1
|
Jin X, Lu Y, Fan Z. Exploring NamiRNA networks and time-series gene expression in osteogenic differentiation of adipose-derived stem cells. Ann Med 2025; 57:2478323. [PMID: 40100054 PMCID: PMC11921168 DOI: 10.1080/07853890.2025.2478323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 02/12/2025] [Accepted: 02/28/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND Adipose-derived stem cells (ADSCs) are a type of stem cell found in adipose tissue with the capacity to differentiate into multiple lineages, including osteoblasts. The differentiation of ADSCs into osteoblasts underlies osteogenic and pathological cellular basis in osteoporosis, bone damage and repair. METHODS Focused on ADSCs osteogenic differentiation, we conducted mRNA, microRNA expression and bioinformatics analysis, including gene differential expression, time series-based trend analysis, functional enrichment, and generates potential nuclear activating miRNAs (NamiRNA) regulatory network. The screened mRNAs in NamiRNA regulatory network were validated with correlation analysis. RESULTS The NamiRNA Regulatory Network reveals 4 mRNAs (C12orf61, MIR31HG, NFE2L1, and PCYOX1L) significantly downregulated in differentiated group and may be associated with ADSCs stemness. Furthermore, the significantly upregulated 10 genes (ACTA2, TAGLN, LY6E, IFITM3, NGFRAP1, TCEAL4, ATP5C1, CAV1, RPSA, and KDELR3) were significantly enriched in osteogenic-related pathways, and negatively correlated with ADSCs cell stemness in vitro. CONCLUSION These findings uncover potential genes related to ADSCs osteogenic differentiation, and provide theoretical basis for underlying ADSCs osteogenic differentiation and related diseases.
Collapse
Affiliation(s)
- Xin Jin
- Department of Plastic Surgery, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yi Lu
- Department of Plastic Surgery, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhihong Fan
- Department of Plastic Surgery, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
2
|
Ren X, Liu G, Zhou J. Nuclear-activating miRNAs: unveiling the intricacies of subcellular miRNA function and regulation in cancer and immunity disease. Cancer Cell Int 2025; 25:147. [PMID: 40234876 PMCID: PMC11998458 DOI: 10.1186/s12935-025-03760-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 03/19/2025] [Indexed: 04/17/2025] Open
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs that traditionally recognized as negative regulators of gene expression through post-transcriptional regulation in the cytoplasm. However, recent discoveries have unveiled some novel miRNA functions in the cell nucleus, where a subset of miRNAs, termed nuclear-activating miRNAs (NamiRNAs), play pivotal roles in gene activation and transcriptional regulation for cancer and immunity disease. The discovery of NamiRNAs demonstrated a complementary regulatory function of miRNA, showing their differential activities in the nucleus and cytoplasm. This review aims to explore the biogenesis, mechanisms, and regulatory functions of NamiRNAs, deciphering their involvement in NamiRNA-gene network for gene expression modulation, and emerging significance as drug targets against cancer.
Collapse
Affiliation(s)
- Xiang Ren
- Department of Gastrointestinal Surgery, The First Hospital of China Medical University, Nanjing Street 155, Shenyang, 110001, China
- Department of Colorectal Hernia Surgery, Binzhou Medical University Hospital, Yantai, China
| | - Gang Liu
- Department of Gastrointestinal Surgery, The First Hospital of China Medical University, Nanjing Street 155, Shenyang, 110001, China
- Shenyang Medical Nutrition Clinical Medical Research Center, Shenyang, China
| | - Jianping Zhou
- Department of Gastrointestinal Surgery, The First Hospital of China Medical University, Nanjing Street 155, Shenyang, 110001, China.
- Shenyang Medical Nutrition Clinical Medical Research Center, Shenyang, China.
