carolina
- Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina
- UNC School of Pharmacy, Chapel Hill, North Carolina
| | - Mostafa Yazdimamaghani
- Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina
- UNC School of Pharmacy, Chapel Hill, North Carolina
| | - Mengying Hu
- Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina
- UNC School of Pharmacy, Chapel Hill, North Carolina
| | - Nikhila Sultanpuram
- Department of Radiation Oncology, UNC School of Medicine, Chapel Hill, North Carolina
- Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina
| | - Jie Wang
- Department of Radiation Oncology, UNC School of Medicine, Chapel Hill, North Carolina
- Dalian Municipal Central Hospital, Dalian, China
| | - Amber S Moody
- UNC School of Pharmacy, Chapel Hill, North Carolina
- Joint Department of Biomedical Engineering, University of North Carolina and, North Carolina State University, Chapel Hill, North Carolina
| | - Ellie McCabe
- Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina
- UNC School of Pharmacy, Chapel Hill, North Carolina
| | - Jiajie Zhang
- Department of Radiation Oncology, UNC School of Medicine, Chapel Hill, North Carolina
| | - Amanda Graboski
- Department of Radiation Oncology, UNC School of Medicine, Chapel Hill, North Carolina
| | - Liantao Li
- Department of Radiation Oncology, UNC School of Medicine, Chapel Hill, North Carolina
| | - Juan D Rojas
- Joint Department of Biomedical Engineering, University of North Carolina and, North Carolina State University, Chapel Hill, North Carolina
| | - Paul A Dayton
- UNC School of Pharmacy, Chapel Hill, North Carolina
- Joint Department of Biomedical Engineering, University of North Carolina and, North Carolina State University, Chapel Hill, North Carolina
| | - Leaf Huang
- Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina
- UNC School of Pharmacy, Chapel Hill, North Carolina
| | - Andrew Z Wang
- Department of Radiation Oncology, UNC School of Medicine, Chapel Hill, North Carolina.
- Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina
- UNC School of Pharmacy, Chapel Hill, North Carolina
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
2
|
Hu Y, Du G, Li G, Peng X, Zhang Z, Zhai Y. The miR-122 inhibition alleviates lipid accumulation and inflammation in NAFLD cell model. Arch Physiol Biochem 2021; 127:385-389. [PMID: 31311339 DOI: 10.1080/13813455.2019.1640744] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 06/02/2019] [Accepted: 07/02/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Accumulating evidence showed that the expression of miR-122 was abnormal in NAFLD patients; however, the role of miR-122 on lipid accumulation and inflammation in NAFLD is not clear. METHODS RT-qPCR was applied to detect the expression levels of miR-122 and pro-inflammatory cytokines following transfected with miR-122 inhibitor or treated with oleic acid (OA). Detection of lipid accumulation was performed by triglyceride content test and oil red o staining assay. Western blotting was applied to detect the protein levels of TLR7, TLR4, MyD88 and NF-κBp65. RESULTS We found that the OA promoted lipid accumulation and pro-inflammatory cytokines secretion and activated TLR4/MyD88/NF-κBp65 signalling pathway, which were restored following transfected with miR-122 inhibitor. CONCLUSIONS These results suggested that miR-122 inhibition alleviates lipid accumulation and inflammation in L02 cell induced by OA may through inhibiting TLR4/MyD88/NF-κBp65 signalling pathway. The protective mechanism of miR-122 inhibition in NAFLD must be explored in future studies.
Collapse
Affiliation(s)
- Yiyi Hu
- Department of Gastroenterology, Shunde Hospital of Southern Medical University, Foshan, China
| | - Guoping Du
- Department of Gastroenterology, Shunde Hospital of Southern Medical University, Foshan, China
| | - Guohua Li
- Department of Gastroenterology, Shunde Hospital of Southern Medical University, Foshan, China
| | - Xuetao Peng
- Department of Gastroenterology, Shunde Hospital of Southern Medical University, Foshan, China
| | - Zhiqiao Zhang
- Department of Infectious Diseases, Shunde Hospital of Southern Medical University, Foshan, China
| | - Yingji Zhai
- Department of Gastroenterology, Shunde Hospital of Southern Medical University, Foshan, China
| |
Collapse
|
3
|
Fawzy MS, Toraih EA. MicroRNA signatures as predictive biomarkers in transarterial chemoembolization‐treated hepatocellular carcinoma. PRECISION MEDICAL SCIENCES 2021. [DOI: 10.1002/prm2.12031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Manal S. Fawzy
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine Suez Canal University Ismailia Egypt
- Biochemistry Department, Faculty of Medicine Northern Border University Arar KSA
| | - Eman A. Toraih
- Department of Surgery Tulane University, School of Medicine New Orleans Louisiana USA
- Genetics Unit, Department of Histology and Cell Biology, Faculty of Medicine Suez Canal University Ismailia Egypt
| |
Collapse
|
4
|
Huang PS, Wang CS, Yeh CT, Lin KH. Roles of Thyroid Hormone-Associated microRNAs Affecting Oxidative Stress in Human Hepatocellular Carcinoma. Int J Mol Sci 2019; 20:E5220. [PMID: 31640265 PMCID: PMC6834183 DOI: 10.3390/ijms20205220] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 10/17/2019] [Accepted: 10/18/2019] [Indexed: 02/07/2023] Open
Abstract
Oxidative stress occurs as a result of imbalance between the generation of reactive oxygen species (ROS) and antioxidant genes in cells, causing damage to lipids, proteins, and DNA. Accumulating damage of cellular components can trigger various diseases, including metabolic syndrome and cancer. Over the past few years, the physiological significance of microRNAs (miRNA) in cancer has been a focus of comprehensive research. In view of the extensive level of miRNA interference in biological processes, the roles of miRNAs in oxidative stress and their relevance in physiological processes have recently become a subject of interest. In-depth research is underway to specifically address the direct or indirect relationships of oxidative stress-induced miRNAs in liver cancer and the potential involvement of the thyroid hormone in these processes. While studies on thyroid hormone in liver cancer are abundantly documented, no conclusive information on the potential relationships among thyroid hormone, specific miRNAs, and oxidative stress in liver cancer is available. In this review, we discuss the effects of thyroid hormone on oxidative stress-related miRNAs that potentially have a positive or negative impact on liver cancer. Additionally, supporting evidence from clinical and animal experiments is provided.
