51
|
Shermane Lim YW, Xiang X, Garg M, Le MT, Li-Ann Wong A, Wang L, Goh BC. The double-edged sword of H19 lncRNA: Insights into cancer therapy. Cancer Lett 2020; 500:253-262. [PMID: 33221454 DOI: 10.1016/j.canlet.2020.11.006] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 10/25/2020] [Accepted: 11/06/2020] [Indexed: 01/03/2023]
Abstract
H19 long non-coding RNA (lncRNA) has many functions in cancer. Some studies have reported that H19 acts as an oncogene and is involved in cancer progression by activating epithelial-mesenchymal transition (EMT), the cell cycle and angiogenesis via mechanisms like microRNA (miRNA) sponging - the binding to and inhibition of miRNA activity. This makes H19 lncRNA a potential target for cancer therapeutics. However, several conflicting studies have also found that H19 suppresses tumour development. In this review, we shed light on the possible reasons for these conflicting findings. We also summarise the current literature on the applications of H19 lncRNA in cancer therapy in many cancers and explore new avenues for future research. This includes the use of H19 in recombinant vectors, chemoresistance, epigenetic regulation, tumour microenvironment alteration and cancer immunotherapy. The relationship between H19 and the master tumour suppressor gene p53 is also explored. In most studies, H19 knockdown via RNA interference (RNAi) or epigenetic silencing inhibits cancer development. Thus, H19 lncRNA could be a promising target for the development of cancer therapeutics. This warrants further investigations into its translational research to improve cancer therapy outcomes.
Collapse
Affiliation(s)
- Yun Wei Shermane Lim
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore; Institute for Digital Medicine and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
| | - Xiaoqiang Xiang
- Department of Clinical Pharmacy, School of Pharmacy, Fudan University, Shanghai, 201203, China.
| | - Manoj Garg
- Amity Institute of Molecular Medicine & Stem Cell Research (AIMMSCR), Amity University, Sector-125, Noida, 201313, India
| | - Minh Tn Le
- Institute for Digital Medicine and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
| | - Andrea Li-Ann Wong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore; Department of Haematology-Oncology, National University Cancer Institute, Singapore, 119228, Singapore; Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
| | - Lingzhi Wang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore; Institute for Digital Medicine and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
| | - Boon-Cher Goh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore; Institute for Digital Medicine and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore; Department of Haematology-Oncology, National University Cancer Institute, Singapore, 119228, Singapore; Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
| |
Collapse
|
52
|
Pegoraro S, Ros G, Sgubin M, Petrosino S, Zambelli A, Sgarra R, Manfioletti G. Targeting the intrinsically disordered architectural High Mobility Group A (HMGA) oncoproteins in breast cancer: learning from the past to design future strategies. Expert Opin Ther Targets 2020; 24:953-969. [PMID: 32970506 DOI: 10.1080/14728222.2020.1814738] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Triple-negative breast cancer (TNBC) is the most difficult breast cancer subtype to treat because of its heterogeneity and lack of specific therapeutic targets. High Mobility Group A (HMGA) proteins are chromatin architectural factors that have multiple oncogenic functions in breast cancer, and they represent promising molecular therapeutic targets for this disease. AREAS COVERED We offer an overview of the strategies that have been exploited to counteract HMGA oncoprotein activities at the transcriptional and post-transcriptional levels. We also present the possibility of targeting cancer-associated factors that lie downstream of HMGA proteins and discuss the contribution of HMGA proteins to chemoresistance. EXPERT OPINION Different strategies have been exploited to counteract HMGA protein activities; these involve interfering with their nucleic acid binding properties and the blocking of HMGA expression. Some approaches have provided promising results. However, some unique characteristics of the HMGA proteins have not been exploited; these include their extensive protein-protein interaction network and their intrinsically disordered status that present the possibility that HMGA proteins could be involved in the formation of proteinaceous membrane-less organelles (PMLO) by liquid-liquid phase separation. These unexplored characteristics could open new pharmacological avenues to counteract the oncogenic contributions of HMGA proteins.
Collapse
Affiliation(s)
- Silvia Pegoraro
- Department of Life Sciences, University of Trieste , Trieste, Italy
| | - Gloria Ros
- Department of Life Sciences, University of Trieste , Trieste, Italy
| | - Michela Sgubin
- Department of Life Sciences, University of Trieste , Trieste, Italy
| | - Sara Petrosino
- Department of Life Sciences, University of Trieste , Trieste, Italy
| | | | - Riccardo Sgarra
- Department of Life Sciences, University of Trieste , Trieste, Italy
| | | |
Collapse
|
53
|
Zhou Y, Chen B. GAS5‑mediated regulation of cell signaling (Review). Mol Med Rep 2020; 22:3049-3056. [PMID: 32945519 PMCID: PMC7453608 DOI: 10.3892/mmr.2020.11435] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 06/19/2020] [Indexed: 12/11/2022] Open
Abstract
In recent years, an increasing number of long non-coding RNAs (lncRNAs) have been discovered using microarrays and nucleic acid sequencing technology. LncRNAs exert crucial biological functions by regulating signaling pathways. In particular, the lncRNA growth arrest-specific transcript 5 (GAS5) has been documented to serve a crucial role in numerous signaling pathways. This article discusses the latest developments in the association between GAS5 and microRNA (miRNA), p53, mTOR, glucocorticoid response element (GRE) and AKT in order to investigate the roles served by GAS5. miRNAs can activate related signaling pathways and GAS5 can combine with miRNA to regulate related signaling pathways. GAS5 may regulate p53 expression via derivation of snoRNA, but the underlying mechanism requires further investigation. GAS5 overxpresion reduces the expression level of mTOR, which is induced by inhibiting miR-106a-5p expression. GAS5 is a sponge of GR, and serves a role in controlling and maintaining glucocorticoid sensitivity and drug resistance via competitive combination with GR. GAS5 can interact with miRNAs, such as miR-21 and miR-532-5p, to regulate the expression of AKT signaling pathway, affecting cell survival and apoptosis. Collectively, the data indicate that GAS5 serves a key role in the miRNA, p53, mTOR, GRE, and AKT signaling pathways. GAS5 regulates complex intracellular signaling pathways primarily through three modes of action, all of which are interrelated: Signal, decoy and guide. In the present article, latest developments in the association between GAS5 and a number of cellular signaling pathways are discussed to examine the tumor suppressive role of GAS5.
Collapse
Affiliation(s)
- Yang Zhou
- Department of Urology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212000, P.R. China
| | - Binghai Chen
- Department of Urology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212000, P.R. China
| |
Collapse
|
54
|
Shi X, Wei YT, Li H, Jiang T, Zheng XL, Yin K, Zhao GJ. Long non-coding RNA H19 in atherosclerosis: what role? Mol Med 2020; 26:72. [PMID: 32698876 PMCID: PMC7374855 DOI: 10.1186/s10020-020-00196-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 07/07/2020] [Indexed: 12/14/2022] Open
Abstract
Atherosclerosis (AS) is widely accepted to be a multistep pathophysiological process associated with several other processes such as angiogenesis and inflammatory response. Long non-coding RNAs (lncRNAs) are non-protein coding RNAs (more than 200 nucleotides in length) and can regulate gene expression at the transcriptional and post-transcriptional levels. Recent studies suggest that lncRNA-H19 plays important roles in the regulation of angiogenesis, adipocyte differentiation, lipid metabolism, inflammatory response, cellular proliferation and apoptosis. In this review, we primarily discuss the roles of lncRNA-H19 in atherosclerosis-related pathophysiological processes and the potential mechanisms by which lncRNA-H19 regulates the development of atherosclerosis, to help provide a better understanding of the biological functions of lncRNA-H19 in atherosclerosis.
Collapse
Affiliation(s)
- Xian Shi
- School of Medicine, Guilin Medical University, Guilin, 541100, Guangxi, China
| | - Ya-Ting Wei
- School of Medicine, Guilin Medical University, Guilin, 541100, Guangxi, China
| | - Heng Li
- Institute of Cardiovascular Research, Key Laboratory for Arteriosclerology of Hunan Province, University of South China, Hengyang, 421001, Hunan, China
| | - Ting Jiang
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan City People's Hospital, Qingyuan, 511518, Guangdong, China
| | - Xi-Long Zheng
- Department of Biochemistry and Molecular Biology, The Libin Cardiovascular Institute of Alberta, The University of Calgary, Health Sciences Center, Calgary, AB, Canada.,Key Laboratory of Molecular Targets and Clinical Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, Guangdong, China
| | - Kai Yin
- Center for Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, The Second Affiliated Hospital of Guilin Medical University, Guilin, 541100, Guangxi, China.
| | - Guo-Jun Zhao
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan City People's Hospital, Qingyuan, 511518, Guangdong, China.
| |
Collapse
|
55
|
Xu Z, Zhang L, Yu Q, Zhang Y, Yan L, Chen ZJ. The estrogen-regulated lncRNA H19/miR-216a-5p axis alters stromal cell invasion and migration via ACTA2 in endometriosis. Mol Hum Reprod 2020; 25:550-561. [PMID: 31323679 DOI: 10.1093/molehr/gaz040] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 06/27/2019] [Accepted: 07/10/2019] [Indexed: 12/11/2022] Open
Abstract
Fibrotic tissue may contribute to the origin of some endometriosis-related symptoms, such as chronic pelvic pain and infertility. Alterations in the H19/miR-216a-5p/ACTA2 pathway may mediate the regulation of eutopic endometrial stromal cell (euESC) invasion and migration and may represent a potential mechanism underlying fibrous tissue formation or fibrosis in women with endometriosis. In this study, we aimed to determine the expression of H19 and ACTA2 in endometrial tissues of women with endometriosis. Two groups of 23 infertile women with endometriosis and 23 matched infertile women without endometriosis were investigated. Primary cultured cells of endometrial tissues were analyzed using RT-PCR and western blotting (WB) to determine expression of H19 and ACTA2. 5-Ethyl-2'-deoxyuridine, CCK8 and Transwell assays were used to study the functions of H19 and ACTA2. Human embryonic kidney 293 cells were used for luciferase assays to study miR-216a-5p binding sites with H19 and ACTA2. We found that H19 and ACTA2 levels were significantly higher in endometriosis euESCs than in control euESCs (P < 0.05) and were positively correlated in endometriosis euESCs. Luciferase assays indicated that H19 regulates ACTA2 expression via competition for inhibitory miR-216a-5p binding sites. Our results indicate that alterations in the estrogen/H19/miR-216a-5p/ACTA2 pathway regulated endometriosis euESC invasion and migration. Downregulation of H19 or ACTA2 inhibited endometriosis euESC invasion and migration; however, estrogen promoted endometriosis euESC invasion and migration via H19. The main limitation of our study was that experiments were conducted in vitro and further in vivo studies are required in the future. However, our study showed that primary cultured cells represented endometriosis cells more clearly than cell lines.
Collapse
Affiliation(s)
- Zhen Xu
- School of Medicine, Shandong University, 44 Wenhua West Road, Jinan, China.,Reproductive Hospital Affiliated to Shandong University, 157 Jingliu Road, Jinan, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Key Laboratory of Reproductive Endocrinology, Shandong University, Ministry of Education, 157 Jingliu Road, Jinan, China.,Shandong Provincial Key Laboratory of Reproductive Medicine, 157 Jingliu Road, Jinan, China
| | - Liping Zhang
- School of Medicine, Shandong University, 44 Wenhua West Road, Jinan, China.,Reproductive Hospital Affiliated to Shandong University, 157 Jingliu Road, Jinan, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Key Laboratory of Reproductive Endocrinology, Shandong University, Ministry of Education, 157 Jingliu Road, Jinan, China.,Shandong Provincial Key Laboratory of Reproductive Medicine, 157 Jingliu Road, Jinan, China
| | - Qian Yu
- School of Medicine, Shandong University, 44 Wenhua West Road, Jinan, China.,Reproductive Hospital Affiliated to Shandong University, 157 Jingliu Road, Jinan, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Key Laboratory of Reproductive Endocrinology, Shandong University, Ministry of Education, 157 Jingliu Road, Jinan, China.,Shandong Provincial Key Laboratory of Reproductive Medicine, 157 Jingliu Road, Jinan, China
| | - Yanan Zhang
- School of Medicine, Shandong University, 44 Wenhua West Road, Jinan, China.,Reproductive Hospital Affiliated to Shandong University, 157 Jingliu Road, Jinan, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Key Laboratory of Reproductive Endocrinology, Shandong University, Ministry of Education, 157 Jingliu Road, Jinan, China.,Shandong Provincial Key Laboratory of Reproductive Medicine, 157 Jingliu Road, Jinan, China
| | - Lei Yan
- School of Medicine, Shandong University, 44 Wenhua West Road, Jinan, China.,Reproductive Hospital Affiliated to Shandong University, 157 Jingliu Road, Jinan, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Key Laboratory of Reproductive Endocrinology, Shandong University, Ministry of Education, 157 Jingliu Road, Jinan, China.,Shandong Provincial Key Laboratory of Reproductive Medicine, 157 Jingliu Road, Jinan, China
| | - Zi-Jiang Chen
- School of Medicine, Shandong University, 44 Wenhua West Road, Jinan, China.,Reproductive Hospital Affiliated to Shandong University, 157 Jingliu Road, Jinan, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Key Laboratory of Reproductive Endocrinology, Shandong University, Ministry of Education, 157 Jingliu Road, Jinan, China.,Shandong Provincial Key Laboratory of Reproductive Medicine, 157 Jingliu Road, Jinan, China.,Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, 845 Lingshan Road, Shanghai, China
| |
Collapse
|
56
|
Han CL, Liu YP, Guo CJ, Du TT, Jiang Y, Wang KL, Shao XQ, Meng FG, Zhang JG. The lncRNA H19 binding to let-7b promotes hippocampal glial cell activation and epileptic seizures by targeting Stat3 in a rat model of temporal lobe epilepsy. Cell Prolif 2020; 53:e12856. [PMID: 32648622 PMCID: PMC7445408 DOI: 10.1111/cpr.12856] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/27/2020] [Accepted: 05/16/2020] [Indexed: 12/18/2022] Open
Abstract
Objectives Glial cell activation contributes to the inflammatory response and occurrence of epilepsy. Our preliminary study demonstrated that the long non‐coding RNA, H19, promotes hippocampal glial cell activation during epileptogenesis. However, the precise mechanisms underlying this effect remain unclear. Materials and methods H19 and let‐7b were overexpressed or silenced using an adeno‐associated viral vector in vivo. Their expression in a kainic acid‐induced epilepsy model was evaluated by real‐time quantitative PCR, fluorescence in situ hybridization, and cytoplasmic and nuclear RNA isolation. A dual‐luciferase reporter assay was used to evaluate the direct binding of let‐7b to its target genes and H19. Western blot, video camera monitoring and Morris water maze were performed to confirm the role of H19 and let7b on epileptogenesis. Results H19 was increased in rat hippocampus neurons after status epilepticus, which might be due to epileptic seizure‐induced hypoxia. Increased H19 aggravated the epileptic seizures, memory impairment and mossy fibre sprouting of the epileptic rats. H19 could competitively bind to let‐7b to suppress its expression. Overexpression of let‐7b inhibited hippocampal glial cell activation, inflammatory response and epileptic seizures by targeting Stat3. Moreover, overexpressed H19 reversed the inhibitory effect of let‐7b on glial cell activation. Conclusions LncRNA H19 could competitively bind to let‐7b to promote hippocampal glial cell activation and epileptic seizures by targeting Stat3 in a rat model of temporal lobe epilepsy.