| |
Collapse
|
3
|
Jiao L, Junfang Z, Yanna L, Caixia J, Chen Z, Song J, Jie X, Xiaoli Y, Xin G, Libo X, Feng W, lixia L, Chunli X, Lei X. miR-153 promotes neural differentiation by activating the cell adhesion/Ca 2+ signaling pathway and targeting ion channel activity in HT-22 cells by bioinformatic analysis. Heliyon 2024; 10:e30204. [PMID: 38694121 PMCID: PMC11061740 DOI: 10.1016/j.heliyon.2024.e30204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 04/21/2024] [Accepted: 04/22/2024] [Indexed: 05/03/2024] Open
Abstract
MicroRNAs have been studied extensively in neurodegenerative diseases. In a previous study, miR-153 promoted neural differentiation and projection formation in mouse hippocampal HT-22 cells. However, the pathways and molecular mechanism underlying miR-153-induced neural differentiation remain unclear. To explore the molecular mechanism of miR-153 on neural differentiation, we performed RNA sequencing on miR-153-overexpressed HT-22 cells. Based on RNA sequencing, differentially expressed genes (DEGs) and pathways in miR-153-overexpressed cells were identified. The Database for Annotation, Visualization and Integrated Discovery and Gene Set Enrichment Analysis were used to perform functional annotation and enrichment analysis of DEGs. Targetscan predicted the targets of miR-153. The Search Tool for the Retrieval of Interacting Genes and Cytoscape, were used to construct protein-protein interaction networks and identify hub genes. Q-PCR was used to detect mRNA expression of the identified genes. The expression profiles of the identified genes were compared between embryonic days 9.5 (E9.5) and E11.5 in the embryotic mouse brain of the GDS3442 dataset. Cell Counting Kit-8 assay was used to determine cell proliferation and cellular susceptibility to amyloid β-protein (Aβ) toxicity in miR-153-overexpressed cells. The results indicated that miR-153 increased cell adhesion/Ca2+ (Cdh5, Nrcam, and P2rx4) and Bdnf/Ntrk2 neurotrophic signaling pathway, and decreased ion channel activity (Kcnc3, Kcna4, Clcn5, and Scn5a). The changes in the expression of the identified genes in miR-153-overexpressed cells were consistent with the expression profile of GDS3442 during neural differentiation. In addition, miR-153 overexpression decreased cellular susceptibility to Aβ toxicity in HT-22 cells. In conclusion, miR-153 overexpression may promote neural differentiation by inducing cell adhesion and the Bdnf/Ntrk2 pathway, and regulating electrophysiological maturity by targeting ion channels. MiR-153 may play an important role in neural differentiation; the findings provide a useful therapeutic direction for neurodegenerative diseases.
Collapse
Affiliation(s)
- Li Jiao
- Teaching Laboratory Center, Tongji University School of Medicine, Shanghai, China
| | - Zhang Junfang
- Teaching Laboratory Center, Tongji University School of Medicine, Shanghai, China
| | - Li Yanna
- Teaching Laboratory Center, Tongji University School of Medicine, Shanghai, China
| | - Jin Caixia
- Department of Biochemistry and Molecular Biology, Tongji University School of Medicine, Shanghai, China
| | - Zhang Chen
- Department of Laboratory Research Center, Tongji University School of Medicine, Shanghai, China
| | - Jia Song
- Teaching Laboratory Center, Tongji University School of Medicine, Shanghai, China
| | - Xu Jie
- Teaching Laboratory Center, Tongji University School of Medicine, Shanghai, China
| | - Yan Xiaoli
- Teaching Laboratory Center, Tongji University School of Medicine, Shanghai, China
| | - Gui Xin
- School of Life Science and Technology, Tongji University, Shanghai, China
| | - Xing Libo
- School of Life Science and Technology, Tongji University, Shanghai, China
| | - Wang Feng
- Department of Neurology, The Seventh People's Hospital of Integrated Traditional Chinese and Western Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lu lixia
- Department of Biochemistry and Molecular Biology, Tongji University School of Medicine, Shanghai, China
| | - Xu Chunli
- Department of Neurology, The Seventh People's Hospital of Integrated Traditional Chinese and Western Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xu Lei
- Department of Biochemistry and Molecular Biology, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
4
|
Xia J, Wang D, Guo W, Pei Y, Zhang L, Bao L, Li Y, Qu Y, Zhao Y, Hao C, Yao W. Exposure to micron-grade silica particles triggers pulmonary fibrosis through cell-to-cell delivery of exosomal miR-107. Int J Biol Macromol 2024; 266:131058. [PMID: 38522707 DOI: 10.1016/j.ijbiomac.2024.131058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 02/17/2024] [Accepted: 03/04/2024] [Indexed: 03/26/2024]
Abstract
Long-term exposure to inhalable silica particles may lead to severe systemic pulmonary disease, such as silicosis. Exosomes have been demonstrated to dominate the pathogenesis of silicosis, but the underlying mechanisms remain unclear. Therefore, this study aimed to explore the roles of exosomes by transmitting miR-107, which has been linked to the toxic pulmonary effects of silica particles. We found that miR-107, miR-122-5p, miR-125a-5p, miR-126-5p, and miR-335-5p were elevated in exosomes extracted from the serum of patients with silicosis. Notably, an increase in miR-107 in serum exosomes and lung tissue was observed in the experimental silicosis mouse model, while the inhibition of miR-107 reduced pulmonary fibrosis. Moreover, exosomes helped the migration of miR-107 from macrophages to lung fibroblasts, triggering the transdifferentiation of cell phenotypes. Further experiments demonstrated that miR-107 targets CDK6 and suppresses the expression of retinoblastoma protein phosphorylation and E2F1, resulting in cell-cycle arrest. Overall, micron-grade silica particles induced lung fibrosis through exosomal miR-107 negatively regulating the cell cycle signaling pathway. These findings may open a new avenue for understanding how silicosis is regulated by exosome-mediated cell-to-cell communication and suggest the prospect of exosomes as therapeutic targets.