Collapse
Affiliation(s)
- Po-Shuan Huang
- Department of Biochemistry, College of Medicine, Chang-Gung University, Taoyuan 33302, Taiwan.
- Department of Biomedical Sciences, College of Medicine, Chang-Gung University, Taoyuan 33302, Taiwan.
| | - Chia-Siu Wang
- Department of General Surgery, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan.
| | - Chau-Ting Yeh
- Liver Research Center, Chang Gung Memorial Hospital, Linkou, Taoyuan 33302, Taiwan.
| | - Kwang-Huei Lin
- Department of Biochemistry, College of Medicine, Chang-Gung University, Taoyuan 33302, Taiwan.
- Department of Biomedical Sciences, College of Medicine, Chang-Gung University, Taoyuan 33302, Taiwan.
- Liver Research Center, Chang Gung Memorial Hospital, Linkou, Taoyuan 33302, Taiwan.
- Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33302, Taiwan.
| |
Collapse
|
5
|
Rana MA, Ijaz B, Daud M, Tariq S, Nadeem T, Husnain T. Interplay of Wnt β-catenin pathway and miRNAs in HBV pathogenesis leading to HCC. Clin Res Hepatol Gastroenterol 2019; 43:373-386. [PMID: 30377095 DOI: 10.1016/j.clinre.2018.09.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 09/05/2018] [Accepted: 09/24/2018] [Indexed: 02/04/2023]
Abstract
The prevalence of Hepatocellular carcinoma (HCC) has been identified world-wide. Plethora of factors including chronic infection of HBV/HCV has been characterized for the development of HCC. Although the onset and progression of HCC has been linked with awry of various signaling pathways but precise mechanism, still lies under the multitude layers of curiosity. HBV is spreading with insane speed throughout the world and has been found a main culprit in HCC development after regulating the several cellular pathways including Wnt/β-catenin, Raf/MAPK, Akt and affecting cell multiplication to genomic instability. The role of Wnt/FZD/β-catenin signaling pathway is centralized in liver functions and its anomalous activation leads to HCC development. β-catenin mainly plays a pivotal role in canonical pathway of the system. Altered mainly overexpression of β-catenin along its nuclear localization tunes the aberrations in liver functions and set disease progression. In the development of HCC, modulation of Wnt/FZD/β-catenin signaling pathway by HBV has been established. As HBV infects the cell it affects the miRNAs, the master regulators of cell. Previous studies showed the connection between HBV and cellular miRNAs. In the present review, we unveiled how HBV is deciphering the cellular miRNAs like miR-26a, miR-15a, miR-16-1, miR-148a, miR-132, miR-122, miR-34a, miR-21, miR-29a, miR-222 and miR-199a/b-3p to modulate the Wnt/FZD/β-catenin signaling pathway and develop HCC. These HBV mediated miRNAs may prove future therapeutic options to treat HBV-Wnt/FZD/β-catenin associated HCC.
Collapse
Affiliation(s)
- Muhammad Adeel Rana
- Department of microbiology, Quaid-i-Azam University, Islamabad, Pakistan; Centre of Excellence in Molecular Biology, University of the Punjab, Lahore Pakistan
| | - Bushra Ijaz
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore Pakistan.
| | - Muhammad Daud
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore Pakistan
| | - Sommyya Tariq
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore Pakistan
| | - Tariq Nadeem
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore Pakistan
| | - Tayyab Husnain
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore Pakistan
| |
Collapse
|
6
|
Chen S, Yang L, Pan A, Duan S, Li M, Li P, Huang J, Gao X, Huang X, Lin Y. Inhibitory Effect on the Hepatitis B Cells through the Regulation of miR-122-MAP3K2 signal pathway. AN ACAD BRAS CIENC 2019; 91:e20180941. [PMID: 31141015 DOI: 10.1590/0001-3765201920180941] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 01/07/2019] [Indexed: 11/22/2022] Open
Abstract
The aim of this study was to investigate the inhibitory effect of regulation of miR-122-MAP3K2 signal pathway on the hepatitis B cells. We detected the content of MAP3K2 from patients with HBV blood serum samples and analyzed the correlation between content of MAP3K2 and copies of HBV-DNA. Wound healing and Transwell assays were used to detect the function of cells from control group (wild type) and observer group (overexpresses miR-122). Secretion levels of HBsAg and MAP3K2 in the supernatant and level of MAP3K2 in cells were detected by ELISA and western blot, respectively. The results showed that there was a positive correlation between the copies of HBV-DNA and MAP3K2 in serum. In the assays involving detection of the number of HBV-DNA copies, the supernatant levels of HBsAg and MAP3K2, and the level of MAP3K2 in the cells, the rate of increase of these indicators significantly slowed as culture time. In conclusion, overexpression of miR-122 could inhibit the migration of hepatoblastoma cells; however, following transfection with miR-122, DNA synthesis and the secretion of HBsAg were inhibited. Overexpression of miR-122 can also downregulate MAP3K2. Consequently, we concluded that regulating the miR-122-MAP3K2 signaling pathway exerts an inhibitory effect in hepatitis B cells.