Collapse
Affiliation(s)
- Chun-Lei Han
- Department of Functional Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Neurostimulation, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yun-Peng Liu
- Department of Functional Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Neurostimulation, Beijing, China
| | - Chen-Jia Guo
- Department of Pathology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Ting-Ting Du
- Department of Functional Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Neurostimulation, Beijing, China
| | - Ying Jiang
- Department of Functional Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Neurostimulation, Beijing, China
| | - Kai-Liang Wang
- Department of Functional Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Neurostimulation, Beijing, China
| | - Xiao-Qiu Shao
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Fan-Gang Meng
- Department of Functional Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Neurostimulation, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jian-Guo Zhang
- Department of Functional Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Neurostimulation, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
57
|
Role of long non-coding RNAs and MYC interaction in cancer metastasis: A possible target for therapeutic intervention. Toxicol Appl Pharmacol 2020; 399:115056. [PMID: 32445756 DOI: 10.1016/j.taap.2020.115056] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 05/13/2020] [Accepted: 05/16/2020] [Indexed: 01/17/2023]
Abstract
The c-MYC is one of the most commonly discussed oncogenes in almost all cancers. c-MYC, as a proto-oncogene in normal cells, has found to be tightly controlled and regulated, both genetically and epigenetically. Evasion of the controlled checkpoint mechanisms during cancer causes a deregulated expression of c-MYC. Overexpression of c-MYC causes the onset of many hallmarks of cancer. Despite c-MYC being centrally located in several cancers, it is not feasible to target c-MYC in therapeutic resistant cancers. Similarly, long non-coding RNAs (lncRNAs) are deregulated during the genesis and progression of different cancers. LncRNAs contribute to almost 27% human genome and recent findings by tumor genome sequencing revealed many of the lncRNAs loci that are modified, deleted, amplified, and mutated during the different stages of cancer development. Recent studies also reported that multiple lncRNAs regulate c-MYC by different mechanisms and vice versa. Thus, oncogenic lncRNAs and c-MYC interaction are positioned to provide an interesting choice for therapeutic interventions in cancers. In this mini-review, we summarize the recent discoveries and explain how the interaction between oncogenic lncRNAs and c-MYC could be used as a possible target for therapeutic intervention in cancers, especially the therapeutic resistant metastatic cancers.
Collapse
|
58
|
Liu T, Wang X, Zhai J, Wang Q, Zhang B. Long Noncoding RNA UCA1 Facilitates Endometrial Cancer Development by Regulating KLF5 and RXFP1 Gene Expressions. Cancer Biother Radiopharm 2020; 36:521-533. [PMID: 32412793 DOI: 10.1089/cbr.2019.3278] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Objective: Long noncoding RNA urothelial carcinoma associated 1 (UCA1) was found to facilitate endometrial cancer cell metastasis, and high UCA1 expression predicted endometrial cancer development and patients' worsened outcomes. This research aimed to investigate the cancer promoting role and mechanism of UCA1 in endometrial cancer. Materials and Methods: Around 64 endometrioid adenocarcinoma patients' tissue specimens were analyzed by qRT-PCR. Primary endometrial cancer cell culture was established in vitro. UCA1 overexpression or knockdown was executed by adenoviral transduction. Cell proliferation, apoptosis, colony formation, transwell invasion, and epithelia-to-mesenchymal transition of primary endometrial cancer cells were assessed. Interactions among UCA1, microRNAs (miRNAs), and mRNAs were investigated by luciferase reporter assay and argonaute 2 (AGO2)-RNA immunoprecipitation. Nude mouse xenograft assay was used to explore the role of UCA1 in endometrial cancer in vivo. Results: UCA1 was significantly upregulated in endometrial cancer tissues compared to normal tissues. High expression of UCA1 associated with endometrial cancer progression and patients' decreased survival. Overexpressing UCA1 significantly increased the malignancy of primary endometrial cancer cells in vitro, while UCA1 knockdown showed opposite effect. Kruppel-like factor 5 (KLF5) and relaxin like family peptide receptor 1 (RXFP1) were found as two UCA1 co-expressing genes in endometrial cancer. UCA1 increased the malignancy of endometrial cells partially through KLF5, and it increased the relaxin 2-induced endometrial cancer cell metastasis through RXFP1. UCA1 reduced the si-RNA-induced silencing of KLF5 and RXFP1 genes in endometrial cancer cells. MiR-143-3p and miR-1-3p were found to interact with both UCA1 and KLF5 mRNA. In addition, knockdown of UCA1 suppressed tumor growth in endometrial cancer in vivo. Conclusion: UCA1 might facilitate endometrial cancer development by upregulating KLF5 and RXFP1 gene expression by sponging miR-143-3p and miR-1-3p.
Collapse
Affiliation(s)
- Tong Liu
- Department of Obstetrics and Gynecology, Xuzhou Central Hospital, Xuzhou, China
| | - Xia Wang
- Department of Obstetrics and Gynecology, Xuzhou Central Hospital, Xuzhou, China
| | - Jingfang Zhai
- Department of Obstetrics and Gynecology, Xuzhou Central Hospital, Xuzhou, China
| | - Qing Wang
- Department of Obstetrics and Gynecology, Xuzhou Central Hospital, Xuzhou, China
| | - Bei Zhang
- Department of Obstetrics and Gynecology, Xuzhou Central Hospital, Xuzhou, China
| |
Collapse
|
59
|
Guo J, Li Y, Duan H, Yuan L. Metformin Suppresses the Proliferation and Promotes the Apoptosis of Colon Cancer Cells Through Inhibiting the Expression of Long Noncoding RNA-UCA1. Onco Targets Ther 2020; 13:4169-4181. [PMID: 32523353 PMCID: PMC7234977 DOI: 10.2147/ott.s245091] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 04/28/2020] [Indexed: 12/17/2022] Open
Abstract
PURPOSE LncRNA-UCA1 has been proven to facilitate the proliferation and metastasis of colon cancer. Whether metformin inhibits the progression of colon cancer by suppressing lncRNA-UCA1 remains unknown. In this research, we aimed to explore the role of Metformin playing in pathogenesis of colon cancer. MATERIALS AND METHODS Using qRT-PCR, we measured the expression of five tumor-promoting lncRNAs in SW480 and SW620 colon cancer cells. Then, we conducted Western blotting and immunohistochemistry to evaluate the effects of MET or UCA1 knockdown or the combined MET+ UCA1 knockdown on the activities of the PI3K/AKT and ERK pathways in vitro and in tumor tissues obtained from tumor-bearing nude mice. RESULTS The results from CCK-8 assays showed that MET dose-dependently and time-dependently inhibited the viability of the colon cancer cells in vitro. Flow cytometry revealed that MET promoted the apoptosis of the SW480 and SW620 cells. qRT-PCR showed that lncRNA-UCA1 had the highest expression among the five lncRNAs. Suppressing UCA1 expression by siRNA or shRNA could further enhance the metformin-mediated anticancer effects against colon cancer in vitro and in vivo. Metformin decreased the UCA1 expression and further inhibited the proliferation and promoted the apoptosis of the colon cancer cells, which were associated with inactivation of the PI3K/AKT and ERK signaling pathways in vitro and in the tumor tissues obtained from the mice. CONCLUSION These results indicated that metformin has potential anticancer properties and revealed the anticancer mechanisms of metformin against colon cancer via regulating lncRNA-UCA1.
Collapse
Affiliation(s)
- Jianbo Guo
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Yan Li
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, People’s Republic of China
| | - He Duan
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Lu Yuan
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, People’s Republic of China
| |
Collapse
|
60
|
Liang T, Han L, Guo L. Rewired functional regulatory networks among miRNA isoforms (isomiRs) from let-7 and miR-10 gene families in cancer. Comput Struct Biotechnol J 2020; 18:1238-1248. [PMID: 32542110 PMCID: PMC7280754 DOI: 10.1016/j.csbj.2020.05.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 05/05/2020] [Accepted: 05/05/2020] [Indexed: 01/05/2023] Open
Abstract
Classical microRNA (miRNA) has been so far believed as a single sequence, but it indeed contains multiple miRNA isoforms (isomiR) with various sequences and expression patterns. It is not clear whether these diverse isomiRs have potential relationships and whether they contribute to miRNA:mRNA interactions. Here, we aimed to reveal the potential evolutionary and functional relationships of multiple isomiRs based on let-7 and miR-10 gene families that are prone to clustering together on chromosomes. Multiple isomiRs within gene families showed similar functions to their canonical miRNAs, indicating selection of the predominant sequence. IsomiRs containing novel seed regions showed increased/decreased biological function depending on whether they had more/less specific target mRNAs than their annotated seed. Few gene ontology(GO) terms and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways were shared among the target genes of the annotated seeds and the novel seeds. Various let-7 isomiRs with novel seed regions may cause opposing drug responses despite the fact that they are generated from the same miRNA locus and have highly similar sequences. IsomiRs, especially the dominant isomiRs with shifted seeds, may disturb the coding-non-coding RNA regulatory network. These findings provide insight into the multiple isomiRs and isomiR-mediated control of gene expression in the pathogenesis of cancer.
Collapse
Key Words
- ACC, adrenocortical carcinoma
- BLCA, bladder urothelial carcinoma
- BRCA, breast invasive carcinoma
- CESC, cervical squamous cell carcinoma and endocervical adenocarcinoma
- CHOL, cholangiocarcinoma
- COAD, colon adenocarcinoma
- ESCA, esophageal carcinoma
- Function
- GBM, glioblastoma multiforme
- HNSC, head and neck squamous cell carcinoma
- IsomiR
- KICH, kidney chromophobe
- KIRC, kidney renal clear cell carcinoma
- KIRP, kidney renal papillary cell carcinoma
- LAML, acute myeloid leukemia
- LGG, brain Lower grade glioma
- LIHC, liver hepatocellular carcinoma
- LUAD, lung adenocarcinoma
- LUSC, lung squamous cell carcinoma
- Let-7
- MESO, mesothelioma
- MicroRNA (miRNA)
- Network
- OV, ovarian serous cystadenocarcinoma
- PAAD, pancreatic adenocarcinoma
- PCPG, pheochromocytoma and paraganglioma
- PRAD, prostate adenocarcinoma
- READ, rectum adenocarcinoma
- SARC, sarcoma
- SKCM, skin cutaneous melanoma
- STAD, stomach adenocarcinoma
- TGCT, testicular germ cell tumors
- THCA, thyroid carcinoma
- THYM, thymoma
- TSG, tumor suppressor gene
- UCEC, uterine corpus endometrial carcinoma
- UCS, uterine carcinosarcoma
- UVM, uveal melanoma
- miR-10
Collapse
Affiliation(s)
- Tingming Liang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China
| | - Leng Han
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX 77030, USA
| | - Li Guo
- Department of Bioinformatics, Smart Health Big Data Analysis and Location Services Engineering Lab of Jiangsu Province, School of Geographic and Biologic Information, Nanjing University of Posts and Telecommunications, Nanjing, China
| |
Collapse
|
61
|
Cao T, Jiang Y, Li D, Sun X, Zhang Y, Qin L, Tellides G, Taylor HS, Huang Y. H19/TET1 axis promotes TGF-β signaling linked to endothelial-to-mesenchymal transition. FASEB J 2020; 34:8625-8640. [PMID: 32374060 PMCID: PMC7364839 DOI: 10.1096/fj.202000073rrrrr] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 04/16/2020] [Accepted: 04/17/2020] [Indexed: 12/21/2022]
Abstract
While emerging evidence suggests the link between endothelial activation of TGF-β signaling, induction of endothelial-to-mesenchymal transition (EndMT), and cardiovascular disease (CVD), the molecular underpinning of this connection remains enigmatic. Here, we report aberrant expression of H19 lncRNA and TET1 in endothelial cells (ECs) of human atherosclerotic coronary arteries. Using primary human umbilical vein endothelial cells (HUVECs) and aortic endothelial cells (HAoECs) we show that TNF-α, a known risk factor for endothelial dysfunction and CVD, induces H19 expression which in turn activates TGF-β signaling and EndMT via a TET1-dependent epigenetic mechanism. We also show that H19 regulates TET1 expression at the posttranscriptional level. Further, we provide evidence that this H19/TET1-mediated regulation of TGF-β signaling and EndMT occurs in mouse pulmonary microvascular ECs in vivo under hyperglycemic conditions. We propose that endothelial activation of the H19/TET1 axis may play an important role in EndMT and perhaps CVD.