Collapse
Affiliation(s)
- Jiarui Xia
- Department of Occupational and Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Di Wang
- Department of Occupational and Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Wei Guo
- Department of Occupational Disease, Henan Institute for Occupational Medicine, Zhengzhou 450052, China
| | - Yangqing Pei
- Department of Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Lin Zhang
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Maternal and Child Health Care Hospital of Shandong Province, Shandong University, Jinan 250001, China
| | - Lei Bao
- Department of Occupational and Environmental Health, School of Public Health, Hebei Medical University, Shijiazhuang 050017, China
| | - Yiping Li
- Department of Occupational and Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Yaqian Qu
- Department of Occupational and Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Youliang Zhao
- Department of Occupational and Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Changfu Hao
- Department of Occupational and Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou 450001, China.
| | - Wu Yao
- Department of Occupational and Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
5
|
Jin L, Zhang X, Fan M, Li W, Zhang X. NamiRNA-mediated high expression of KNSTRN correlates with poor prognosis and immune infiltration in hepatocellular carcinoma. Contemp Oncol (Pozn) 2023; 27:163-175. [PMID: 38239867 PMCID: PMC10793618 DOI: 10.5114/wo.2023.133507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 11/12/2023] [Indexed: 01/22/2024] Open
Abstract
Introduction Mutations of kinetochore-localized astrin/sperm-associated antigen 5 (KNSTRN) can interfere with chromatid cohesion, increase aneuploidy in tumours, and enhance tumourigenesis. However, the role of the KNSTRN-binding protein in hepatocellular carcinoma (HCC) remains unclear. Material and methods Using The Cancer Genome Atlas databases, we investigated the potential oncogenic functions of KNSTRN in HCC along with R and various computational tools. Results Detailed results revealed that elevated expression of KNSTRN was considerably associated with poor overall survival (HR = 1.48, 95% CI: 1.05-2.09, p = 0.027) and progress-free interval (HR = 1.41, 95% CI: 1.05-1.89, p = 0.021) in HCC. Gene ontology/Kyoto Encyclopedia of Genes and Genomes functional enrichment analysis showed that KNSTRN is closely related to organelle fission, chromosomal region, tubulin binding, and cell cycle signalling pathway. TIMER database analysis showed the correlations between KNSTRN expression and tumour-infiltrating immune cells, biomarkers of immune cells, and immune checkpoint expression. Moreover, the KNSTRN level was significantly positively associated with immunosuppressive cells in the tumour microenvironment, including regulatory T-cells, myeloid-derived suppressor cells, and cancer-associated fibrocytes. Finally, a possible nuclear activating miRNA (NamiRNA)-enhancer network of hsa-miR-107, which activates the KNSTRN expression in liver hepatocellular carcinoma, was constructed by correlation analysis. Conclusions NamiRNA-mediated upregulation of KNSTRN correlated with poor prognosis and tumour immune infiltration in HCC. KNSTRN could serve as an effective biomarker for the diagnosis and prognosis of HCC and support the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Liang Jin
- Department of Hepatobiliary Surgery, Xijing Hospital, Air Force Medical University, Xi’an, Shaanxi, China
| | - Xiaojing Zhang
- Department of Hepatobiliary Surgery, Xijing Hospital, Air Force Medical University, Xi’an, Shaanxi, China
| | - Ming Fan
- Department of Hepatobiliary Surgery, Xijing Hospital, Air Force Medical University, Xi’an, Shaanxi, China
| | - Weimin Li
- Department of Hepatobiliary Surgery, Xijing Hospital, Air Force Medical University, Xi’an, Shaanxi, China
| | - Xuan Zhang
- Department of Hepatobiliary Surgery, Xijing Hospital, Air Force Medical University, Xi’an, Shaanxi, China
| |
Collapse
|
6
|
Meng F, Han L, Liang Q, Lu S, Huang Y, Liu J. The Lnc-RNA APPAT Suppresses Human Aortic Smooth Muscle Cell Proliferation and Migration by Interacting With MiR-647 and FGF5 in Atherosclerosis. J Endovasc Ther 2023; 30:937-950. [PMID: 35880306 DOI: 10.1177/15266028221112247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE LncRNA-Atherosclerotic plaque pathogenesis-associated transcript (APPAT) could be detected in circulating blood and has been demonstrated to correlate with the development of atherosclerosis in our previous work. It could be a potential noninvasive biomarker for earlier diagnoses of clinical cardiovascular disease. Moreover, the expression of miR-647 increased in ox-LDL-treated vascular smooth muscle cells and peripheral blood of patients with coronary heart disease. A negative correlation between APPAT and miR-647 