Collapse
Affiliation(s)
- Songlin Chen
- First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning 530023, Guangxi, People's Republic of China
| | - Lei Yang
- First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning 530023, Guangxi, People's Republic of China
| | - Aiping Pan
- First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning 530023, Guangxi, People's Republic of China
| | - Siliang Duan
- Medical College of Guangxi University of Science and Technology, Liuzhou 545005, Guangxi, People's Republic of China
| | - Mingfen Li
- First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning 530023, Guangxi, People's Republic of China
| | - Ping Li
- First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning 530023, Guangxi, People's Republic of China
| | - Jingjing Huang
- First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning 530023, Guangxi, People's Republic of China
| | - Xingxin Gao
- First Affiliated Hospital of Guangxi Medical University, Nanning 530023, Guangxi, People's Republic of China
| | - Xiaoqi Huang
- First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning 530023, Guangxi, People's Republic of China
| | - Yinghui Lin
- First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning 530023, Guangxi, People's Republic of China
| |
Collapse
|
7
|
Sendi H, Mead I, Wan M, Mehrab-Mohseni M, Koch K, Atala A, Bonkovsky HL, Bishop CE. miR-122 inhibition in a human liver organoid model leads to liver inflammation, necrosis, steatofibrosis and dysregulated insulin signaling. PLoS One 2018; 13:e0200847. [PMID: 30024933 PMCID: PMC6053181 DOI: 10.1371/journal.pone.0200847] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 07/03/2018] [Indexed: 12/12/2022] Open
Abstract
To investigate the role of miR-122 in the development and regression of non-alcoholic fatty liver disease (NAFLD) in vitro, we used multicellular 3D human liver organoids developed in our laboratory. These organoids consist of primary human hepatocytes, Kupffer cells, quiescent stellate cells and liver sinusoidal endothelial cells. They remain viable and functional for 4 weeks expressing typical markers of liver function such as synthesis of albumin, urea, and alpha-1 p450 drug metabolism. Before mixing, hepatic cells were transduced with lentivirus to inhibit miR122 expression (ABM, CA). Immediately after the organoids were fully formed (day 4) or after 1 or 2 weeks of additional incubation (days 11 or 18), the organoids were analyzed using fluorescent live/dead staining and ATP production; total RNA was extracted for qPCR gene expression profiling. Our results show that miR-122 inhibition in liver organoids leads to inflammation, necrosis, steatosis and fibrosis. This was associated with increase in inflammatory cytokines (IL6, TNF), chemokines (CCL2, CCL3) and increase in a subset of Matrix Metaloproteinases (MMP8, MMP9). An altered expression of key genes in lipid metabolism (i.e LPL, LDLR) and insulin signaling (i.e GLUT4, IRS1) was also identified. Conclusion: Our results highlight the role of miR-122 inhibition in liver inflammation, steatofibrosis and dysregulation of insulin signaling. Patients with NAFLD are known to have altered levels of miR-122, therefore we suggest that miR-122 mimics could play a useful role in reversing liver steatofibrosis and insulin resistance seen in patients with NAFLD.
Collapse
Affiliation(s)
- Hossein Sendi
- The Laboratory for Liver Diseases and Metabolic Disorders, Section on Gastroenterology, Department of Internal Medicine, Wake Forest Baptist Medical Center, Winston-Salem, NC, United States of America
- Wake Forest Institute of Regenerative Medicine, Winston-Salem, NC, United States of America
- * E-mail:
| | - Ivy Mead
- Wake Forest Institute of Regenerative Medicine, Winston-Salem, NC, United States of America
| | - Meimei Wan
- Wake Forest Institute of Regenerative Medicine, Winston-Salem, NC, United States of America
| | - Marjan Mehrab-Mohseni
- The Laboratory for Liver Diseases and Metabolic Disorders, Section on Gastroenterology, Department of Internal Medicine, Wake Forest Baptist Medical Center, Winston-Salem, NC, United States of America
| | - Kenneth Koch
- The Laboratory for Liver Diseases and Metabolic Disorders, Section on Gastroenterology, Department of Internal Medicine, Wake Forest Baptist Medical Center, Winston-Salem, NC, United States of America
| | - Anthony Atala
- Wake Forest Institute of Regenerative Medicine, Winston-Salem, NC, United States of America
| | - Herbert L. Bonkovsky
- The Laboratory for Liver Diseases and Metabolic Disorders, Section on Gastroenterology, Department of Internal Medicine, Wake Forest Baptist Medical Center, Winston-Salem, NC, United States of America
| | - Colin E. Bishop
- Wake Forest Institute of Regenerative Medicine, Winston-Salem, NC, United States of America
| |
Collapse
|
8
|
Suehiro T, Miyaaki H, Kanda Y, Shibata H, Honda T, Ozawa E, Miuma S, Taura N, Nakao K. Serum exosomal microRNA-122 and microRNA-21 as predictive biomarkers in transarterial chemoembolization-treated hepatocellular carcinoma patients. Oncol Lett 2018; 16:3267-3273. [PMID: 30127924 DOI: 10.3892/ol.2018.8991] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 05/22/2018] [Indexed: 12/13/2022] Open
Abstract
Exosomal microRNAs (miRNAs) have been investigated as potential novel biomarkers, and miR-122 and miR-21 were shown to be important in hepatocellular carcinoma (HCC). We analyzed the importance of serum exosomal miRNA expression levels in HCC patients that underwent transarterial chemoembolization (TACE). Seventy-five HCC patients who underwent TACE as the initial treatment in Nagasaki University Hospital were enrolled. Exosomal miRNAs were isolated from serum samples collected before and after TACE. Exosomal miR-122 expression levels significantly decreased after TACE (P=0.012), while the exosomal miR-21 expression levels did not significantly change. The expression levels of exosomal miR-122 before TACE were shown to correlate significantly with aspartate aminotransferase (r=0.31, P=0.004) and alanine aminotransferase (r=0.33, P=0.003) levels, tumor diameter (r=0.29, P=0.010) and Child-Pugh score (r=-0.28, P=0.013). The median survival time for all patients was 47 months, and neither of the investigated exosomal miRNAs were shown to be independent factors associated with the disease-specific survival. According to the median relative expression of miR-122 after TACE/before TACE (miR-122 ratio) in liver cirrhosis patients (n=57), the patients with a higher miR-122 ratio had significantly longer disease-specific survival, compared with that of the patients with the lower miR-122 ratio (P=0.0461). Multivariate Cox proportional hazards regression analysis of clinical parameters revealed that a lower exosomal miR-122 ratio (HR 2.720; 95% confidence interval, 1.035-8.022; P=0.042) is associated with the disease-specific survival. Taken together, our results demonstrate that the exosomal miR-122 level alterations may represent a predictive biomarker in HCC patients with liver cirrhosis treated with TACE.