Collapse
Affiliation(s)
- Tiefeng Cao
- Department of Obstetrics, Gynecology, & Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA.,Department of Gynecology and Obstetrics, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Ying Jiang
- Department of Obstetrics, Gynecology, & Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA.,Department of Obstetrics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Da Li
- Department of Obstetrics, Gynecology, & Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA.,Department of Obstetrics and Gynecology, Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaoli Sun
- Department of Obstetrics, Gynecology, & Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA.,Department of Obstetrics and Gynecology, Center of Reproductive Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Yuanyuan Zhang
- Department of Obstetrics, Gynecology, & Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA.,Department of Obstetrics and Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lingfeng Qin
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
| | - George Tellides
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
| | - Hugh S Taylor
- Department of Obstetrics, Gynecology, & Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Yingqun Huang
- Department of Obstetrics, Gynecology, & Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
62
|
Abstract
Introduction: Ovarian carcinoma (OC) is the leading cause of death in women with gynecologic cancers. Most patients are diagnosed at an advanced stage with a low five-year survival rate of 20-30%. Discovering novel biomarkers for early detection and outcome prediction of OC is an urgent medical need. miRNAs, a group of small non-coding RNAs, play critical roles in multiple biologic processes and cancer pathogenesis.Areas covered: We provide an in-depth look at the functions of miRNAs in OC, particularly focusing on their roles in chemoresistance and metastasis in OC. We also discuss the biological and clinical significance of miRNAs in exosomes and expand on long non-coding RNA which acts as ceRNA of miRNAs.Expert opinion: miRNAs participate in many biological processes including proliferation, apoptosis, chemoresistance, metastasis, epithelial-mesenchymal transition, and cancer stem cell. They will substantially contribute to our understanding of OC pathogenesis. Given their resistance to the degradation of ribonucleases and availability in plasma exosomes, miRNAs may serve as emerging biomarkers for cancer detection, therapeutic assessment, and prognostic prediction. Being a messenger, exosomal miRNAs are crucial for the crosstalk between cancer cells and stromal cells in tumor microenvironment.
Collapse
Affiliation(s)
- Huilin Zhang
- Department of Surgical Pathology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Bingjian Lu
- Department of Surgical Pathology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| |
Collapse
|
63
|
He D, Zeng H, Chen J, Xiao L, Zhao Y, Liu N. H19 regulates trophoblastic spheroid adhesion by competitively binding to let-7. Reproduction 2020; 157:423-430. [PMID: 30780128 PMCID: PMC6433002 DOI: 10.1530/rep-18-0339] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 02/18/2019] [Indexed: 12/22/2022]
Abstract
Integrin β3 (ITGB3), which is the target gene of the miRNA let-7 that can be antagonized by long noncoding RNA (lncRNA) H19, is well known to have a critical role in endometrium receptivity. However, the regulation of ITGB3 in cell–cell or cell–extracellular matrix adhesion and invasion for the maintenance of early pregnancy remains unknown. This study aimed to explore the role of the H19/let-7/ITGB3 axis in regulating trophoblastic spheroid adhesion and in vitro invasion ability using the HTR-8/SVneo cell line and to investigate the expression levels of lncRNA H19 and ITGB3 in human products of conception. The in vitro knockdown of H19 resulted in decreased expression of ITGB3 at the mRNA and protein levels and reduced the adhesion and invasion ability. In the embryonic chorion tissue of spontaneous abortion (SA), the expressions of H19 and ITGB3 at both the mRNA and protein levels decreased. The results of quantitative RT-PCR, Western blot analysis, dual-luciferase report gene and functional miRNA let-7 rescue experiments, adhesion assay and in vitro transwell invasion assay confirmed that H19 regulated trophoblastic spheroid adhesion with endometrial stromal cells through the H19/let-7/ITGB3 axis, thereby providing an improved understanding of the molecular mechanism of SA.
Collapse
Affiliation(s)
- Dongmei He
- Reproductive Medicine Center, Xiangya Hospital, Central South University, Changsha, China
| | - Hong Zeng
- Reproductive Medicine Center, Xiangya Hospital, Central South University, Changsha, China
| | - Jingfei Chen
- Department of Obstetrics and Gynecology, Xiangya Hospital, Central South University, Changsha, China
| | - Lan Xiao
- Reproductive Medicine Center, Xiangya Hospital, Central South University, Changsha, China
| | - Yuhao Zhao
- Reproductive Medicine Center, Xiangya Hospital, Central South University, Changsha, China
| | - Nenghui Liu
- Reproductive Medicine Center, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
64
|
Zhao B, Luo J, Yu T, Zhou L, Lv H, Shang P. Anticancer mechanisms of metformin: A review of the current evidence. Life Sci 2020; 254:117717. [PMID: 32339541 DOI: 10.1016/j.lfs.2020.117717] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 04/15/2020] [Accepted: 04/21/2020] [Indexed: 02/07/2023]
Abstract
Metformin, a US Food and Drug Administration-approved "star" drug used for diabetes mellitus type 2, has become a topic of increasing interest to researchers due to its anti-neoplastic effects. Growing evidence has demonstrated that metformin may be a promising chemotherapeutic agent, and several clinical trials of metformin use in cancer treatment are ongoing. However, the anti-neoplastic effects of metformin and its underlying mechanisms have not been fully elucidated. In this review, we present the newest findings on the anticancer activities of metformin, and highlight its diverse anticancer mechanisms. Several clinical trials, as well as the limitations of the current evidence are also demonstrated. This review explores the crucial roles of metformin and provides supporting evidence for the repurposing of metformin as a treatment of cancer.
Collapse
Affiliation(s)
- Bin Zhao
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen 518057, China; School of Life Science, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China; Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Science, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Jie Luo
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen 518057, China; School of Life Science, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China; Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Science, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Tongyao Yu
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen 518057, China; School of Life Science, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China; Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Science, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Liangfu Zhou
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen 518057, China; School of Life Science, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China; Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Science, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Huanhuan Lv
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen 518057, China; School of Life Science, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China; Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Science, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Peng Shang
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen 518057, China; Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Science, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China.
| |
Collapse
|
65
|
Verjans R, van Bilsen M, Schroen B. Reviewing the Limitations of Adult Mammalian Cardiac Regeneration: Noncoding RNAs as Regulators of Cardiomyogenesis. Biomolecules 2020; 10:biom10020262. [PMID: 32050588 PMCID: PMC7072544 DOI: 10.3390/biom10020262] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 02/03/2020] [Accepted: 02/07/2020] [Indexed: 12/18/2022] Open
Abstract
The adult mammalian heart is incapable of regeneration following cardiac injury, leading to a decline in function and eventually heart failure. One of the most evident barriers limiting cardiac regeneration is the inability of cardiomyocytes to divide. It has recently become clear that the mammalian heart undergoes limited cardiomyocyte self-renewal throughout life and is even capable of modest regeneration early after birth. These exciting findings have awakened the goal to promote cardiomyogenesis of the human heart to repair cardiac injury or treat heart failure. We are still far from understanding why adult mammalian cardiomyocytes possess only a limited capacity to proliferate. Identifying the key regulators may help to progress towards such revolutionary therapy. Specific noncoding RNAs control cardiomyocyte division, including well explored microRNAs and more recently emerged long noncoding RNAs. Elucidating their function and molecular mechanisms during cardiomyogenesis is a prerequisite to advance towards therapeutic options for cardiac regeneration. In this review, we present an overview of the molecular basis of cardiac regeneration and describe current evidence implicating microRNAs and long noncoding RNAs in this process. Current limitations and future opportunities regarding how these regulatory mechanisms can be harnessed to study myocardial regeneration will be addressed.
Collapse
Affiliation(s)
- Robin Verjans
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6200 MD Maastricht, The Netherlands;
| | - Marc van Bilsen
- Department of Physiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6200 MD Maastricht, The Netherlands;
| | - Blanche Schroen
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6200 MD Maastricht, The Netherlands;
- Correspondence: ; Tel.: +31-433882949
| |
Collapse
|
66
|
Correlation between polymorphisms in IGF2/H19 gene locus and epithelial ovarian cancer risk in Chinese population. Genomics 2020; 112:2510-2515. [PMID: 32045670 DOI: 10.1016/j.ygeno.2020.02.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 12/29/2019] [Accepted: 02/07/2020] [Indexed: 01/14/2023]
Abstract
To investigate the association between SNPs in human IGF2/H19 gene locus and epithelial ovarian cancer (EOC) risk, we performed a case-control study in 422 individuals (219 EOC patients and 203 cancer-free controls). Four SNPs (rs2525885, rs2839698, rs3741206, rs3741219) were found to be related with EOC risk. Specifically, the minor allele C of rs2525885 and allele A of rs2839698 was associated with elevated EOC genetic susceptibility under both dominant and recessive models (TC + CC vs TT: adjusted OR: 1.61, P = .031; CC vs TT + TC: adjusted OR: 4.87, P = .014; GA + AA vs GG: adjusted OR: 1.63, P = .023; AA vs GG + GA: adjusted OR: 2.43, P = .007). For rs3741206, the genotype TC + CC was associated with a significant decrease in EOC risk with the TT genotype as reference in a dominant genetic model (adjusted OR: 0.44, P = .003), while for rs3741219, genotype AA was associated with a 59% decrease in EOC risk only in the recessive model (adjusted OR: 0.41, P = .038). In the stratified analysis, an increased risk associated with the variant genotypes was observed in only subjects aged >47 years for rs2525885 (adjusted OR = 2.04, P = .024), rs2839698 (adjusted OR = 2.50, P = .047) and rs3741206 (adjusted OR = 0.37, P = .009), respectively. What's more, the TC + CC genotype of rs2525885 was significantly associated with advanced FIGO stage (III vs II, adjusted OR = 2.73, P = .040).
Collapse
|
67
|
You G, Long X, Song F, Huang J, Tian M, Xiao Y, Deng S, Wu Q. Metformin Activates the AMPK-mTOR Pathway by Modulating lncRNA TUG1 to Induce Autophagy and Inhibit Atherosclerosis. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:457-468. [PMID: 32099330 PMCID: PMC7006854 DOI: 10.2147/dddt.s233932] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 01/11/2020] [Indexed: 12/25/2022]
Abstract
Background Metformin has been shown to inhibit the proliferation and migration of vascular wall cells. However, the mechanism through which metformin acts on atherosclerosis (AS) via the long non-coding RNA taurine up-regulated gene 1 (lncRNA TUG1) is still unknown. Thus, this research investigated the effect of metformin and lncRNA TUG1 on AS. Methods First, qRT-PCR was used to detect the expression of lncRNA TUG1 in patients with coronary heart disease (CHD). Then, the correlation between metformin and TUG1 expression in vitro and their effects on proliferation, migration, and autophagy in vascular wall cells were examined. Furthermore, in vivo experiments were performed to verify the anti-AS effect of metformin and TUG1 to provide a new strategy for the prevention and treatment of AS. Results qRT-PCR results suggested that lncRNA TUG1 expression was robustly upregulated in patients with CHD. In vitro experiments indicated that after metformin administration, the expression of lncRNA TUG1 decreased in a time-dependent manner. Metformin and TUG1 knockdown via small interfering RNA both inhibited proliferation and migration while promoted autophagy via the AMPK/mTOR pathway in vascular wall cells. In vivo experiments with a rat AS model further demonstrated that metformin and sh-TUG1 could inhibit the progression of AS. Conclusion Taken together, our data demonstrate that metformin might function to prevent AS by activating the AMPK/mTOR pathway via lncRNA TUG1.
Collapse
Affiliation(s)
- Ganhua You
- Guizhou University School of Medicine, Guiyang 550025, People's Republic of China.,Guizhou Institute for Food and Drug Control, Guiyang 550004, People's Republic of China
| | - Xiangshu Long
- Department of Cardiology, Guizhou Provincial People's Hospital, Guiyang 550002, People's Republic of China.,Department of Cardiology, People's Hospital of Guizhou University, Guiyang 550002, People's Republic of China
| | - Fang Song
- Department of Cardiology, Guizhou Provincial People's Hospital, Guiyang 550002, People's Republic of China.,Department of Cardiology, People's Hospital of Guizhou University, Guiyang 550002, People's Republic of China
| | - Jing Huang
- Department of Cardiology, Guizhou Provincial People's Hospital, Guiyang 550002, People's Republic of China.,Department of Cardiology, People's Hospital of Guizhou University, Guiyang 550002, People's Republic of China
| | - Maobo Tian
- Department of Cardiology, Guizhou Provincial People's Hospital, Guiyang 550002, People's Republic of China.,Department of Cardiology, People's Hospital of Guizhou University, Guiyang 550002, People's Republic of China
| | - Yan Xiao
- Department of Cardiology, Guizhou Provincial People's Hospital, Guiyang 550002, People's Republic of China.,Department of Cardiology, People's Hospital of Guizhou University, Guiyang 550002, People's Republic of China
| | - Shiyan Deng
- Department of Cardiology, Guizhou Provincial People's Hospital, Guiyang 550002, People's Republic of China.,Department of Cardiology, People's Hospital of Guizhou University, Guiyang 550002, People's Republic of China
| | - Qiang Wu
- Department of Cardiology, Guizhou Provincial People's Hospital, Guiyang 550002, People's Republic of China.,Department of Cardiology, People's Hospital of Guizhou University, Guiyang 550002, People's Republic of China
| |
Collapse
|
68
|
High Mobility Group A (HMGA): Chromatin Nodes Controlled by a Knotty miRNA Network. Int J Mol Sci 2020; 21:ijms21030717. [PMID: 31979076 PMCID: PMC7038092 DOI: 10.3390/ijms21030717] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 01/16/2020] [Accepted: 01/17/2020] [Indexed: 12/11/2022] Open
Abstract
High mobility group A (HMGA) proteins are oncofoetal chromatin architectural factors that are widely involved in regulating gene expression. These proteins are unique, because they are highly expressed in embryonic and cancer cells, where they play a relevant role in cell proliferation, stemness, and the acquisition of aggressive tumour traits, i.e., motility, invasiveness, and metastatic properties. The HMGA protein expression levels and activities are controlled by a connected set of events at the transcriptional, post-transcriptional, and post-translational levels. In fact, microRNA (miRNA)-mediated RNA stability is the most-studied mechanism of HMGA protein expression modulation. In this review, we contribute to a comprehensive overview of HMGA-targeting miRNAs; we provide detailed information regarding HMGA gene structural organization and a comprehensive evaluation and description of HMGA-targeting miRNAs, while focusing on those that are widely involved in HMGA regulation; and, we aim to offer insights into HMGA-miRNA mutual cross-talk from a functional and cancer-related perspective, highlighting possible clinical implications.