was confirmed, and FGF5 was screened as molecular target of miR-647. However, it is largely unclear how APPAT, miR-647, and FGF5 interact and function in disease development. Here, we aim to explore the underlying molecular mechanism in this progression. MATERIALS AND METHODS APPAT, miR-647, and FGF5 expression levels were detected by quantitative reverse transcription polymerase chain reaction; cell proliferation was detected by EdU incorporation assay; cell migration was detected by wound-healing assay; the molecular interaction of APPAT/FGF5 with miR-647 was verified by dual-luciferase reporter assay; the western blot was performed to determine the gene expression at protein levels; subcellular localizations of APPAT and miR-647 were observed by fluorescence in situ hybridization; cytosolic and nucleus fractionation assay was performed to further detect the distribution of miR-647. RESULTS APPAT and miR-647 have inverse effects on human aortic smooth muscle cells' (HASMCs) proliferation and migration. APPAT negatively regulated the cell activity, whereas miR-647 did it in a positive way (p<0.05). Three pairs of molecular interplay were found: mutual negative regulation between APPAT and miR-647, APPAT downregulated FGF5, miR-647 regulation on FGF5 (p<0.05). Subcellular location assay confirmed the molecular interaction of APPAT and miR-647. CONCLUSIONS APPAT could suppress the migration and proliferation of ox-LDL-treated HASMCs via interacting with miR-647 and FGF5. We revealed a nontypical competing endogenous RNA mechanism of long noncoding RNA in the progression of atherosclerosis.
Collapse
Affiliation(s)
- Fanming Meng
- School of Basic Medical Sciences, Central South University, Changsha, People's Republic of China
| | - Luyang Han
- School of Basic Medical Sciences, Central South University, Changsha, People's Republic of China
| | - Qin Liang
- School of Basic Medical Sciences, Central South University, Changsha, People's Republic of China
| | - Shanshan Lu
- School of Basic Medical Sciences, Central South University, Changsha, People's Republic of China
| | - Yanqing Huang
- School of Basic Medical Sciences, Central South University, Changsha, People's Republic of China
| | - Junwen Liu
- School of Basic Medical Sciences, Central South University, Changsha, People's Republic of China
| |
Collapse
|
7
|
Tahamtan A, Samadizadeh S, Salimi V, Natarelli L, Nakstad B. Editorial: miRNAs and inflammation: from biogenesis to therapeutic option. Front Immunol 2023; 14:1296589. [PMID: 37854607 PMCID: PMC10579885 DOI: 10.3389/fimmu.2023.1296589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 09/22/2023] [Indexed: 10/20/2023] Open
Affiliation(s)
- Alireza Tahamtan
- Department of Microbiology, Golestan University of Medical Sciences, Gorgan, Iran
- Infectious Diseases Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Saeed Samadizadeh
- Department of Microbiology, Golestan University of Medical Sciences, Gorgan, Iran
- Infectious Diseases Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Vahid Salimi
- Department of Virology, Tehran University of Medical Sciences, Tehran, Iran
| | - Lucia Natarelli
- Institute for Cardiovascular Prevention, Ludwig-Maximillians University, Munich, Germany
| | - Britt Nakstad
- Department of Paediatrics and Adolescent Health, University of Botswana, Gaborone, Botswana
- Division of Paediatrics and Adeolescent Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
8
|
Luo ZD, Wang YF, Zhao YX, Yu LC, Li T, Fan YJ, Zeng SJ, Zhang YL, Zhang Y, Zhang X. Emerging roles of non-coding RNAs in colorectal cancer oxaliplatin resistance and liquid biopsy potential. World J Gastroenterol 2023; 29:1-18. [PMID: 36683709 PMCID: PMC9850945 DOI: 10.3748/wjg.v29.i1.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 10/11/2022] [Accepted: 11/04/2022] [Indexed: 01/04/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most common malignancies of the digestive tract, with the annual incidence and mortality increasing consistently. Oxaliplatin-based chemotherapy is a preferred therapeutic regimen for patients with advanced CRC. However, most patients will inevitably develop resistance to oxaliplatin. Many studies have reported that non-coding RNAs (ncRNAs), such as microRNAs, long non-coding RNAs, and circular RNAs, are extensively involved in cancer progression. Moreover, emerging evidence has revealed that ncRNAs mediate chemoresistance to oxaliplatin by transcriptional and post-transcriptional regulation, and by epigenetic modification. In this review, we summarize the mechanisms by which ncRNAs regulate the initiation and development of CRC chemoresistance to oxaliplatin. Furthermore, we investigate the clinical application of ncRNAs as promising biomarkers for liquid CRC biopsy. This review provides new insights into overcoming oxaliplatin resistance in CRC by targeting ncRNAs.