Collapse
Affiliation(s)
- Tomoyuki Suehiro
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Hisamitsu Miyaaki
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Yasuko Kanda
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Hidetaka Shibata
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Takuya Honda
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Eisuke Ozawa
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Satoshi Miuma
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Naota Taura
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Kazuhiko Nakao
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| |
Collapse
|
9
|
Xie Y, He S, Wang J. MicroRNA-373 facilitates HSV-1 replication through suppression of type I IFN response by targeting IRF1. Biomed Pharmacother 2018; 97:1409-1416. [DOI: 10.1016/j.biopha.2017.11.071] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Revised: 10/23/2017] [Accepted: 11/10/2017] [Indexed: 01/31/2023] Open
|
10
|
Chelomina GN. Genomics and transcriptomics of the Chinese liver fluke Clonorchis sinensis (Opisthorchiidae, Trematoda). Mol Biol 2017. [DOI: 10.1134/s0026893317020078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
11
|
Wang N, Wang Q, Shen D, Sun X, Cao X, Wu D. Downregulation of microRNA-122 promotes proliferation, migration, and invasion of human hepatocellular carcinoma cells by activating epithelial-mesenchymal transition. Onco Targets Ther 2016; 9:2035-47. [PMID: 27103830 PMCID: PMC4827919 DOI: 10.2147/ott.s92378] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Objective To investigate the effects of microRNA-122 (miR-122) on proliferation, migration, and invasion in human hepatocellular carcinoma (HCC) cells by activating epithelial–mesenchymal transition (EMT) pathways. Methods miR-122 mimics, miR-122 inhibitors, relevant control oligonucleotides, and Wnt1 were transfected into HepG2 and huh7 cell lines which were then divided into six groups: miR-122 group, anti-miR-122 group, miR-negative control (NC) group, anti-miR-NC group, miR-122 + Wnt1 group, and miR-122 + vector group. The miR-122 expressions and mRNA expressions of Wnt1 and EMT-related genes (E-cadherin, vimentin, β-cadherin, and N-cadherin) were quantified by quantitative real-time polymerase chain reaction (qRT-PCR). Protein expression levels of Wnt1, E-cadherin, vimentin, β-cadherin, and N-cadherin were measured by Western blot. Cell proliferation, migration, and invasion were evaluated using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, wound-healing assay, and Transwell assay, respectively. Results Dual luciferase reporter gene results showed that Wnt1 is a direct target gene of miR-122 in both HepG2 and huh7 cell lines. Compared to miR-NC, anti-miR-NC, and miR-122 + Wnt1 groups, miR-122 expression was markedly higher in the miR-122 group and miR-122 + vector group, but was sharply decreased in anti-miR-122 group (both P<0.05), and the mRNA and protein levels of Wnt1, vimentin, β-cadherin, and N-cadherin decreased significantly; also E-cadherin increased, and cell proliferation, migration, and invasion decreased in the miR-122 group and miR-122 + vector group (all P<0.05), but the situation was totally reversed in the anti-miR-122 group (all P<0.05). Conclusion Downregulation of miR-122 promoted proliferation, migration, and invasion of human HCC cells by targeting Wnt1 and regulating Wnt/β-catenin pathway which activated the EMT pathways.