Collapse
|
69
|
Yu TX, Chung HK, Xiao L, Piao JJ, Lan S, Jaladanki SK, Turner DJ, Raufman JP, Gorospe M, Wang JY. Long Noncoding RNA H19 Impairs the Intestinal Barrier by Suppressing Autophagy and Lowering Paneth and Goblet Cell Function. Cell Mol Gastroenterol Hepatol 2019; 9:611-625. [PMID: 31862317 PMCID: PMC7078540 DOI: 10.1016/j.jcmgh.2019.12.002] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 12/09/2019] [Accepted: 12/10/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS The protective intestinal mucosal barrier consists of multiple elements including mucus and epithelial layers and immune defense; nonetheless, barrier dysfunction is common in various disorders. The imprinted and developmentally regulated long noncoding RNA H19 is involved in many cell processes and diseases. Here, we investigated the role of H19 in regulating Paneth and goblet cells and autophagy, and its impact on intestinal barrier dysfunction induced by septic stress. METHODS Studies were conducted in H19-deficient (H19-/-) mice, mucosal tissues from patients with sepsis, primary enterocytes, and Caco-2 cells. Septic stress was induced by cecal ligation and puncture (CLP), and gut permeability was detected by tracer fluorescein isothiocyanate-dextran assays. The function of Paneth and goblet cells was examined by immunostaining for lysozyme and mucin 2, respectively, and autophagy was examined by microtubule-associated proteins 1A/1B light chain 3 II immunostaining and Western blot analysis. Intestinal organoids were isolated from H19-/- and control littermate mice and treated with lipopolysaccharide (LPS). RESULTS Intestinal mucosal tissues in mice 24 hours after exposure to CLP and in patients with sepsis showed high H19 levels, associated with intestinal barrier dysfunction. Targeted deletion of the H19 gene in mice enhanced the function of Paneth and goblet cells and promoted autophagy in the small intestinal mucosa. Knockout of H19 protected Paneth and goblet cells against septic stress, preserved autophagy activation, and promoted gut barrier function after exposure to CLP. Compared with organoids from control littermate mice, intestinal organoids isolated from H19-/- mice had increased numbers of lysozyme- and mucin 2-positive cells and showed increased tolerance to LPS. Conversely, ectopic overexpression of H19 in cultured intestinal epithelial cells prevented rapamycin-induced autophagy and abolished the rapamycin-induced protection of the epithelial barrier against LPS. CONCLUSIONS In investigations of mice, human tissues, primary organoids, and intestinal epithelial cells, we found that increased H19 inhibited the function of Paneth and goblet cells and suppressed autophagy, thus potentially contributing to barrier dysfunction in intestinal pathologies.
Collapse
Affiliation(s)
- Ting-Xi Yu
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland
| | - Hee K Chung
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland
| | - Lan Xiao
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland; Baltimore Veterans Affairs Medical Center, Baltimore, Maryland
| | - Jun-Jie Piao
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland
| | - Shaoyang Lan
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland
| | - Suraj K Jaladanki
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland
| | - Douglas J Turner
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland; Baltimore Veterans Affairs Medical Center, Baltimore, Maryland
| | - Jean-Pierre Raufman
- Baltimore Veterans Affairs Medical Center, Baltimore, Maryland; Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Myriam Gorospe
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, Maryland
| | - Jian-Ying Wang
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland; Baltimore Veterans Affairs Medical Center, Baltimore, Maryland; Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland.
| |
Collapse
|
70
|
Mishra R, Haldar S, Suchanti S, Bhowmick NA. Epigenetic changes in fibroblasts drive cancer metabolism and differentiation. Endocr Relat Cancer 2019; 26:R673-R688. [PMID: 31627186 PMCID: PMC6859444 DOI: 10.1530/erc-19-0347] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 10/17/2019] [Indexed: 12/17/2022]
Abstract
Genomic changes that drive cancer initiation and progression contribute to the co-evolution of the adjacent stroma. The nature of the stromal reprogramming involves differential DNA methylation patterns and levels that change in response to the tumor and systemic therapeutic intervention. Epigenetic reprogramming in carcinoma-associated fibroblasts are robust biomarkers for cancer progression and have a transcriptional impact that support cancer epithelial progression in a paracrine manner. For prostate cancer, promoter hypermethylation and silencing of the RasGAP, RASAL3 that resulted in the activation of Ras signaling in carcinoma-associated fibroblasts. Stromal Ras activity initiated a process of macropinocytosis that provided prostate cancer epithelia with abundant glutamine for metabolic conversion to fuel its proliferation and a signal to transdifferentiate into a neuroendocrine phenotype. This epigenetic oncogenic metabolic/signaling axis seemed to be further potentiated by androgen receptor signaling antagonists and contributed to therapeutic resistance. Intervention of stromal signaling may complement conventional therapies targeting the cancer cell.
Collapse
Affiliation(s)
- Rajeev Mishra
- Department of Biosciences, Manipal University Jaipur, Jaipur, Rajasthan, India
| | - Subhash Haldar
- Department of Biotechnology, Brainware University, Kolkata, India
| | - Surabhi Suchanti
- Department of Biosciences, Manipal University Jaipur, Jaipur, Rajasthan, India
| | - Neil A Bhowmick
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Research, Greater Los Angeles Veterans Administration, Los Angeles, California, USA
- Correspondence should be addressed to N A Bhowmick:
| |
Collapse
|
71
|
Wang J, Sun J, Yang F. The role of long non-coding RNA H19 in breast cancer. Oncol Lett 2019; 19:7-16. [PMID: 31897110 PMCID: PMC6924119 DOI: 10.3892/ol.2019.11093] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 07/26/2019] [Indexed: 12/24/2022] Open
Abstract
Breast cancer is the most common malignant tumor in women in the majority of countries, such as China, Britain and Australia, and its morbidity and mortality rates remain very high. Long non-coding RNAs (lncRNAs) are non-coding RNAs (ncRNAs) >200 nucleotides in length that lack open reading frames. LncRNA H19 is a transcription product of the H19 gene, and the aberrant expression of H19 can be demonstrated in various types of tumor cell. The purpose of the present review was to elaborate the role of H19 in breast cancer. H19 can regulate gene expression in breast cancer at multiple levels, including epigenetic, transcriptional and posttranscriptional. The abnormal expression of H19 is closely associated with the tumorigenesis and progression of breast cancer via different underlying molecular mechanisms, such as encoding microRNA-675, competing endogenous RNA regulation and interacting with MYC. A large number of clinical studies have suggested that H19 can serve as a potential biomarker for the diagnosis of breast cancer. High expression levels of H19 increases the drug resistance of breast cancer cells and is associated with poor prognosis within patients with breast cancer. Therefore, serum H19 levels may have momentous significance in the clinical setting.
Collapse
Affiliation(s)
- Ji Wang
- The First Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Jinyu Sun
- The First Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Fen Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| |
Collapse
|
72
|
lncRNA SNHG5 Modulates Endometrial Cancer Progression via the miR-25-3p/BTG2 Axis. JOURNAL OF ONCOLOGY 2019; 2019:7024675. [PMID: 31885582 PMCID: PMC6925730 DOI: 10.1155/2019/7024675] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 07/01/2019] [Accepted: 07/18/2019] [Indexed: 01/11/2023]
Abstract
Endometrial carcinoma (EC) is one of the most common malignancies of the female genital tract, although the mechanisms of EC initiation and development remain incompletely understood. In this study, we demonstrated that the noncoding RNA SNHG5 can inhibit the proliferation, migration, and invasion of EC cells by suppressing the expression of its putative target miR-25-3p. Overexpression of miR-25-3p significantly promoted the proliferation, migration, and invasion of EC cells. In addition, we showed that miR-25-3p represses the expression of BTG2 by directly binding to the 3′-UTR of BTG2 mRNA. Furthermore, increased miR-25-3p expression and decreased SNHG5 and BTG2 expression were observed in EC tissues, and the expression of SNHG5 was negatively correlated to that of miR-25-3p but positively correlated to that of BTG2. In summary, for the first time, we report that the SNHG5/miR-25-3p/BTG2 axis plays an important role in EC progression and is of great potential clinical significance for EC diagnosis and therapy.
Collapse
|
73
|
Andreeva–Gateva PA, Mihaleva ID, Dimova II. Type 2 diabetes mellitus and cardiovascular risk; what the pharmacotherapy can change through the epigenetics. Postgrad Med 2019; 132:109-125. [DOI: 10.1080/00325481.2019.1681215] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Pavlina A. Andreeva–Gateva
- Department of Pharmacology and Toxicology, Faculty of Medicine, Medical University of Sofia, Sofia, Bulgaria
- Department of Pharmacology, Medical Faculty, Sofia University “St Kliment Ohridski”, Sofia, Bulgaria
| | - Ivelina D. Mihaleva
- Department of Pharmacology and Toxicology, Faculty of Medicine, Medical University of Sofia, Sofia, Bulgaria
| | - Ivanka I. Dimova
- Department of Medical Genetics, Faculty of Medicine, Medical University of Sofia, Sofia, Bulgaria
| |
Collapse
|
74
|
Zhao JF, Zha ZA, Xie WH, Wang HB, Li XM, Sun Q, Sun ML. [Effect of long chain non-coding RNA H19 on the migration and invasion of oral cancer cells and its molecular mechanism]. HUA XI KOU QIANG YI XUE ZA ZHI = HUAXI KOUQIANG YIXUE ZAZHI = WEST CHINA JOURNAL OF STOMATOLOGY 2019; 37:378-383. [PMID: 31512829 DOI: 10.7518/hxkq.2019.04.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To investigate the effect of the long chain non-coding RNA H19 (lncRNA H19) on the invasion and migration of oral cancer cells and its related molecular mechanism. METHODS The expression levels of lncRNA H19, miR-107, and cyclin-dependent kinase 6 (CDK6) in the immortalized oral epithelial cell line HIOEC and the oral cancer cell line CAL27 were detected by real-time quantitative polymerase chain reaction. CAL27 cells were transfected with siRNA H19, miR-107 mimics, pcDNA H19, or anti-miR-107, and the effects of H19 and miR-107 on the invasion and migration of cells were examined via Transwell assay. The TargetScan database predicted the targeting of H19, miR-107, and CDK6. Double luciferase reporter gene assay was performed to detect interactions among H19, miR-107, and CDK6. Western blot analysis was conducted to examine the effects of H19 and miR-107 on the protein level of the target gene CDK6. RESULTS Compared with that in HIOEC cells, the expression of H19 was significantly increased in CAL27 cells (P<0.05). After transfection with siRNA H19, the expression of H19 decreased, and the invasion and migration ability of CAL27 cells were inhibited (P<0.05). H19 could bind specifically to the 3'-UTR of miR-107 to modulate the expression of miR-107. Compared with that in HIOEC cells, the expression of miR-107 significantly decreased in CAL27 cells (P<0.05). The expression of miR-107 increased after transfection with siRNA H19, and anti-mir-107 co-transfection could promote the invasion and migration ability of siRNA H19 in CAL27 cells (P<0.05). Compared with that in HIOEC cells, CDK6 expression significantly increased in CAL27 cells (P<0.05), and the expression level of the gene was coregulated by H19 and miR-107 (P<0.05). CONCLUSIONS lncRNA H19 plays an important role in the development of oral cancer. It can regulate the invasion and migration of oral cancer cells by targeting the miR-107/CDK6 signaling axis.
Collapse
Affiliation(s)
- Jun-Fang Zhao
- Dept. of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Zhi-An Zha
- Dept. of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Wei-Hong Xie
- Dept. of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Hai-Bin Wang
- Dept. of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Xin-Ming Li
- Dept. of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Qiang Sun
- Dept. of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Ming-Lei Sun
- Dept. of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
75
|
Huang MC, Chou YH, Shen HP, Ng SC, Lee YC, Sun YH, Hsu CF, Yang SF, Wang PH. The clinicopathological characteristic associations of long non-coding RNA gene H19 polymorphisms with uterine cervical cancer. J Cancer 2019; 10:6191-6198. [PMID: 31772651 PMCID: PMC6856740 DOI: 10.7150/jca.36707] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 09/18/2019] [Indexed: 12/14/2022] Open
Abstract
The purposes of the current study were conducted to explore the relationships among long non-coding RNA gene H19 (LncRNA H19) polymorphisms and clinicopathological characteristics of uterine cervical cancer, and patient prognosis in Taiwan. Five genetic variants of LncRNA H19 rs3024270, rs2839698, rs3741219, rs2107425 and rs217727 were recruited from one hundred and thirty-four patients with invasive cancer, 101 with high-grade cervical intraepithelial neoplasia (CIN) of uterine cervix and 325 controls and their genetic distributions were determined. It indicated no associations of these LncRNA H19 genetic variants with development of cervical cancer. CC/CT in LncRNA H19 rs2839698 exhibited less risk to have pelvic lymph node metastasis [Odds ratio (OR): 0.19, 95% Confidence interval (CI):0.04-0.82, p=0.028)], as compared with TT. Meanwhile, cervical cancer patients with AA/AG in rs3741219 also had less risk to develop pelvic lymph node metastasis (OR: 0.17, 95% CI: 0.05-0.63, p=0.008), large tumor (OR: 0.17, 95% CI: 0.04-0.82, p=0.014) as well as parametrium (OR: 0.26, 95% CI: 0.07-0.95, p=0.045) and vagina invasion (OR: 0.25, 95% CI: 0.07-0.91, p=0.041, as compared to those with GG. However, only positive pelvic lymph node metastasis was related to worse recurrence-free survival and poor overall survival. Conclusively, it indicated no association of LncRNA H19 SNPs with cervical carcinogensis in Taiwanese women. Although genotypes TT in LncRNA H19 rs2839698 and GG in rs3741219 are related to some poor clinicopathological parameters of cervical cancer, only pelvic lymph node status could predict 5 year patient survival significantly.