Collapse
Affiliation(s)
- Zheng-Dong Luo
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan 250012, Shandong Province, China
| | - Yi-Feng Wang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan 250012, Shandong Province, China
| | - Yu-Xiao Zhao
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan 250012, Shandong Province, China
| | - Long-Chen Yu
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan 250012, Shandong Province, China
| | - Tian Li
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan 250012, Shandong Province, China
| | - Ying-Jing Fan
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan 250012, Shandong Province, China
| | - Shun-Jie Zeng
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan 250012, Shandong Province, China
| | - Yan-Li Zhang
- Department of Clinical Laboratory, Shandong Provincial Third Hospital, Jinan 250012, Shandong Province, China
| | - Yi Zhang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan 250012, Shandong Province, China
| | - Xin Zhang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan 250012, Shandong Province, China
| |
Collapse
|
9
|
Hu X, Yin G, Zhang Y, Zhu L, Huang H, Lv K. Recent advances in the functional explorations of nuclear microRNAs. Front Immunol 2023; 14:1097491. [PMID: 36911728 PMCID: PMC9992549 DOI: 10.3389/fimmu.2023.1097491] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 02/10/2023] [Indexed: 02/24/2023] Open
Abstract
Approximately 22 nucleotide-long non-coding small RNAs (ncRNAs) play crucial roles in physiological and pathological activities, including microRNAs (miRNAs). Long ncRNAs often stay in the cytoplasm, modulating post-transcriptional gene expression. Briefly, miRNA binds with the target mRNA and builds a miRNA-induced silencing complex to silence the transcripts or prevent their translation. Interestingly, data from recent animal and plant studies suggested that mature miRNAs are present in the nucleus, where they regulate transcriptionally whether genes are activated or silenced. This significantly broadens the functional range of miRNAs. Here, we reviewed and summarized studies on the functions of nuclear miRNAs to better understand the modulatory networks associated with nuclear miRNAs.
Collapse
Affiliation(s)
- Xiaozhu Hu
- Central Laboratory, The First Affiliated Hospital of Wannan Medical College, Wuhu, China.,Key Laboratory of Non-Coding RNA Transformation Research of Anhui Higher Education Institutes (Wannan Medical College), Wuhu, China
| | - Guoquan Yin
- Central Laboratory, The First Affiliated Hospital of Wannan Medical College, Wuhu, China.,Key Laboratory of Non-Coding RNA Transformation Research of Anhui Higher Education Institutes (Wannan Medical College), Wuhu, China
| | - Yuan Zhang
- Central Laboratory, The First Affiliated Hospital of Wannan Medical College, Wuhu, China.,Key Laboratory of Non-Coding RNA Transformation Research of Anhui Higher Education Institutes (Wannan Medical College), Wuhu, China
| | - Liangyu Zhu
- Central Laboratory, The First Affiliated Hospital of Wannan Medical College, Wuhu, China.,Key Laboratory of Non-Coding RNA Transformation Research of Anhui Higher Education Institutes (Wannan Medical College), Wuhu, China
| | - Haoyu Huang
- Central Laboratory, The First Affiliated Hospital of Wannan Medical College, Wuhu, China.,Key Laboratory of Non-Coding RNA Transformation Research of Anhui Higher Education Institutes (Wannan Medical College), Wuhu, China
| | - Kun Lv
- Central Laboratory, The First Affiliated Hospital of Wannan Medical College, Wuhu, China.,Key Laboratory of Non-Coding RNA Transformation Research of Anhui Higher Education Institutes (Wannan Medical College), Wuhu, China
| |
Collapse
|
10
|