Collapse
Affiliation(s)
- Nanyao Wang
- Department of Oncology, The Affiliated Jiangyin Hospital, School of Medicine, Southeast University, Jiangyin, Jiangsu, People's Republic of China
| | - Qiong Wang
- Department of Oncology, The Affiliated Jiangyin Hospital, School of Medicine, Southeast University, Jiangyin, Jiangsu, People's Republic of China
| | - Dong Shen
- Department of Oncology, The Affiliated Jiangyin Hospital, School of Medicine, Southeast University, Jiangyin, Jiangsu, People's Republic of China
| | - Xia Sun
- Department of Oncology, The Affiliated Jiangyin Hospital, School of Medicine, Southeast University, Jiangyin, Jiangsu, People's Republic of China
| | - Xiangming Cao
- Department of Oncology, The Affiliated Jiangyin Hospital, School of Medicine, Southeast University, Jiangyin, Jiangsu, People's Republic of China
| | - Dan Wu
- Department of Oncology, The Affiliated Jiangyin Hospital, School of Medicine, Southeast University, Jiangyin, Jiangsu, People's Republic of China
| |
Collapse
|
12
|
He J, Zhao K, Zheng L, Xu Z, Gong W, Chen S, Shen X, Huang G, Gao M, Zeng Y, Zhang Y, He F. Upregulation of microRNA-122 by farnesoid X receptor suppresses the growth of hepatocellular carcinoma cells. Mol Cancer 2015; 14:163. [PMID: 26302777 PMCID: PMC4547435 DOI: 10.1186/s12943-015-0427-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 08/04/2015] [Indexed: 12/31/2022] Open
Abstract
Background microRNA-122 (miR-122) is the most abundant and specific miRNA in the liver. It acts as an important tumor suppressor in hepatocellular carcinoma (HCC) through regulating its target genes, but details of its own regulation are largely unknown. Farnesoid X receptor (FXR), a transcription factor with multiple functions, plays an important role in protecting against liver carcinogenesis, but it is unclear whether the anti-HCC effect of FXR is involved in the regulation of miR-122. Methods The levels of miR-122 and FXR in HCC tissues and cell lines were examined by quantitative real-time PCR (qRT-PCR). qRT-PCR was also used to detect the expression of miR-122 target genes at mRNA level, while Western blotting was used to analyze that of their protein products. The effect of FXR on the transcriptional activity of miR-122 promoter was evaluated by a luciferase reporter assay. Electrophoretic mobility shift assay (EMSA) and chromatin immunoprecipitation (ChIP) assay were performed to identify the FXR binding site within miR-122 promoter region. The cell proliferation was analyzed by a CCK-8 assay. The influence of FXR on tumor growth and miR-122 expression in vivo was monitored using HCC xenografts in nude mice. Results The expression of FXR was positively correlated with that of miR-122 in HCC tissues and cell lines. Activation of FXR in HCC cells upregulated miR-122 expression and in turn downregulated the expression of miR-122 target genes including insulin-like growth factor-1 receptor and cyclin G1. FXR bound directly to the DR2 element (−338 to −325) in miR-122 promoter region, and enhanced the promoter’s transcriptional activity. Functional experiments showed that the FXR-mediated upregulation of miR-122 suppressed the proliferation of HCC cells in vitro and the growth of HCC xenografts in vivo. Conclusions miR-122 is a novel target gene of FXR, and the upregulation of miR-122 by FXR represses the growth of HCC cells, suggesting that FXR may serve as a key transcriptional regulator for manipulating miR-122 expression, and the FXR/miR-122 pathway may therefore be a novel target for the treatment of HCC. Electronic supplementary material The online version of this article (doi:10.1186/s12943-015-0427-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jialin He
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing, 400038, China.
| | - Kai Zhao
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing, 400038, China.
| | - Lu Zheng
- Department of Hepatobiliary Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
| | - Zhizhen Xu
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing, 400038, China.
| | - Wei Gong
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing, 400038, China.
| | - Shan Chen
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing, 400038, China.
| | - Xiaodong Shen
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing, 400038, China.
| | - Gang Huang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing, 400038, China.
| | - Min Gao
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing, 400038, China.
| | - Yijun Zeng
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing, 400038, China.
| | - Yan Zhang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing, 400038, China.
| | - Fengtian He
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing, 400038, China.
| |
Collapse
|
13
|
Sendi H, Mehrab-Mohseni M, Foureau DM, Ghosh S, Walling TL, Steuerwald N, Zamor PJ, Kaplan KJ, Jacobs C, Ahrens WA, Russo MW, Clemens MG, Schrum LW, Bonkovsky HL. MiR-122 decreases HCV entry into hepatocytes through binding to the 3' UTR of OCLN mRNA. Liver Int 2015; 35:1315-23. [PMID: 25302477 DOI: 10.1111/liv.12698] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 10/02/2014] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Analysis in silico suggests that occludin (OCLN), a key receptor for HCV, is a candidate target of miR-122; the most abundant hepatic micro RNA. We aimed to determine if miR-122 can decrease HCV entry through binding to the 3' UTR of OCLN mRNA. DESIGN Huh7.5 cells were cotransfected with luciferase construct containing 3' UTR of OCLN (pLuc-OCLN) and with selected miRNAs (0-50 nM) and luciferase activity was measured. Huh7.5 cells were also infected by viral particles containing lenti-miR122 genome or control virus. After 48 h, the cells were infected with HCV pseudo-particles (HCVpp) and VSV pseudo-particles (VSVpp). After 72 h of infection, luciferase activity was measured and HCVpp activity was normalized to VSVpp activity. RESULTS miR-122 binds to the 3'-UTR of OCLN and down-regulates its expression; cotransfection of miR-122 mimic with pLuc-OCLN resulted in a significant decrease in luciferase activity [by 55% (P < 0.01)], while a non-specific miRNA and a mutant miR-122 did not have any effect. miR-122 mimic significantly down-regulated [by 80% (P < 0.01)] OCLN protein in Huh7.5 cells. Accordingly, patients with chronic hepatitis C and higher levels of hepatic miR-122 have lower hepatic expression of OCLN. Immuno-fluorescence imaging showed a decrease in colocalization of OCLN and CLDN following miR-122 over-expression in HCV infected cells. Huh7.5 cells transiently expressing Lenti-miR122 system showed 42% (P < 0.01) decrease in HCV entry. CONCLUSION This study uncovers a novel antiviral effect of miR-122 on human liver cells and shows that over-expression of miR-122 can decrease HCV entry into hepatocytes through down-regulation of OCLN.