Collapse
Affiliation(s)
- Ming-Chao Huang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Obstetrics and Gynecology, Hsinchu MacKay Memorial Hospital, Hsinchu, Taiwan.,Mackay Medicine, Nursing, and Management College, Taipei, Taiwan
| | - Ying-Hsiang Chou
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Medical Imaging and Radiological Sciences, Chung Shan Medical University, Taichung, Taiwan.,Department of Radiation Oncology, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Huang-Pin Shen
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Obstetrics and Gynecology, Chung Shan Medical University Hospital, Taichung, Taiwan.,School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Soo-Cheen Ng
- Department of Obstetrics and Gynecology, Chung Shan Medical University Hospital, Taichung, Taiwan.,School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Yueh-Chun Lee
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Radiation Oncology, Chung Shan Medical University Hospital, Taichung, Taiwan.,School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Yi-Hung Sun
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Obstetrics and Gynecology, Chi-Mei Foundation Medical Center, Tainan, Taiwan
| | - Chun-Fang Hsu
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Obstetrics and Gynecology, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Po-Hui Wang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Obstetrics and Gynecology, Chung Shan Medical University Hospital, Taichung, Taiwan.,School of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
76
|
Liu H, Wan J, Chu J. Long non-coding RNAs and endometrial cancer. Biomed Pharmacother 2019; 119:109396. [PMID: 31505425 DOI: 10.1016/j.biopha.2019.109396] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 08/11/2019] [Accepted: 08/28/2019] [Indexed: 12/17/2022] Open
Abstract
Endometrial cancer (EC) is one of the most common gynecologic malignancies. In spite of the advance in chemotherapy, radiotherapy or surgical techniques for EC in recent years, the survival rate of advanced stage EC patients remains unsatisfactory. Long non-coding RNAs (lncRNAs) are known as transcripts longer than 200 nucleotides exhibiting no or limited protein-coding potential. Growing evidence suggested lncRNAs may be a critical class of pervasive genes involved in cancer progression. However, the function and biological relevance between lncRNAs and EC remain not yet fully understood. Accumulating evidence has indicated that lncRNAs are dysregulated in EC, and closely related to tumorigenesis, metastasis and chemoresistance. In this review, we summarize the known regulation and functional roles of lncRNAs in EC. Besides, we will discuss the potential of lncRNAs as diagnostic biomarkers and therapeutic targets in EC.
Collapse
Affiliation(s)
- Hongyang Liu
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China.
| | - Junhu Wan
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Jie Chu
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| |
Collapse
|
77
|
Zhao Z, Wen J, Peng L, Liu H. Upregulation of Insulin-Like Growth Factor-1 Receptor (IGF-1R) Reverses the Inhibitory Effect of Let-7g-5p on Migration and Invasion of Nasopharyngeal Carcinoma. Med Sci Monit 2019; 25:5747-5756. [PMID: 31374070 PMCID: PMC6689202 DOI: 10.12659/msm.914555] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background Let-7 microRNAs (miRNAs) have the effects of inhibiting tumor growth and metastasis, however, the research in nasopharyngeal carcinoma (NPC) is limited. This study focused on the effects of Let-7 on NPC migration and invasion and the mechanism of action. Material/Methods Plasmid transfection was used to upregulate the expression levels of Let-7g-5p and insulin-like growth factor-1 receptor (IGF-1R). Cell counting kit-8 (CCK-8) assay was applied to test the cell viability. Scratch assay and Transwell assay were performed to detect the migration and invasion abilities. Bioinformatics prediction and luciferase reporter assay were used to determine and verify the downstream target genes for Let-7g-5p. Protein and mRNA were detected by western blot and real-time quantitative polymerase chain reaction (RT-qPCR), respectively. Results Let-7g-5p was under-expressed in human NPC cells. Overexpression of Let-7g-5p could inhibit cell viability and inhibit the migration and invasion of SUNE1 cells. The dual-luciferase reporter assay showed that IGF-1R was a direct target gene of Let-7g-5p, which was directly regulated IGF-1R expression by 3′UTR. Let-7g-5p overexpression could inhibit the expression of IGF-1R gene, and upregulation of IGF-1R gene expression reversed the inhibitory effect of Let-7g-5p on cell viability and epithelial-mesenchymal transition processes. Conclusions Let-7g-5p is lowly expressed in NPC and it was the first to discover that IGF-1R was a target gene of let-7g-5p in NPC. Upregulation of IGF-1R reversed the inhibitory effect of Let-7g-5p on epithelial-mesenchymal transition.
Collapse
Affiliation(s)
- Zhecheng Zhao
- Department of Otolaryngology Head and Neck Surgery, Zhuzhou Central Hospital, Zhuzhou, Hunan, China (mainland)
| | - Jianxue Wen
- Department of Otolaryngology Head and Neck Surgery, Zhuzhou Central Hospital, Zhuzhou, Hunan, China (mainland)
| | - Lihua Peng
- Department of Otolaryngology Head and Neck Surgery, Zhuzhou Central Hospital, Zhuzhou, Hunan, China (mainland)
| | - Hanbo Liu
- Department of Otolaryngology Head and Neck Surgery, Zhuzhou Central Hospital, Zhuzhou, Hunan, China (mainland)
| |
Collapse
|
78
|
Wang J, Xie S, Yang J, Xiong H, Jia Y, Zhou Y, Chen Y, Ying X, Chen C, Ye C, Wang L, Zhou J. The long noncoding RNA H19 promotes tamoxifen resistance in breast cancer via autophagy. J Hematol Oncol 2019; 12:81. [PMID: 31340867 PMCID: PMC6657081 DOI: 10.1186/s13045-019-0747-0] [Citation(s) in RCA: 179] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 05/31/2019] [Indexed: 12/16/2022] Open
Abstract
Background Tamoxifen resistance remains a clinical challenge for hormone receptor-positive breast cancer. Recently, dysregulations in autophagy have been suggested as a potential mechanism for tamoxifen resistance. Although the long noncoding RNA H19 is involved in various stages of tumorigenesis, its role in tamoxifen resistance remains unknown. Here, we assessed the role of H19 in the development of tamoxifen-resistant breast cancer. Methods Quantitative real-time PCR analyzed expression of H19 in tamoxifen-resistant breast cancer tissues. Knockdown of H19 was used to assess the sensitivity to tamoxifen in vitro and in vivo. Both knockdown and overexpression of H19 were used to analyze the status of autophagy. Real-time quantitative methylation-specific polymerase chain reaction, chromatin immunoprecipitation, immunofluorescence, and Western blot were used to explore the tamoxifen resistance mechanism of H19. Results In this study, we observed that the expression of H19 was substantially upregulated in tamoxifen-resistant breast cancer cell line and tumor tissues, and knockdown of H19 enhanced the sensitivity to tamoxifen both in vitro and in vivo. Furthermore, knockdown of H19 significantly inhibited autophagy in MCF7 tamoxifen-resistant (MCF7/TAMR) cells. Conversely, overexpression of H19 promoted autophagy. Interestingly, overexpression of H19 in MCF7 tamoxifen-sensitive cells could recapitulate tamoxifen resistance. Moreover, an increase in methylation in the promoter region of Beclin1 was observed in MCF7/TAMR-shH19 cells. In the double knockdown groups, both shH19+shSAHH and shH19+shDNMT3B rescued the Beclin1 promoter region methylation levels and reactivated autophagy functions. A chromatin immunoprecipitation assay further validated that DNMT3B binds to the Beclin1 promoter region and the knockdown of H19 increases this binding. Conclusions Our findings demonstrate that H19 induces autophagy activation via the H19/SAHH/DNMT3B axis, which could contribute to tamoxifen resistance in breast cancer. Electronic supplementary material The online version of this article (10.1186/s13045-019-0747-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ji Wang
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No.3 Eastern Qingchun Road, Hangzhou, 310016, Zhejiang, China.,Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China
| | - Shuduo Xie
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No.3 Eastern Qingchun Road, Hangzhou, 310016, Zhejiang, China.,Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China
| | - Jingjing Yang
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No.3 Eastern Qingchun Road, Hangzhou, 310016, Zhejiang, China.,Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China
| | - Hanchu Xiong
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No.3 Eastern Qingchun Road, Hangzhou, 310016, Zhejiang, China.,Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China
| | - Yunlu Jia
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No.3 Eastern Qingchun Road, Hangzhou, 310016, Zhejiang, China.,Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China
| | - Yulu Zhou
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No.3 Eastern Qingchun Road, Hangzhou, 310016, Zhejiang, China.,Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China
| | - Yongxia Chen
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No.3 Eastern Qingchun Road, Hangzhou, 310016, Zhejiang, China.,Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China
| | - Xiaogang Ying
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No.3 Eastern Qingchun Road, Hangzhou, 310016, Zhejiang, China.,Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China
| | - Cong Chen
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No.3 Eastern Qingchun Road, Hangzhou, 310016, Zhejiang, China.,Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China
| | - Chenyang Ye
- Cancer Institute (Key Laboratory of Cancer Prevention & Intervention, National Ministry of Education), Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Linbo Wang
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No.3 Eastern Qingchun Road, Hangzhou, 310016, Zhejiang, China. .,Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China.
| | - Jichun Zhou
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No.3 Eastern Qingchun Road, Hangzhou, 310016, Zhejiang, China. .,Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China.
| |
Collapse
|
79
|
Shu C, Yan D, Chen C, Mo Y, Wu L, Gu J, Shah NK, He J, Dong S. Metformin exhibits its therapeutic effect in the treatment of pre-eclampsia via modulating the Met/H19/miR-148a-5p/P28 and Met/H19/miR-216-3p/EBI3 signaling pathways. Int Immunopharmacol 2019; 74:105693. [PMID: 31203154 DOI: 10.1016/j.intimp.2019.105693] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 05/16/2019] [Accepted: 06/08/2019] [Indexed: 11/25/2022]
Abstract
Metformin (Met) has been found to modify the methylation of H19 and to alter its expression. In addition, IL-27, one of the downstream factors in the H19 signaling pathway, plays an important role in the pathogenesis of pre-eclampsia (PE). In this study, we investigated the molecular mechanism underlying the therapeutic effect of Met in the management of PE both in vivo and in vitro. The role of H19 signaling pathway in PE was validated using online bioinformatics tools, luciferase assays, real-time PCR and Western Blot. A tail-cuff method was used to examine the blood pressures in PE rats with or without Met treatment. Cells exhibited a dose-dependent increase of H19 methylation, which inhibited the expression of H19. Additionally, upon the Met treatment, levels of miR-148-5p and miR-216-3p were both elevated in a dose-dependent manner while levels of p28 mRNA and EBI3 mRNA were both inhibited by Met treatment. Also, H19 was found to regulate the expression of miR-148a-5p and miR-216-3p, while P28 and EBI3 were respectively identified as target genes of miR-148a-5p and miR-216-3p. Therefore, the Met/H19/miR-148a-5p/P28 and Met/H19/miR-216-3p/EBI3 signaling pathways were implicated in the pathogenesis of PE. Met was implicated in the pathogenesis of PE via modulating the H19 signaling pathway. The methylation of H19 reduced H19 expression, which in turn could up-regulate the expression of miR-148-5p/miR-216-3p. And the expressions of subunits of IL-27, P28 and EBI3, were thus suppressed. Therefore, Met-induced inhibition of H19 also led to the reduction of IL-27 expression, TNF-α and IL-6 in vivo.
Collapse
Affiliation(s)
- Chang Shu
- Department of Obstetrics and Gynecology, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Dongmei Yan
- Department of Immunology, The Norman Bethune Medical Institute of Jilin University, Changchun, Jilin 130021, China
| | - Chen Chen
- Department of Obstetrics and Gynecology, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Yanxiang Mo
- Department of Obstetrics and Gynecology, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Linlin Wu
- Department of Obstetrics and Gynecology, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Jishuang Gu
- Department of Obstetrics and Gynecology, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Neelam Kumari Shah
- Department of Obstetrics and Gynecology, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Jin He
- Department of Obstetrics and Gynecology, The First Hospital of Jilin University, Changchun, Jilin 130021, China.
| | - Shuai Dong
- Department of Obstetrics and Gynecology, The First Hospital of Jilin University, Changchun, Jilin 130021, China.
| |
Collapse
|
80
|
Yang S, Wang H, Li D, Li M. Role of Endometrial Autophagy in Physiological and Pathophysiological Processes. J Cancer 2019; 10:3459-3471. [PMID: 31293650 PMCID: PMC6603423 DOI: 10.7150/jca.31742] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 04/24/2019] [Indexed: 12/17/2022] Open
Abstract
Endometrium is the mucosal lining of the uterus which expressed a cyclic process of proliferation, secretion and scaling under the control of hormones secreted by the ovary, and it also plays an indispensable role in the embryo implantation, the constitution of fetal-maternal interface, and the maintaining of pregnancy. In pathophysiological conditions, the abnormality or disorder of endometrium may lead to endometrium-related diseases, such as endometriosis, endometrium hyperplasia and even endometrial carcinoma. In recent years, more and more evidence revealed that autophagy exists in both the endometrium stroma cells and epithelial cells, and the activity of autophagy is changed in the different phases of menstruation, as well as in the endometrium-related diseases. Here, we aim to review the activity level, the regulatory factors and the function of autophagy in physiological and pathophysiological endometria, and to discuss the potential value of autophagy as a target for therapies of endometrium-related diseases.
Collapse
Affiliation(s)
- Shaoliang Yang
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University, 200080, People's Republic of China
| | - Haiyan Wang
- Department of Gynecology of Integrated Traditional Chinese and Western Medicine, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, 200011, People's Republic of China
| | - Dajin Li
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University, 200080, People's Republic of China
| | - Mingqing Li
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University, 200080, People's Republic of China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, People's Republic of China
| |
Collapse
|
81
|
Wang D, Liu N, Tian Y, Li Y, Shen X, Chen Y, Wu F. Expression profile of Let-7s in peripheral blood mononuclear cells of normal and severe preeclampsia pregnant women. Exp Mol Pathol 2019; 110:104263. [PMID: 31128089 DOI: 10.1016/j.yexmp.2019.104263] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 05/20/2019] [Accepted: 05/21/2019] [Indexed: 11/18/2022]
Abstract
HEADINGS AIM We aimed to investigate if the let-7 s expression level in the serum of peripheral blood from pregnant women with severe pre-eclampsia and normal pregnant women is related to the incidence of severe pre-eclampsia. METHODS Total RNA was extracted from collected peripheral blood mononuclear cells from 20 or over weeks pregnant women diagnosed with severe pre-eclampsia (age: 31.57 ± 4.94) and normal pregnant women (age: 29.75 ± 4.6) respectively, followed by real-time PCR to examine the expression of let-7 s. Correlation between let-7 s expression level and maternal age or body mass index of the normal pregnant women were also analyzed using SPSS21.0 software. RESULTS Let-7a and let-7 g were significantly increased in pregnant women with severe pre-eclampsia by 4.67 fold and 2.37 fold respectively compared to the normal pregnant women, whereas there was no significant difference in let-7b and let-7i. Moreover, there was no correlation between maternal age or body mass index and the expression level of let-7a, let-7b, let-7 g, and let-7i. CONCLUSIONS In conclusion, let-7a and let-7 g were significantly increased in the PBMCs of severe pre-eclampsia women compared to normal controls. Moreover, their expression level was not correlated to the maternal age or body mass of patients. Our data indicated that let-7a and let-7 g may be considered as predictive markers for SPE.