Li H, Da D, Yu W, Chen L, Yang S, Zhang B, Wang Y, Li L, Dang C. Tumor suppressor genes are reactivated by miR-26A1 via enhancer reprogramming in NSCLC. Hum Mol Genet 2022; 32:79-92. [PMID: 35921230 PMCID: PMC9838096 DOI: 10.1093/hmg/ddac185] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 06/23/2022] [Accepted: 08/01/2022] [Indexed: 01/25/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) is one of the most malignant epithelial tumors. Studies have suggested that DNA hypermethylation of promoters and abnormal histone modifications could induce tumor suppressor genes (TSGs) downregulation in NSCLC. However, the exact mechanism of TSGs downregulation remains unclear. In this study, we found that there is no difference in the regions of most TSGs promoters in NSCLC. Moreover, we found that there is no DNA methylation difference in the region of VILL promoter in NSCLC compared with adjacent tissue samples by pyrosequencing. We further demonstrated that VILL was markedly reactivated in A549 and H1703 cells infected with miR-26A1 lentivirus while this activation was inhibited by JQ1, an enhancer inhibitor. In addition, we identified that miR-26A1 could function as a tumor suppressor to inhibit proliferation and metastasis of NSCLC cells. Chromatin immunoprecipitation assays revealed that overexpression of miR-26A1 could significantly induce the enrichment of H3K27ac at the enhancer regions in A549 cells. To sum up, our findings revealed that enhancer-mediated TSGs regulation occured in NSCLC, suggesting that miR-26A1 could serve as a key regulator and may be a potential therapeutic target for NSCLC.
Collapse
Affiliation(s)
- Hongling Li
- To whom correspondence should be addressed at: Department of Oncology, Gansu Provincial Hospital, Lanzhou 730000, PR China. Tel: +86-0931-8281563;
| | | | | | - Lu Chen
- Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200000, PR China
| | - Shuai Yang
- Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200000, PR China
| | - Baolong Zhang
- Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200000, PR China
| | - Yongying Wang
- Department of Oncology, Gansu Provincial Hospital, The First Clinical Medical College of Gansu University of Chinese Medicine, Lanzhou 730000, PR China
| | - Linyu Li
- Department of Oncology, Gansu Provincial Hospital, The First Clinical Medical College of Gansu University of Chinese Medicine, Lanzhou 730000, PR China
| | - Chunyan Dang
- Department of Oncology, Gansu Provincial Hospital, The First Clinical Medical College of Gansu University of Chinese Medicine, Lanzhou 730000, PR China
| |
Collapse
|
11
|
Ju D, Liang Y, Hou G, Zheng W, Zhang G, Dun X, Wei D, Yan F, Zhang L, Lai D, Yuan J, Zheng Y, Wang F, Meng P, Wang Y, Yu W, Yuan J. FBP1
/miR-24-1/enhancer axis activation blocks renal cell carcinoma progression via Warburg effect. Front Oncol 2022; 12:928373. [PMID: 35978816 PMCID: PMC9376222 DOI: 10.3389/fonc.2022.928373] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 06/30/2022] [Indexed: 11/13/2022] Open
Abstract
Warburg effect is a pivotal hallmark of cancers and appears prevalently in renal cell carcinoma (RCC). FBP1 plays a negative role in Warburg effect as a rate-limiting enzyme in gluconeogenesis, yet its mechanism in RCC remains to be further characterized. Herein, we revealed that FBP1 was downregulated in RCC tissue samples and was related to the poor survival rate of RCC. Strikingly, miR-24-1 whose DNA locus is overlapped with enhancer region chr9:95084940-95087024 was closely linked with the depletion of FBP1 in RCC. Of note, miRNAs like miR-24-1 whose DNA loci are enriched with H3K27ac and H3K4me1 modifications are belonging to nuclear activating miRNAs (NamiRNAs), which surprisingly upregulate target genes in RCC through enhancer beyond the conventional role of repressing target gene expression. Moreover, miR-24-1 reactivated the expression of FBP1 to suppress Warburg effect in RCC cells, and subsequently inhibited proliferation and metastasis of RCC cells. In mechanism, the activating role of miR-24-1 was dependent on enhancer integrity by dual luciferase reporter assay and CRISPR/Cas9 system. Ultimately, animal assay in vivo validated the suppressive function of FBP1 on 786-O and ACHN cells. Collectively, the current study highlighted that activation of FBP1 by enhancer-overlapped miR-24-1 is capable of contributing to Warburg effect repression through which RCC progression is robustly blocked, providing an alternative mechanism for RCC development and as well implying a potential clue for RCC treatment strategy.