Collapse
Affiliation(s)
- Hossein Sendi
- The Liver-Biliary-Pancreatic Center, Carolinas Medical Center, Charlotte, NC, USA; Department of Biology, University of North Carolina at Charlotte, Charlotte, NC, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Gao D, Zhai A, Qian J, Li A, Li Y, Song W, Zhao H, Yu X, Wu J, Zhang Q, Kao W, Wei L, Zhang F, Zhong Z. Down-regulation of suppressor of cytokine signaling 3 by miR-122 enhances interferon-mediated suppression of hepatitis B virus. Antiviral Res 2015; 118:20-8. [PMID: 25766860 DOI: 10.1016/j.antiviral.2015.03.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2014] [Revised: 02/26/2015] [Accepted: 03/01/2015] [Indexed: 12/21/2022]
Abstract
MicroRNA-122 (miR-122) is involved in the pathogenesis of several liver diseases, including chronic hepatitis B infection and hepatocellular carcinoma. This study aimed to explore the potential role of miR-122 in the interferon (IFN)-mediated suppression of hepatitis B virus (HBV) in hepatocytes. We found that elevated expression of suppressor of cytokine signaling 3 (SOCS3) following HBV infection, contributed to the inactivation of the IFN signaling pathway. Based on previous studies from our laboratory showing that miR-122 can modulate type I IFN expression by inhibiting SOCS1 expression, we analyzed the SOCS3 mRNA sequence for putative miR-122 binding sites. We demonstrate that miR-122 inhibits SOCS3 expression by targeting the 3'-untranslated region of the SOCS3 mRNA within the region 1887-1910 nucleotides. Finally, we demonstrate that significantly increased levels of IFN lead to decreased HBV expression in miR-122 mimic-treated Huh7 cells, whereas inhibition of endogenous miR-122 leads to enhanced viral production, owing to a marked decrease in IFN expression. Taken together, our results demonstrate that miR-122 down-regulates SOCS3, thus positively affecting the anti-HBV efficiency of endogenous type I IFN. Our study suggests that suppression of miR-122 induced by HBV infection, leads to the inactivation of IFN expression, which in turn enhances HBV replication, contributing to viral persistence and hepatocarcinogenesis.
Collapse
Affiliation(s)
- Dongni Gao
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, The Heilongjiang Key Laboratory of Immunity and Infection, Pathogenic Biology, Harbin 150086, Heilongjiang, China; Laboratory of Microbiology, College of Life Science, Heilongjiang University, Harbin, China
| | - Aixia Zhai
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, The Heilongjiang Key Laboratory of Immunity and Infection, Pathogenic Biology, Harbin 150086, Heilongjiang, China
| | - Jun Qian
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, The Heilongjiang Key Laboratory of Immunity and Infection, Pathogenic Biology, Harbin 150086, Heilongjiang, China
| | - Aimei Li
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, The Heilongjiang Key Laboratory of Immunity and Infection, Pathogenic Biology, Harbin 150086, Heilongjiang, China
| | - Yujun Li
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, The Heilongjiang Key Laboratory of Immunity and Infection, Pathogenic Biology, Harbin 150086, Heilongjiang, China
| | - Wuqi Song
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, The Heilongjiang Key Laboratory of Immunity and Infection, Pathogenic Biology, Harbin 150086, Heilongjiang, China
| | - Hong Zhao
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, The Heilongjiang Key Laboratory of Immunity and Infection, Pathogenic Biology, Harbin 150086, Heilongjiang, China
| | - Xin Yu
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, The Heilongjiang Key Laboratory of Immunity and Infection, Pathogenic Biology, Harbin 150086, Heilongjiang, China
| | - Jing Wu
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, The Heilongjiang Key Laboratory of Immunity and Infection, Pathogenic Biology, Harbin 150086, Heilongjiang, China
| | - Qingmeng Zhang
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, The Heilongjiang Key Laboratory of Immunity and Infection, Pathogenic Biology, Harbin 150086, Heilongjiang, China
| | - Wenping Kao
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, The Heilongjiang Key Laboratory of Immunity and Infection, Pathogenic Biology, Harbin 150086, Heilongjiang, China
| | - Lanlan Wei
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, The Heilongjiang Key Laboratory of Immunity and Infection, Pathogenic Biology, Harbin 150086, Heilongjiang, China
| | - Fengmin Zhang
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, The Heilongjiang Key Laboratory of Immunity and Infection, Pathogenic Biology, Harbin 150086, Heilongjiang, China.
| | - Zhaohua Zhong
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, The Heilongjiang Key Laboratory of Immunity and Infection, Pathogenic Biology, Harbin 150086, Heilongjiang, China.
| |
Collapse
|
15
|
Zhang Z, Chen J, He Y, Zhan X, Zhao R, Huang Y, Xu H, Zhu Z, Liu Q. miR-125b inhibits hepatitis B virus expression in vitro through targeting of the SCNN1A gene. Arch Virol 2014; 159:3335-43. [PMID: 25173609 DOI: 10.1007/s00705-014-2208-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Accepted: 08/19/2014] [Indexed: 12/17/2022]
Abstract
microRNAs (miRNAs) are small noncoding RNAs that modulate gene expression at the posttranscriptional level, playing an important role in many diseases. However, reports concerning the role of miRNA in hepatitis B virus (HBV) infection are limited. miRNA chips were used to investigate miRNA changes during HBV infection in vitro. Bioinformatics analysis was used to explore possible miRNA and target genes during HBV infection. The expression of miR-125b and its potential target gene, sodium channel, non-voltage-gated 1 alpha (SCNN1A), was further analyzed. A total of 136 miRNAs were analyzed in an HBV transient transfection model (HepG2-HBV1.3), and 78 miRNAs were differentially expressed in HepG2.2.15 cells compared with HepG2 cells. miR-125b expression was decreased in both HepG2-HBV1.3 and HepG2.2.15 cells, and ectopic expression of miR-125b inhibited HBV DNA intermediates and secretion of HBsAg and HBeAg. miR-125b also inhibited the mRNA and protein levels of SCNN1A. Using a dual luciferase reporter system, we found that SCNN1A was one of the targets of miR-125b. In this study, we found that miR-125b inhibits HBV expression in vitro by regulating SCNN1A expression.