Collapse
Affiliation(s)
- Dandan Wang
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, Jilin Province 130041, China; Department of Obstetrics and Gynecology, Longnan Hospital of Daqing, Daqing, Heilongjiang Province 163458, China
| | - Ning Liu
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, Jilin Province 130041, China
| | - Yuan Tian
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, Jilin Province 130041, China
| | - Yang Li
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, Jilin Province 130041, China
| | - Xinyang Shen
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, Jilin Province 130041, China
| | - Yang Chen
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, Jilin Province 130041, China
| | - Fuju Wu
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, Jilin Province 130041, China.
| |
Collapse
|
82
|
Cao T, Jiang Y, Wang Z, Zhang N, Al-Hendy A, Mamillapalli R, Kallen AN, Kodaman P, Taylor HS, Li D, Huang Y. H19 lncRNA identified as a master regulator of genes that drive uterine leiomyomas. Oncogene 2019; 38:5356-5366. [PMID: 31089260 PMCID: PMC6755985 DOI: 10.1038/s41388-019-0808-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 02/11/2019] [Accepted: 03/19/2019] [Indexed: 12/17/2022]
Abstract
Uterine leiomyomas or fibroids (UFs) are benign tumors characterized by hyperplastic smooth muscle cells and excessive deposition of extracellular matrix (ECM). Afflicting ~80% of women, and symptomatic in 25%, UFs bring tremendous suffering and are an economic burden worldwide; they cause severe pain and bleeding, and are the leading cause of hysterectomy. Yet, UFs are severely understudied with few effective treatment options available; those that are available frequently have significant side effects such as menopausal symptoms. Recently, integrated genome-scale studies have revealed mutations and fibroid subtype-specific expression changes in key driver genes, with MED12 and HMGA2 together contributing to nearly 90% of all UFs, but their regulation of expression is poorly characterized. Here we report that the expression of H19 long noncoding RNA (lncRNA) is aberrantly increased in UFs. Using cell culture and genome-wide transcriptome and methylation profiling analyses, we demonstrate that H19 promotes expression of MED12, HMGA2, and key ECM-remodeling genes via multiple mechanisms including a new class of epigenetic modification by TET3. Our results mark the first example of an evolutionarily conserved lncRNA in pathogenesis of UFs and regulation of TET expression. Given the link between a H19 single-nucleotide polymorphism (SNP) and increased risk and tumor size of UFs, and the existence of multiple fibroid subtypes driven by key pathway genes regulated by H19, we propose a unifying mechanism for pathogenesis of uterine fibroids mediated by H19 and identify a pathway for future exploration of novel target therapies for uterine leiomyomas.
Collapse
Affiliation(s)
- Tiefeng Cao
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, 06510, USA.,Department of Gynecology and Obstetrics, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510070, China
| | - Ying Jiang
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, 06510, USA.,Department of Obstetrics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
| | - Zhangsheng Wang
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, 06510, USA.,Department of Cardiology, Fifth People's Hospital of Shanghai, Fudan University, Shanghai, 200240, China
| | - Na Zhang
- Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Ayman Al-Hendy
- Department of Obstetrics and Gynecology, University of Illinois College of Medicine, Chicago, IL, 60612, USA
| | - Ramanaiah Mamillapalli
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Amanda N Kallen
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Pinar Kodaman
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Hugh S Taylor
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Da Li
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital, China Medical University, Shenyang, 110004, China.
| | - Yingqun Huang
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, 06510, USA.
| |
Collapse
|
83
|
Xiao L, Gorospe M, Wang JY. Long noncoding RNAs in intestinal epithelium homeostasis. Am J Physiol Cell Physiol 2019; 317:C93-C100. [PMID: 31042423 DOI: 10.1152/ajpcell.00092.2019] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The epithelium of the mammalian intestinal mucosa is a rapidly self-renewing tissue in the body, and its homeostasis is preserved through well-controlled mechanisms. Long noncoding RNAs (lncRNAs) regulate a variety of biological functions and are intimately involved in the pathogenesis of diverse human diseases. Here we highlight the roles of several lncRNAs expressed in the intestinal epithelium, including uc.173, SPRY4-IT1, H19, and Gata6, in maintaining the integrity of the intestinal epithelium, focusing on the emerging evidence of lncRNAs in the regulation of intestinal mucosal regeneration and epithelial barrier function. We also discuss recent results that the interactions between lncRNAs with microRNAs and the RNA-binding protein HuR influence epithelial homeostasis. With rapidly advancing knowledge of lncRNAs, there is also growing recognition that lncRNAs in the intestinal epithelium might be promising therapeutic targets in our efforts to protect the integrity of the intestinal epithelium in response to stressful environments.
Collapse
Affiliation(s)
- Lan Xiao
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine , Baltimore, Maryland.,Baltimore Veterans Affairs Medical Center , Baltimore, Maryland
| | - Myriam Gorospe
- Laboratory of Genetics and Genomics, National Institute on Aging-Intramural Research Program, National Institutes of Health , Baltimore, Maryland
| | - Jian-Ying Wang
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine , Baltimore, Maryland.,Baltimore Veterans Affairs Medical Center , Baltimore, Maryland.,Department of Pathology, University of Maryland School of Medicine , Baltimore, Maryland
| |
Collapse
|
84
|
Chen Z, Wei H, Zhao X, Xin X, Peng L, Ning Y, Wang Y, Lan Y, Zhang Q. Metformin treatment alleviates polycystic ovary syndrome by decreasing the expression of MMP‐2 and MMP‐9 via H19/miR‐29b‐3p and AKT/mTOR/autophagy signaling pathways. J Cell Physiol 2019; 234:19964-19976. [PMID: 30989649 DOI: 10.1002/jcp.28594] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 03/01/2019] [Accepted: 03/06/2019] [Indexed: 12/27/2022]
Affiliation(s)
- Zhilan Chen
- Department of Obstetrics and Gynecology The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology Wuhan China
| | - Huafang Wei
- Department of Obstetrics and Gynecology Central Theater General Hospital of PLA Wuhan China
| | - Xiaoling Zhao
- Department of Obstetrics and Gynecology Central Theater General Hospital of PLA Wuhan China
| | - Xin Xin
- Department of Obstetrics and Gynecology The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology Wuhan China
| | - Ling Peng
- Department of Obstetrics and Gynecology The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology Wuhan China
| | - Yang Ning
- Department of Obstetrics and Gynecology The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology Wuhan China
| | - Yapei Wang
- Department of Obstetrics and Gynecology The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology Wuhan China
| | - Yanli Lan
- Department of Obstetrics and Gynecology Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science Xiangyang China
| | - Qinghua Zhang
- Department of Obstetrics and Gynecology The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology Wuhan China
| |
Collapse
|
85
|
Dong L, Li G, Li Y, Zhu Z. Upregulation of Long Noncoding RNA GAS5 Inhibits Lung Cancer Cell Proliferation and Metastasis via miR-205/PTEN Axis. Med Sci Monit 2019; 25:2311-2319. [PMID: 30926767 PMCID: PMC6452771 DOI: 10.12659/msm.912581] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Background Long noncoding RNA (lncRNA) is a key part of noncoding RNA class and increasing evidences have manifested that it plays a significant role in the physiology and pathology. The growth arrest-specific transcript 5 (GAS5) is a vital tumor suppressor in some types of cancers. However, the function of GAS5 in lung cancer remains largely no clear. The purpose of the current study was to identify the biological role of GAS5 in non-small cell lung cancer (NSCLC). Material/Methods To study the role of GAS5 in the NSCLC, the RT-PCR, Western Blot, Luciferase assay, and RNA immunoprecipitation assay was employed to determine the relationship of GAS5, miR-205, and PTEN. CCK8 assay, Cell migration and invasion assay was used for the role of GAS5 in lung cancer cell proliferation and metastasis. Results The results indicated that GAS5 was drastically downregulated in lung cancer cell lines. Further functional analysis showed that down-expression of GAS5 remarkably induced NSCLC growth, migration, and invasion. The luciferase reporter assays determined that miR-205 was a direct target of GAS5 in lung cancer. Moreover, the Phosphatase and tensin homologue (PTEN) was known as a direct target of miR-205 and miR-205/PTEN rescued the effects of GAS5 in NSCLC cells. Conclusions To sum up, our results illustrate that upregulation of GAS5 in NSCLC suppresses its growth, migration, and invasion via the miR-205/PTEN axis.
Collapse
Affiliation(s)
- Lizhen Dong
- Department of Clinical Laboratory, Tianjin Medical University General Hospital, Tianjin, China (mainland)
| | - Guangming Li
- Department of Pathogen Biology, Tianjin Medical University, Tianjin, China (mainland)
| | - Yongmei Li
- Department of Pathogen Biology, Tianjin Medical University, Tianjin, China (mainland)
| | - Ze Zhu
- Department of Pathogen Biology, Tianjin Medical University, Tianjin, China (mainland)
| |
Collapse
|
86
|
Wu P, Tang Y, Fang X, Xie C, Zeng J, Wang W, Zhao S. Metformin Suppresses Hypopharyngeal Cancer Growth by Epigenetically Silencing Long Non-coding RNA SNHG7 in FaDu Cells. Front Pharmacol 2019; 10:143. [PMID: 30853913 PMCID: PMC6395377 DOI: 10.3389/fphar.2019.00143] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 02/06/2019] [Indexed: 12/15/2022] Open
Abstract
Local recurrence after therapy remains a challenging problem for hypopharyngeal cancer (HPC) due to the chemotherapy resistance. Metformin is associated with reduced cancer risk through promoting global DNA methylation in cancer cells by controlling S-adenosylhomocysteine (SAHH) activity. However, the mechanisms by which metformin inhibits HPC remain elusive. In this study, we aim to investigate the role of metformin in HPC and illustrate the mechanism by which metformin regulates long non-coding RNAs (lncRNAs) expression. CCK-8 and annexin-V/PI double staining were performed to analyze the cell viability and apoptosis. LncRNA microarray analysis, QPCR, methylation specific PCR, Western blot and RNA Immunoprecipitation were performed to analyze the molecular mechanism, Here, we report that metformin inhibits FaDu cell proliferation in time- and dose-dependent manner by suppressing lncRNA SNHG7. Further investigations revealed that SNHG7 interacted with SAHH and metformin decreased SNHG7 expression by activating SAHH activity. Increased SAHH activity resulted in upregulating DNMT1 expression, leading to hypermethylation of SNHG7 promotor. In addition, upregulation of SNHG7 was associated with advanced stage. The patients with high SNHG7 have lower overall survival than that of with low SNHG7. Interestingly, SNHG7 levels were higher in taxol resistant patients than in taxol sensitive patients. Metformin sensitizes FaDu cells to taxol and irradiation through decreasing SNHG7. In conclusion, our recent study demonstrates that metformin inhibits FaDu cell proliferation by decreasing SNHG7 expression via SAHH-mediated DNA methylation. These findings indicate that combined metformin with paclitaxel or irradiation would be a novel therapeutic strategy to overcome resistance and prevent recurrence in HPC.
Collapse
Affiliation(s)
- Ping Wu
- Department of Otorhinolaryngology Head and Neck Surgery - Province Key Laboratory of Otolaryngology Critical Diseases, Xiangya Hospital of Central South University, Changsha, China
| | - Yaoyun Tang
- Department of Otorhinolaryngology Head and Neck Surgery - Province Key Laboratory of Otolaryngology Critical Diseases, Xiangya Hospital of Central South University, Changsha, China
| | - Xing Fang
- Department of Otorhinolaryngology Head and Neck Surgery - Province Key Laboratory of Otolaryngology Critical Diseases, Xiangya Hospital of Central South University, Changsha, China
| | - Chubo Xie
- Department of Otorhinolaryngology Head and Neck Surgery - Province Key Laboratory of Otolaryngology Critical Diseases, Xiangya Hospital of Central South University, Changsha, China
| | - Junfeng Zeng
- Department of Otorhinolaryngology Head and Neck Surgery - Province Key Laboratory of Otolaryngology Critical Diseases, Xiangya Hospital of Central South University, Changsha, China
| | - Wei Wang
- Department of Otorhinolaryngology Head and Neck Surgery - Province Key Laboratory of Otolaryngology Critical Diseases, Xiangya Hospital of Central South University, Changsha, China
| | - Suping Zhao
- Department of Otorhinolaryngology Head and Neck Surgery - Province Key Laboratory of Otolaryngology Critical Diseases, Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
87
|
Exploring lncRNA-Mediated Regulatory Networks in Endometrial Cancer Cells and the Tumor Microenvironment: Advances and Challenges. Cancers (Basel) 2019; 11:cancers11020234. [PMID: 30781521 PMCID: PMC6406952 DOI: 10.3390/cancers11020234] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 02/07/2019] [Accepted: 02/10/2019] [Indexed: 12/11/2022] Open
Abstract
Recent studies have revealed both the promise and challenges of targeting long non-coding RNAs (lncRNAs) to diagnose and treat endometrial cancer (EC). LncRNAs are upregulated or downregulated in ECs compared to normal tissues and their dysregulation has been linked to tumor grade, FIGO stage, the depth of myometrial invasion, lymph node metastasis and patient survival. Tumor suppressive lncRNAs (GAS5, MEG3, FER1L4 and LINC00672) and oncogenic lncRNAs (CCAT2, BANCR, NEAT1, MALAT1, H19 and Linc-RoR) have been identified as upstream modulators or downstream effectors of major signaling pathways influencing EC metastasis, including the PTEN/PI3K/AKT/mTOR, RAS/RAF/MEK/ERK, WNT/β-catenin and p53 signaling pathways. TUG1 and TDRG1 stimulate the VEGF-A pathway. PCGEM1 is implicated in activating the JAK/STAT3 pathway. Here, we present an overview of the expression pattern, prognostic value, biological function of lncRNAs in EC cells and their roles within the tumor microenvironment, focusing on the influence of lncRNAs on established EC-relevant pathways. We also describe the emerging classification of EC subtypes based on their lncRNA signature and discuss the clinical implications of lncRNAs as valuable biomarkers for EC diagnosis and potential targets for EC treatment.