Collapse
Affiliation(s)
- Dongen Ju
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Ying Liang
- Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Public Health Clinical Center and Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China
- Department of Pharmacy, Precision Pharmacy and Drug Development Center, Tangdu Hospital, Fourth Military Medical University, Xi’an, China
| | - Guangdong Hou
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Wanxiang Zheng
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Geng Zhang
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Xinlong Dun
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Di Wei
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Fei Yan
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Lei Zhang
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Dong Lai
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Jiarui Yuan
- Clinical Medicine Department, St. George’s University School of Medicine, Saint George, Grenada
| | - Yu Zheng
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
- Medical Innovation Center, Fourth Military Medical Univeristy, Xi’an, China
| | - Fuli Wang
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Ping Meng
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Yong Wang
- Department of Urology, Tangdu Hospital, Fourth Military Medical University, Xi’an, China
- *Correspondence: Yong Wang, ; Wenqiang Yu, ; Jianlin Yuan,
| | - Wenqiang Yu
- Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Public Health Clinical Center and Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China
- *Correspondence: Yong Wang, ; Wenqiang Yu, ; Jianlin Yuan,
| | - Jianlin Yuan
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
- *Correspondence: Yong Wang, ; Wenqiang Yu, ; Jianlin Yuan,
| |
Collapse
|
12
|
miR-214-3p Protects and Restores the Myocardial Tissue of Rat Myocardial Infarction Model by Targeting PTEN. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:1175935. [PMID: 35899226 PMCID: PMC9313954 DOI: 10.1155/2022/1175935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/14/2022] [Accepted: 06/30/2022] [Indexed: 11/18/2022]
Abstract
Myocardial infarction (MI), which results in myocardial cell dysfunction and irreversible loss, is one of the most serious health threats today. This study was started with rats, by which the consequence of miRNA expression dysregulation to the occurrence and progression of cardiovascular diseases was explored. We first conducted miRNA sequencing on the myocardial tissues separately from myocardial infarction treatment and sham operation treatment to clarify those differently expressed miRNAs; then, our experiment of functional verification of those key miRNAs was initiated so as to dig out the molecular mechanism behind the miRNA's regulation in myocardial infarction. And it turned out that there were 32 upregulated miRNAs and 16 downregulated miRNAs according to our comparison from the myocardial infarction model group to the sham operation group; of all those upregulated, alteration in miR-214-3p expression was the most conspicuous. Overexpression of miR-214-3p greatly alleviated myocardial infarct area and ameliorated myocardial tissue structure, even reducing myocardial fibrosis and the devastation in the tissues. On the molecular level, miR-214-3p overexpression brought down both the apoptosis rate and cleaved caspase 3 expression. Besides that, we verified that PTEN is the target gene of miR-214-3p through a dual-luciferase assay. A cotransfection of miR-214-3p and PTEN brought about an obvious elevation in the myocardial infarct area, tissue damage, and fibrosis, even in the aspect of cellular apoptosis than a mere transfection of miR-214-3p. All the results above verified miR-214-3p′s effects in protecting myocardial tissues and reducing the infarct area, and it was reasonable to assume that those functions of miR-214-3p came into effect by targeting PTEN, which was then justified by the inversion to miR-214-3p′s protection via PTEN overexpression.