Collapse
Affiliation(s)
- Zhenzhen Zhang
- Department of Infectious Disease, Children's Hospital of Chongqing Medical University, No 136 ZhongShan Er Road, 400012, Chongqing, China,
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Xu G, Yang F, Ding CL, Wang J, Zhao P, Wang W, Ren H. MiR-221 accentuates IFN׳s anti-HCV effect by downregulating SOCS1 and SOCS3. Virology 2014; 462-463:343-50. [PMID: 25019494 DOI: 10.1016/j.virol.2014.06.024] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 06/11/2014] [Accepted: 06/17/2014] [Indexed: 12/30/2022]
Abstract
MiR-221 was reported to be upregulated and play roles in tumorigenesis of hepatitis C virus (HCV) associated hepatocellular carcinoma (HCC). However, the role of miR-221 in HCV infection remains unknown. In this study, it was found that miR-221 was upregulated in serum of HCV chronic hepatitis patients and Huh7.5.1 cells infected with HCVcc. Further studies indicated that miR-221 mimic could accentuate anti-HCV effect of IFN-α in HCVcc model, miR-221 mimic could further repressed 10% HCV RNA expression and 35-42% HCV core or NS5A protein expression in HCVcc infected Huh7.5.1 cells treated with 100IU/mL IFN-α, and miR-221 inhibitor resulted in the reverse effects. Furthermore, two members of suppressor of cytokine signaling (SOCS) family, SOCS1 and SOCS3, which are well established inhibitory factors on IFN/JAK/STAT pathway, were identified as the targets of miR-221 and were involved in the effect of miR-221. In conclusion, miR-221 could accentuate IFN׳s anti-HCV effect by targeting SOCS1 and SOCS3.
Collapse
Affiliation(s)
- Gang Xu
- Department of Microbiology, Shanghai Key Laboratory of Medical Biodefense, Second Military Medical University, Shanghai 200433, China
| | - Fang Yang
- Department of Hepatobiliary Surgery, Fuzhou General Hospital of Nanjing Military Area Command, Fuzhou 350025, China
| | - Cui-Ling Ding
- Department of Microbiology, Shanghai Key Laboratory of Medical Biodefense, Second Military Medical University, Shanghai 200433, China
| | - Jing Wang
- Department of Pharmacy, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Ping Zhao
- Department of Microbiology, Shanghai Key Laboratory of Medical Biodefense, Second Military Medical University, Shanghai 200433, China
| | - Wen Wang
- Department of Microbiology, Shanghai Key Laboratory of Medical Biodefense, Second Military Medical University, Shanghai 200433, China.
| | - Hao Ren
- Department of Microbiology, Shanghai Key Laboratory of Medical Biodefense, Second Military Medical University, Shanghai 200433, China.
| |
Collapse
|
17
|
Nakao K, Miyaaki H, Ichikawa T. Antitumor function of microRNA-122 against hepatocellular carcinoma. J Gastroenterol 2014; 49:589-93. [PMID: 24531873 DOI: 10.1007/s00535-014-0932-4] [Citation(s) in RCA: 115] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2014] [Accepted: 01/06/2014] [Indexed: 02/04/2023]
Abstract
MicroRNA-122 (miR-122), a highly abundant and liver-specific miRNA, acts as a tumor suppressor against hepatocellular carcinoma (HCC). Decreased expression of miR-122 in HCC is frequently observed and is associated with poor differentiation, larger tumor size, metastasis and invasion, and poor prognosis. Mutant mice with knockout (KO) of the miR-122 locus developed steatohepatitis due to increased triglyceride (TG) synthesis and decreased TG secretion from hepatocytes, and eventually developed HCC. Exogenic miR-122 introduction into miR-122 KO mice inhibited the development of HCC. Target genes of miR-122, including cyclin G1, a disintegrin and metalloprotease (ADAM)10, serum response factor, insulin-like growth factor-1 receptor, ADAM17, transcription factor CUTL1, the embryonic isoform of pyruvate kinase (Pkm2), Wnt1, pituitary tumor-transforming gene 1 binding factor, Cut-like homeobox 1, and c-myc, are involved in hepatocarcinogenesis, epithelial mesenchymal transition, and angiogenesis. MiR-122 expression is regulated by liver-enriched transcription factors such as hepatocyte nuclear factor (HNF)1α, HNF3β, HNF4α, HNF6, and CCAAT/enhancer-binding protein (C/EBP)α. A positive feedback loop exists between C/EBPα and miR-122 and between HNF6 and miR-122, whereas a negative feedback loop exists between c-myc and miR-122. Since cotreatment of 5-Aza-Cd and histone deacetylase inhibitor restored miR-122 expression in HCC cells, epigenetic modulation of miR-122 expression is involved in the suppression of miR-122 in HCC. Several experiments suggest that increasing miR-122 levels in HCC with or without antitumor agents may be a promising strategy for HCC treatment.