Collapse
|
88
|
Ning B, Jin R, Wang D, Sun J. The H19/let-7 feedback loop contributes to developmental dysplasia and dislocation of the hip. Physiol Res 2019; 68:275-284. [PMID: 30628827 DOI: 10.33549/physiolres.933920] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Developmental dysplasia and dislocation of the hip (DDH) is the most common type of lower limb deformity in pediatric orthopedics. The mechanism of the signaling pathway has been studied in depth. However, the role of epigenetic regulation, such as lncRNA, is still far from clear. In this study, we successfully established a rat model of DDH and demonstrated that H19 was down-regulated in the development of DDH. Further, we constructed H19 knockdown (KD) and overexpression chondrocytes. H19 KD suppressed the proliferation of normal chondrocytes, while overexpression of H19 promoted cell proliferation of DDH chondrocytes. Finally, we revealed that H19 bound to let-7 and inhibited its function, acting as a competing endogenous RNA. Down-regulation of H19 is closely associated with DDH progression and H19 is an important epigenetic factor that regulates the proliferation of chondrocytes. H19 may thus be a potential clinical marker for DDH diagnosis and treatment.
Collapse
Affiliation(s)
- Bo Ning
- Department of Pediatric Orthopaedics, Children's Hospital of Anhui Medical university, Hefei, Anhui, China.
| | | | | | | |
Collapse
|
89
|
Huang L, Li Y, Wang C, Li N, Hou Y, Chang R, Sun M, Wang R, Zhu L, Qiao C. Overexpression of Collapsin Response Mediator Protein 1 Inhibits Human Trophoblast Cells Proliferation, Migration, and Invasion. Reprod Sci 2018; 26:954-960. [PMID: 30466368 DOI: 10.1177/1933719118799214] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Collapsin response mediator protein 1 (CRMP-1) is widely expressed in the nervous system and has tumor suppressive effects. Our previous studies have demonstrated that CRMP-1 was expressed in the trophoblasts of the whole stage of pregnancy with significantly increasing expression in the placenta of early-onset preeclampsia. Preeclampsia, especially early onset, is strongly associated with the dysfunction of trophoblast including proliferation, apoptosis, migration, and invasion. In this study, we found an inhibitory effect of CRMP-1 on proliferation, migration, invasion, and an enhanced effect on apoptosis in human trophoblast cell lines HTR-8/SVneo and JEG-3 by MTT assay, colony formation assay, cell viability assay, caspase 3/7 activity assay, scratch wound assay, and Matrigel Transwell assay. Overexpression of CRMP-1 in trophoblast cells led to downregulate expression of matrix metalloproteinase 2 and 9. The expression of CRMP-1 was detected by real-time quantitative polymerase chain reaction and Western blot analysis. Thus, we suggested that CRMP-1 might have implications for the pathogenesis of preeclampsia by regulating the biological behavior of trophoblast cells.
Collapse
Affiliation(s)
- Ling Huang
- Department of Obstetrics and Gynecology, Shengjing Hospital, China Medical University, No.36, Sanhao street, Shenyang, Liaoning Province, 110004, China.,Key Laboratory of Maternal-Fetal Medicine, China Medical University, Shenyang, Liaoning Province, China.,Key Laboratory of Obstetrics and Gynecology of Higher Education, China Medical University, Shenyang, Liaoning Province, China
| | - Yuanyuan Li
- Department of Obstetrics and Gynecology, Shengjing Hospital, China Medical University, No.36, Sanhao street, Shenyang, Liaoning Province, 110004, China.,Key Laboratory of Maternal-Fetal Medicine, China Medical University, Shenyang, Liaoning Province, China.,Key Laboratory of Obstetrics and Gynecology of Higher Education, China Medical University, Shenyang, Liaoning Province, China
| | - Chunhui Wang
- Department of Hepatobiliary Surgery, General Hospital of Shenyang Military Region, Shenyang, Liaoning Province, China
| | - Na Li
- Department of Obstetrics and Gynecology, Shengjing Hospital, China Medical University, No.36, Sanhao street, Shenyang, Liaoning Province, 110004, China.,Key Laboratory of Maternal-Fetal Medicine, China Medical University, Shenyang, Liaoning Province, China.,Key Laboratory of Obstetrics and Gynecology of Higher Education, China Medical University, Shenyang, Liaoning Province, China.,Department of Obstetrics and Gynecology, Shenyang Forth People's Hospital, Shenyang, Liaoning Province, China
| | - Yue Hou
- Department of Obstetrics and Gynecology, Shengjing Hospital, China Medical University, No.36, Sanhao street, Shenyang, Liaoning Province, 110004, China.,Key Laboratory of Maternal-Fetal Medicine, China Medical University, Shenyang, Liaoning Province, China.,Key Laboratory of Obstetrics and Gynecology of Higher Education, China Medical University, Shenyang, Liaoning Province, China
| | - Ruijing Chang
- Department of Obstetrics and Gynecology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Man Sun
- Department of Obstetrics and Gynecology, Shengjing Hospital, China Medical University, No.36, Sanhao street, Shenyang, Liaoning Province, 110004, China.,Key Laboratory of Maternal-Fetal Medicine, China Medical University, Shenyang, Liaoning Province, China.,Key Laboratory of Obstetrics and Gynecology of Higher Education, China Medical University, Shenyang, Liaoning Province, China
| | - Ruochen Wang
- Department of Biomedical Engineering, University of Wisconsin at Madison, Madison, WI, USA
| | - Lei Zhu
- Shenyang Academy of Environmental Sciences, Shenyang, Liaoning Province, China
| | - Chong Qiao
- Department of Obstetrics and Gynecology, Shengjing Hospital, China Medical University, No.36, Sanhao street, Shenyang, Liaoning Province, 110004, China. .,Key Laboratory of Maternal-Fetal Medicine, China Medical University, Shenyang, Liaoning Province, China. .,Key Laboratory of Obstetrics and Gynecology of Higher Education, China Medical University, Shenyang, Liaoning Province, China.
| |
Collapse
|
90
|
Wu ZR, Yan L, Liu YT, Cao L, Guo YH, Zhang Y, Yao H, Cai L, Shang HB, Rui WW, Yang G, Zhang XB, Tang H, Wang Y, Huang JY, Wei YX, Zhao WG, Su B, Wu ZB. Inhibition of mTORC1 by lncRNA H19 via disrupting 4E-BP1/Raptor interaction in pituitary tumours. Nat Commun 2018; 9:4624. [PMID: 30397197 PMCID: PMC6218470 DOI: 10.1038/s41467-018-06853-3] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 09/11/2018] [Indexed: 01/02/2023] Open
Abstract
Aberrant expression of long noncoding RNA H19 has been associated with tumour progression, but the underlying molecular tumourigenesis mechanisms remain largely unknown. Here, we report that H19 expression is frequently downregulated in human primary pituitary adenomas and is negatively correlated with tumour progression. Consistently, upregulation of H19 expression inhibits pituitary tumour cell proliferation in vitro and tumour growth in vivo. Importantly, we uncover a function of H19, which controls cell/tumour growth through inhibiting function of mTORC1 but not mTORC2. Mechanistically, we show that H19 could block mTORC1-mediated 4E-BP1 phosphorylation without affecting S6K1 activation. At the molecular level, H19 interacted with 4E-BP1 at the TOS motif and competitively inhibited 4E-BP1 binding to Raptor. Finally, we demonstrate that H19 is more effective than cabergoline treatment in the suppression of pituitary tumours. Together, our study uncovered the role of H19-mTOR-4E-BP1 axis in pituitary tumour growth regulation that may be a potential therapeutic target for human pituitary tumours. LncRNA H19 has been shown to be aberrantly expressed in different cancers. Here, the authors show that H19 lncRNA is downregulated in pituitary adenomas and H19 is able to impede pituitary tumorigenesis via disruption of 4E-BPB1 and Raptor interaction to inhibit the phosphorylation of 4E-BP1.
Collapse
Affiliation(s)
- Ze Rui Wu
- Department of Neurosurgery, Center of Pituitary Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.,Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, 325000, Wenzhou, China
| | - Lichong Yan
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Yan Ting Liu
- Department of Neurosurgery, Center of Pituitary Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.,Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, 325000, Wenzhou, China
| | - Lei Cao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, 100050, Beijing, China
| | - Yu Hang Guo
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, 325000, Wenzhou, China
| | - Yong Zhang
- Department of Neurosurgery, Center of Pituitary Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.,Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, 325000, Wenzhou, China
| | - Hong Yao
- Department of Neurosurgery, Center of Pituitary Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Lin Cai
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, 325000, Wenzhou, China
| | - Han Bing Shang
- Department of Neurosurgery, Center of Pituitary Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Wei Wei Rui
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Gang Yang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, 410000, Chongqing, China
| | - Xiao Biao Zhang
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, 200032 Shanghai, China
| | - Hao Tang
- Department of Neurosurgery, Center of Pituitary Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Yu Wang
- Department of Neurosurgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 200127, Shanghai, China
| | - Jin Yan Huang
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Yong Xu Wei
- Department of Neurosurgery, Center of Pituitary Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Wei Guo Zhao
- Department of Neurosurgery, Center of Pituitary Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Bing Su
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.
| | - Zhe Bao Wu
- Department of Neurosurgery, Center of Pituitary Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China. .,Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, 325000, Wenzhou, China.
| |
Collapse
|
91
|
Geng T, Liu Y, Xu Y, Jiang Y, Zhang N, Wang Z, Carmichael GG, Taylor HS, Li D, Huang Y. H19 lncRNA Promotes Skeletal Muscle Insulin Sensitivity in Part by Targeting AMPK. Diabetes 2018; 67:2183-2198. [PMID: 30201684 PMCID: PMC6198334 DOI: 10.2337/db18-0370] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 08/24/2018] [Indexed: 12/12/2022]
Abstract
Skeletal muscle plays a pivotal role in regulating systemic glucose homeostasis in part through the conserved cellular energy sensor AMPK. AMPK activation increases glucose uptake, lipid oxidation, and mitochondrial biogenesis, leading to enhanced muscle insulin sensitivity and whole-body energy metabolism. Here we show that the muscle-enriched H19 long noncoding RNA (lncRNA) acts to enhance muscle insulin sensitivity, at least in part, by activating AMPK. We identify the atypical dual-specificity phosphatase DUSP27/DUPD1 as a potentially important downstream effector of H19. We show that DUSP27, which is highly expressed in muscle with previously unknown physiological function, interacts with and activates AMPK in muscle cells. Consistent with decreased H19 expression in the muscle of insulin-resistant human subjects and rodents, mice with genetic H19 ablation exhibit muscle insulin resistance. Furthermore, a high-fat diet downregulates muscle H19 via both posttranscriptional and epigenetic mechanisms. Our results uncover an evolutionarily conserved, highly expressed lncRNA as an important regulator of muscle insulin sensitivity.
Collapse
Affiliation(s)
- Tingting Geng
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, CT
- Department of Endocrinology, First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi, People's Republic of China
| | - Ya Liu
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, CT
- Department of Veterinary Medicine, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui, People's Republic of China
| | - Yetao Xu
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, CT
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Ying Jiang
- Department of Obstetrics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People's Republic of China
| | - Na Zhang
- Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT
| | - Zhangsheng Wang
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, CT
- Department of Cardiology, Fifth People's Hospital of Shanghai, Fudan University, Shanghai, People's Republic of China
| | - Gordon G Carmichael
- Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT
| | - Hugh S Taylor
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, CT
| | - Da Li
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, CT
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital, China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Yingqun Huang
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, CT
| |
Collapse
|
92
|
Ansardamavandi A, Tafazzoli-Shadpour M, Shokrgozar MA. Behavioral remodeling of normal and cancerous epithelial cell lines with differing invasion potential induced by substrate elastic modulus. Cell Adh Migr 2018; 12:472-488. [PMID: 29969940 PMCID: PMC6363025 DOI: 10.1080/19336918.2018.1475803] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 05/07/2018] [Indexed: 12/20/2022] Open
Abstract
The micro-environment of cancer cells in the body is mechanically stiffer than that of normal cells. We cultured three breast cell lines of MCF10A-normal, MCF7-noninvasive, and MDA-MB-231-invasive on PDMS substrates with different elastic moduli and different cellular features were examined.Effects of substrate stiffness on cell behavior were evident among all cell lines. Cancerous cells were more sensitive to substrate stiffness for cell behaviors related to cell motility and migration which are necessary for invasion. The invasive cancerous cells were the most motile on substrates with moderate stiffness followed by non-invasive cancerous cells. Gene markers alterations were generally according to the analyzed cell movement parameters. Results suggest that alterations in matrix stiffness may be related to cancer disease and progression.
Collapse
Affiliation(s)
- Arian Ansardamavandi
- Faculty of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | | | | |
Collapse
|
93
|
Lin X, Qiu J, Hua K. Long non-coding RNAs as emerging regulators of epithelial to mesenchymal transition in gynecologic cancers. Biosci Trends 2018; 12:342-353. [PMID: 30146551 DOI: 10.5582/bst.2018.01181] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Gynecologic cancer is a vital global healthcare issue with high rates of mortality and morbidity. Tumor metastasis attributes to most of the death suffering from solid tumors. The epithelial-mesenchymal transition (EMT) plays a pivotal role in initiating metastasis. Long non-coding RNAs (lncRNAs), a well-known group of non-coding RNAs, and a prominent topic in life science research, are misregulated in many malignancies and some are EMT-associated. In the case of gynecologic cancers, several EMT-associated lncRNAs have been identified and found to be implicated in cancer aggressiveness and progression. Mechanically, these lncRNAs participate in the EMT-related metastatic process in multiple ways including interaction with polycomb repressive complex 2 (PRC2), regulation of EMT signaling networks, mediation of EMT-transcription factors (EMT-TFs) and EMT markers, and cooperation with microRNAs (miRNAs). Further studies on these EMT-associated lncRNAs and identification of more relevant lncRNAs are imperative for the lncRNAs-based clinical management of high rate of metastasis in patients with gynecologic cancers.
Collapse
Affiliation(s)
- Xiaojing Lin
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University.,Shanghai Medical College, Fudan University.,Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Fudan University
| | - Junjun Qiu
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University.,Shanghai Medical College, Fudan University.,Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Fudan University
| | - Keqin Hua
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University.,Shanghai Medical College, Fudan University.,Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Fudan University
| |
Collapse
|
94
|
Stojic L, Lun AT, Mangei J, Mascalchi P, Quarantotti V, Barr AR, Bakal C, Marioni JC, Gergely F, Odom DT. Specificity of RNAi, LNA and CRISPRi as loss-of-function methods in transcriptional analysis. Nucleic Acids Res 2018; 46:5950-5966. [PMID: 29860520 PMCID: PMC6093183 DOI: 10.1093/nar/gky437] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 05/03/2018] [Accepted: 05/09/2018] [Indexed: 02/06/2023] Open
Abstract
Loss-of-function (LOF) methods such as RNA interference (RNAi), antisense oligonucleotides or CRISPR-based genome editing provide unparalleled power for studying the biological function of genes of interest. However, a major concern is non-specific targeting, which involves depletion of transcripts other than those intended. Little work has been performed to characterize the off-target effects of these common LOF methods at the whole-transcriptome level. Here, we experimentally compared the non-specific activity of RNAi, antisense oligonucleotides and CRISPR interference (CRISPRi). All three methods yielded non-negligible off-target effects in gene expression, with CRISPRi also exhibiting strong clonal effects. As an illustrative example, we evaluated the performance of each method for determining the role of an uncharacterized long noncoding RNA (lncRNA). Several LOF methods successfully depleted the candidate lncRNA but yielded different sets of differentially expressed genes as well as a different cellular phenotype upon depletion. Similar discrepancies between methods were observed with a protein-coding gene (Ch-TOG/CKAP5) and another lncRNA (MALAT1). We suggest that the differences between methods arise due to method-specific off-target effects and provide guidelines for mitigating such effects in functional studies. Our recommendations provide a framework with which off-target effects can be managed to improve functional characterization of genes of interest.
Collapse
Affiliation(s)
- Lovorka Stojic
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| | - Aaron T L Lun
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| | - Jasmin Mangei
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| | - Patrice Mascalchi
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| | - Valentina Quarantotti
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| | - Alexis R Barr
- Institute of Cancer Research, 237 Fulham Road London SW3 6JB, UK
| | - Chris Bakal
- Institute of Cancer Research, 237 Fulham Road London SW3 6JB, UK
| | - John C Marioni
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
- European Bioinformatics Institute, European Molecular Biology Laboratory (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridgeshire CB10 1SD, UK
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Fanni Gergely
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| | - Duncan T Odom
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| |
Collapse
|
95
|
Li BL, Wan XP. The role of lncRNAs in the development of endometrial carcinoma. Oncol Lett 2018; 16:3424-3429. [PMID: 30127944 DOI: 10.3892/ol.2018.9065] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 07/03/2017] [Indexed: 01/23/2023] Open
Abstract
Endometrial carcinoma (EC) is one of the most common types of gynecological cancer. Long noncoding RNAs (lncRNAs) are associated with the carcinogenesis and progression of EC. In the following review, the emerging role of lncRNAs in EC initiation and progression is considered. The profile of lncRNAs is becoming higher as the contribution of lncRNAs to carcinogenesis through diverse mechanisms is being increasingly recognized, including in EC. A number of lncRNA-profiling studies have identified aberrantly expressed lncRNAs in EC tissue, and the regulatory network associated with these lncRNAs may be critical in EC progression. Additionally, certain lncRNAs may have diagnostic and/or prognostic significance. The potential function of lncRNAs as prospective therapeutic and prognostic targets in EC will be evaluated.
Collapse
Affiliation(s)
- Bi-Lan Li
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 201204, P.R. China
| | - Xiao-Ping Wan
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 201204, P.R. China
| |
Collapse
|
96
|
Bridgeman SC, Ellison GC, Melton PE, Newsholme P, Mamotte CDS. Epigenetic effects of metformin: From molecular mechanisms to clinical implications. Diabetes Obes Metab 2018; 20:1553-1562. [PMID: 29457866 DOI: 10.1111/dom.13262] [Citation(s) in RCA: 133] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 02/13/2018] [Accepted: 02/13/2018] [Indexed: 12/15/2022]
Abstract
There is a growing body of evidence that links epigenetic modifications to type 2 diabetes. Researchers have more recently investigated effects of commonly used medications, including those prescribed for diabetes, on epigenetic processes. This work reviews the influence of the widely used antidiabetic drug metformin on epigenomics, microRNA levels and subsequent gene expression, and potential clinical implications. Metformin may influence the activity of numerous epigenetic modifying enzymes, mostly by modulating the activation of AMP-activated protein kinase (AMPK). Activated AMPK can phosphorylate numerous substrates, including epigenetic enzymes such as histone acetyltransferases (HATs), class II histone deacetylases (HDACs) and DNA methyltransferases (DNMTs), usually resulting in their inhibition; however, HAT1 activity may be increased. Metformin has also been reported to decrease expression of multiple histone methyltransferases, to increase the activity of the class III HDAC SIRT1 and to decrease the influence of DNMT inhibitors. There is evidence that these alterations influence the epigenome and gene expression, and may contribute to the antidiabetic properties of metformin and, potentially, may protect against cancer, cardiovascular disease, cognitive decline and aging. The expression levels of numerous microRNAs are also reportedly influenced by metformin treatment and may confer antidiabetic and anticancer activities. However, as the reported effects of metformin on epigenetic enzymes act to both increase and decrease histone acetylation, histone and DNA methylation, and gene expression, a significant degree of uncertainty exists concerning the overall effect of metformin on the epigenome, on gene expression, and on the subsequent effect on the health of metformin users.
Collapse
Affiliation(s)
- Stephanie Claire Bridgeman
- School of Pharmacy and Biomedical Sciences, and Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia
| | - Gaewyn Colleen Ellison
- School of Pharmacy and Biomedical Sciences, and Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia
| | - Phillip Edward Melton
- School of Pharmacy and Biomedical Sciences, and Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia
- Centre for Genetic Origins of Health and Disease, Faculty of Health and Medical Science, The University of Western Australia, Perth, Western Australia, Australia
| | - Philip Newsholme
- School of Pharmacy and Biomedical Sciences, and Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia
| | - Cyril Desire Sylvain Mamotte
- School of Pharmacy and Biomedical Sciences, and Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia
| |
Collapse
|
97
|
Abstract
Long non-coding RNAs (lncRNAs) refer to functional cellular RNAs molecules longer than 200 nucleotides in length. Unlike microRNAs, which have been widely studied, little is known about the enigmatic role of lncRNAs. However, lncRNAs have motivated extensively attention in the past few years and are emerging as potentially important regulators in pathological processes, including in cancer. We now understand that lncRNAs play role in cancer through their interactions with DNA, protein, and RNA in many instances. Moreover, accumulating evidence has recognized that large classes of lncRNAs are functional for ovarian cancer. Nevertheless, the biological phenomena and molecular mechanisms of lncRNAs in ovarian cancer remain to be better identified. In this review, we outline the dysregulated expression of lncRNAs and their potential clinical implications in ovarian cancer, with a particular emphasis on discussing the well characterized mechanisms underlying lncRNAs in ovarian cancer.
Collapse
Affiliation(s)
- Lei Zhan
- Department of gynecology and obstetrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601 China
| | - Jun Li
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, 230032 China
| | - Bing Wei
- Department of gynecology and obstetrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601 China
| |
Collapse
|
98
|
Liu Y, Li M, Bo X, Li T, Ma L, Zhai T, Huang T. Systematic Analysis of Long Non-Coding RNAs and mRNAs in the Ovaries of Duroc Pigs During Different Follicular Stages Using RNA Sequencing. Int J Mol Sci 2018; 19:ijms19061722. [PMID: 29891752 PMCID: PMC6032137 DOI: 10.3390/ijms19061722] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 06/05/2018] [Accepted: 06/06/2018] [Indexed: 01/22/2023] Open
Abstract
The dynamic process involving the selection and maturation of follicles is regulated and controlled by a highly synchronized and exquisitely timed cascade of gene expression. Studies have shown that long non-coding RNA (lncRNA) is essential for the normal maintenance of animal reproductive function and has an important regulatory function in ovarian development and hormone secretion. In this study, a total of 2076 lncRNAs (1362 known lncRNAs and 714 new lncRNAs) and 25,491 mRNAs were identified in libraries constructed from Duroc ovaries on days 0, 2 and 4 of follicle development. lncRNAs were shorter, had fewer exons, exhibited a shorter ORF (Open Reading Frame) length and lower expression levels, and were less conserved than mRNAs. Furthermore, 1694 transcripts (140 lncRNAs and 1554 mRNAs) were found to be differentially expressed in pairwise comparisons. A total of 6945 co-localized mRNAs were detected in cis in 2076 lncRNAs. The most enriched GO (Gene Ontology) terms were related to developmental processes. KEGG (Kyoto Encyclopedia of Genes and Genomes) pathway analysis revealed that the differentially expressed lncRNAs targeted mRNAs, and the differentially expressed mRNAs were related to the TGF-β signaling pathway, the PI3K-Akt signaling pathway, the Retinol metabolic pathway and the Wnt signaling pathway. This study deepened our understanding of the genetic basis and molecular mechanisms of follicular development in pigs.
Collapse
Affiliation(s)
- Yi Liu
- College of Animal Science and Technology, Shihezi University, 221 North Fourth Road, Shihezi 832000, China.
- State Key Laboratory of Sheep Genetic Improvement and Healthy Production, Institute of Animal Husbandry and Veterinary, Xinjiang Academy of Agricultural and Reclamation Sciences, 221 Wu Yi Road, Shihezi 832000, China.
| | - Mengxun Li
- College of Animal Science and Technology, Shihezi University, 221 North Fourth Road, Shihezi 832000, China.
| | - Xinwen Bo
- State Key Laboratory of Sheep Genetic Improvement and Healthy Production, Institute of Animal Husbandry and Veterinary, Xinjiang Academy of Agricultural and Reclamation Sciences, 221 Wu Yi Road, Shihezi 832000, China.
| | - Tao Li
- College of Animal Science and Technology, Shihezi University, 221 North Fourth Road, Shihezi 832000, China.
| | - Lipeng Ma
- College of Animal Science and Technology, Shihezi University, 221 North Fourth Road, Shihezi 832000, China.
| | - Tenjiao Zhai
- College of Animal Science and Technology, Shihezi University, 221 North Fourth Road, Shihezi 832000, China.
| | - Tao Huang
- College of Animal Science and Technology, Shihezi University, 221 North Fourth Road, Shihezi 832000, China.
| |
Collapse
|
99
|
Li H, Ma SQ, Huang J, Chen XP, Zhou HH. Roles of long noncoding RNAs in colorectal cancer metastasis. Oncotarget 2018; 8:39859-39876. [PMID: 28418892 PMCID: PMC5503659 DOI: 10.18632/oncotarget.16339] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 02/20/2017] [Indexed: 12/11/2022] Open
Abstract
Colorectal cancer (CRC) is the 3rd most common malignancies worldwide. Metastasis is responsible for more than 90% CRC patients' death. Long noncoding RNAs (lncRNAs) are an important class of transcribed RNA molecules greater than 200 nucleotides in length. With the development of whole genome sequencing technologies, they have been gained more attention. Accumulating evidences suggest that abnormal expression of lncRNAs in diverse diseases are involved in various biological functions such as proliferation, apoptosis, metastasis and differentiation by acting as epigenetic, splicing, transcriptional or post-transcriptional regulators. Aberrant expression of lncRNAs has also been found in CRC. Besides, recent studies have indicated that lncRNAs play important roles in tumourigenesis and cancer metastasis. They participate in the process of metastasis by activing or inhibiting the metastatic pathways. However, their functions on the development of cancer metastasis are poorly understood. In this review, we highlight the findings of roles for lncRNAs in CRC metastasis and review the metastatic pathways of lncRNAs leading to cancer metastasis in CRC, including escape of apoptosis, epithelial-mesenchymal transition (EMT), angiogenesis and invasion, migration and proliferation. Furthermore, we also discuss the potential clinical application of lncRNAs in CRC as diagnostic markers and therapeutic targets.
Collapse
Affiliation(s)
- He Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, P.R. China.,Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, P.R. China
| | - Si-Qing Ma
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, P.R. China.,Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, P.R. China
| | - Jin Huang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, P.R. China.,Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, P.R. China
| | - Xiao-Ping Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, P.R. China.,Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, P.R. China.,Hunan Province Cooperation Innovation Center for Molecular Target New Drug Study, Hengyang, P. R. China
| | - Hong-Hao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, P.R. China.,Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, P.R. China.,Hunan Province Cooperation Innovation Center for Molecular Target New Drug Study, Hengyang, P. R. China
| |
Collapse
|
100
|
Long non-coding RNA H19 contributes to apoptosis of hippocampal neurons by inhibiting let-7b in a rat model of temporal lobe epilepsy. Cell Death Dis 2018; 9:617. [PMID: 29795132 PMCID: PMC5966382 DOI: 10.1038/s41419-018-0496-y] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 02/23/2018] [Accepted: 03/02/2018] [Indexed: 01/09/2023]
Abstract
Temporal lobe epilepsy (TLE) is one of the most common types of intractable epilepsy, characterized by hippocampal neuron damage and hippocampal sclerosis. Long noncoding RNAs (lncRNAs) have been increasingly recognized as posttranscriptional regulators. However, their expression levels and functions in TLE remain largely unknown. In the present study, TLE rat model is used to explore the expression profiles of lncRNAs in the hippocampus of epileptic rats using microarray analysis. Our results demonstrate that H19 is the most pronouncedly differentiated lncRNA, significantly upregulated in the latent period of TLE. Moreover, the in vivo studies using gain- and loss-of-function approaches reveal that the overexpression of H19 aggravates SE-induced neuron apoptosis in the hippocampus, while inhibition of H19 protects the rats from SE-induced cellular injury. Finally, we show that H19 might function as a competing endogenous RNA to sponge microRNA let-7b in the regulation of cellular apoptosis. Overall, our study reveals a novel lncRNA H19-mediated mechanism in seizure-induced neural damage and provides a new target in developing lncRNA-based strategies to reduce seizure-induced brain injury.
Collapse
|