Collapse
Affiliation(s)
- Kazuhiko Nakao
- Department of Gastroenterology and Hepatology, Graduate School of Biomedical Sciences, Nagasaki University, Sakamoto 1-7-1, Nagasaki, 852-8501, Japan,
| | | | | |
Collapse
|
18
|
Gupta P, Cairns MJ, Saksena NK. Regulation of gene expression by microRNA in HCV infection and HCV-mediated hepatocellular carcinoma. Virol J 2014; 11:64. [PMID: 24690114 PMCID: PMC3977900 DOI: 10.1186/1743-422x-11-64] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 03/27/2014] [Indexed: 02/06/2023] Open
Abstract
MicroRNA (miRNA) exert a profound effect on Hepatitis C virus (HCV) replication and on the manifestation of HCV-associated hepatocellular carcinoma (HCC). miR-122 in particular, is highly enriched in liver and has been shown to interact with HCV, suggesting this virus has evolved to subvert and manipulate the host gene silencing machinery in order to support its life cycle. It is therefore likely that miR-122 and other miRNAs play an important role in the pathophysiology of HCV infection. The changes in post-transcriptional gene regulation by the miRNAs may play a key role in the manifestation of chronic liver disease and hepatocellular carcinoma. Understanding of HCV-host miRNA interactions will ultimately lead to the design of therapeutic modalities against HCV infection and HCV-mediated HCC and may also provide important biomarkers that direct treatment options. Here, we review the current knowledge on the role of miRNA and gene expression on HCV infection and hepatocellular carcinoma, in addition to the possible role of miRNA as future therapeutic targets.
Collapse
Affiliation(s)
| | | | - Nitin K Saksena
- Centre for Virus Research, Westmead Millennium Institute, Darcy Road, Sydney, Westmead NSW 2145, Australia.
| |
Collapse
|
19
|
Aalaei-andabili SH, Rezaei N. Toll like receptor (TLR)-induced differential expression of microRNAs (MiRs) promotes proper immune response against infections: a systematic review. J Infect 2013; 67:251-64. [PMID: 23850616 DOI: 10.1016/j.jinf.2013.07.016] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Revised: 05/25/2013] [Accepted: 07/06/2013] [Indexed: 12/19/2022]
Abstract
Toll like receptors (TLRs) are one of the major families of pattern recognition receptors (PRRs). MicroRNAs (MiRs) are small noncoding RNAs with regulatory effects on biological process, and it has been recently shown that they can control inflammatory process and the response to an infection by modulating the function of TLRs. In this study, we designed a systematic review to clarify the reciprocal interaction between TLRs and MiRs, in order to identify possible future therapeutic targets and strategies. On the one hand, TLRs stimulation can change expression level of miRs in various ways, which can lead to modulating their effects. On the other hand, MiRs also influence the expression of TLRs and the intensity of the inflammatory reaction. We therefore conclude that the interaction between MiRs and TLRs is a key regulator of innate immune system. Investigations discovering therapeutic approaches by manipulation of miRs expression level may open a new approach for the treatment of inflammatory diseases.
Collapse
|
20
|
Gehrau RC, Mas VR, Maluf DG. Hepatic disease biomarkers and liver transplantation: what is the potential utility of microRNAs? Expert Rev Gastroenterol Hepatol 2013; 7:157-70. [PMID: 23363264 DOI: 10.1586/egh.12.71] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Liver transplantation represents the treatment of choice for acute hepatic failure or chronically induced end stage of liver disease. Molecular characterization of hepatic injury and recovery processes encloses the key for biomarker discovery in the liver transplantation field. Several pathological hepatic processes were demonstrated to be associated with deregulated miRNA profiles. Moreover, abnormal concentration levels of circulating cell-free miRNAs correlate with specific hepatic injury. The high molecular stability and emerging rapid assessment techniques invite further consideration of miRNAs as feasible monitoring and outcome predictive biomarkers for liver disease and liver transplantation. The present review aims to provide an overview of recent achievements in research on the potential applicability of miRNAs as biomarkers in liver disease and liver transplantation.
Collapse
Affiliation(s)
- Ricardo C Gehrau
- Department of Surgery, University of Virginia, Transplant Division, PO Box 800625, 409 Lane Rd, Charlottesville, VA 22908-0625, USA
| | | | | |
Collapse
|
21
|
Can tobacco use promote HCV-induced miR-122 hijacking and hepatocarcinogenesis? Med Hypotheses 2012; 80:131-3. [PMID: 23218444 DOI: 10.1016/j.mehy.2012.11.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Revised: 11/08/2012] [Accepted: 11/09/2012] [Indexed: 01/22/2023]
Abstract
Chronic hepatitis C virus (HCV) infection is a well-recognized risk factor for hepatocellular carcinoma (HCC). As a co-risk factor, the role of tobacco use in HCV-driven carcinogenesis and relevant underlying mechanisms remain largely unclear. The latest discoveries about HCV replication have shown that HCV RNA hijacks cellular miRNA-122 by forming an Ago2-HCV-miR-122 complex that stabilizes the HCV genome and enhances HCV replication. Our previous work has demonstrated that aqueous tobacco smoke extract (TSE) is a potent activator of HIV replication via TSE-mediated viral protection from oxidative stress and activation of a set of genes that can promote viral replication. Since HCV is, like HIV, an enveloped virus that should be equally susceptible to lipid peroxidation, and since one of the TSE-upregulated genes, the DDX3 helicase, is known to facilitate HCV replication, we hypothesize that (1) tobacco use can similarly enhance HCV viability and replication, and promote HCC progression by up-regulation of DDX3, and (2) by competing for binding with miR-122 as a competing endogenous RNA (ceRNA), HCV replication can liberate miR-122's direct target, oncogenic gene cyclin G1 (CCNG1); furthermore, simultaneous tobacco use can synergistically enhance this competing effect via HCV upregulation. Our hypotheses may lay a foundation for better understanding of carcinogenesis in HCV-driven HCC and the potential role of tobacco as a cofactor. Disrupting the HCV ceRNA effect may provide a new strategy for designing anti HCV/HCC drugs.
Collapse
|