Collapse
|
13
|
Liu YY, Zhao RF, Liu C, Zhou J, Yang L, Li L. MiR-320b and miR-320d as Biomarkers to Predict and Participate in the Formation of Platinum Resistance in Ovarian Cancer Patients. Front Oncol 2022; 12:881496. [PMID: 35592674 PMCID: PMC9110861 DOI: 10.3389/fonc.2022.881496] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 04/06/2022] [Indexed: 12/24/2022] Open
Abstract
Patients with ovarian cancer who receive platinum-based chemotherapy typically develop platinum resistance, which leads to tumor recurrence and mortality. Therefore, finding the underlying mechanisms and biomarkers is critical. A total of 51 platinum-resistant and 70 platinum-sensitive ovarian cancer patients were enrolled in this study. We examined the GSE131978 dataset in the National Center for Biotechnology Information (NCBI) Gene Expression Omnibus database for differentially expressed long non-coding RNAs and messenger RNAs (mRNAs) between platinum-resistant and platinum-sensitive patients and completed a microRNA chip analysis. After filtering by Pearson correlation analysis, the competitive endogenous RNA (ceRNA) networks were subsequently constructed. Then, the Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology enrichment analyses about mRNAs in ceRNA networks were accomplished. More crucially, we demonstrated the differentially expressed microRNAs using quantitative real-time PCR and fluorescence in situ hybridization. The feasibility of microRNAs as biomarkers to predict platinum resistance and tumor recurrence was assessed using the receiver operating characteristic curve and survival analysis. MiR-320b and miR-320d exhibited high area under the curve values of 0.757 and 0.702, respectively. In our study, ceRNA networks including miR-320b and miR-320d probably provided novel insights for platinum resistance in ovarian cancer patients.
Collapse
Affiliation(s)
- Yuan-Yuan Liu
- Department of Gynecologic Oncology, Guangxi Medical University Cancer Hospital, Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Ministry of Education, Nanning, China.,Department of Gynecology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Ren-Feng Zhao
- Department of Gynecology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Chao Liu
- Department of Gynecology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Jie Zhou
- Department of Gynecology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Liu Yang
- Department of Gynecology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Li Li
- Department of Gynecologic Oncology, Guangxi Medical University Cancer Hospital, Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Ministry of Education, Nanning, China
| |
Collapse
|
14
|
Uncovering miRNA-mRNA Regulatory Modules in Developing Xylem of Pinus massoniana via Small RNA and Degradome Sequencing. Int J Mol Sci 2021; 22:ijms221810154. [PMID: 34576316 PMCID: PMC8472836 DOI: 10.3390/ijms221810154] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/12/2021] [Accepted: 09/18/2021] [Indexed: 12/21/2022] Open
Abstract
Xylem is required for the growth and development of higher plants to provide water and mineral elements. The thickening of the xylem secondary cell wall (SCW) not only improves plant survival, but also provides raw materials for industrial production. Numerous studies have found that transcription factors and non-coding RNAs regulate the process of SCW thickening. Pinus massoniana is an important woody tree species in China and is widely used to produce materials for construction, furniture, and packaging. However, the target genes of microRNAs (miRNAs) in the developing xylem of P. massoniana are not known. In this study, a total of 25 conserved miRNAs and 173 novel miRNAs were identified via small RNA sequencing, and 58 differentially expressed miRNAs were identified between the developing xylem (PM_X) and protoplasts isolated from the developing xylem (PM_XP); 26 of these miRNAs were significantly up-regulated in PM_XP compared with PM_X, and 32 were significantly down-regulated. A total of 153 target genes of 20 conserved miRNAs and 712 target genes of 113 novel miRNAs were verified by degradome sequencing. There may be conserved miRNA-mRNA modules (miRNA-MYB, miRNA-ARF, and miRNA-LAC) involved in softwood and hardwood formation. The results of qRT-PCR-based parallel validation were in relatively high agreement. This study explored the potential regulatory network of miRNAs in the developing xylem of P. massoniana and provides new insights into wood formation in coniferous species.
Collapse
|
15
|
Liang Y, Lu Q, Li W, Zhang D, Zhang F, Zou Q, Chen L, Tong Y, Liu M, Wang S, Li W, Ren X, Xu P, Yang Z, Dong S, Zhang B, Huang Y, Li D, Wang H, Yu W. Reactivation of tumour suppressor in breast cancer by enhancer switching through NamiRNA network. Nucleic Acids Res 2021; 49:8556-8572. [PMID: 34329471 PMCID: PMC8421228 DOI: 10.1093/nar/gkab626] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 06/08/2021] [Accepted: 07/17/2021] [Indexed: 12/31/2022] Open
Abstract
Dysfunction of Tumour Suppressor Genes (TSGs) is a common feature in carcinogenesis. Epigenetic abnormalities including DNA hypermethylation or aberrant histone modifications in promoter regions have been described for interpreting TSG inactivation. However, in many instances, how TSGs are silenced in tumours are largely unknown. Given that miRNA with low expression in tumours is another recognized signature, we hypothesize that low expression of miRNA may reduce the activity of TSG related enhancers and further lead to inactivation of TSG during cancer development. Here, we reported that low expression of miRNA in cancer as a recognized signature leads to loss of function of TSGs in breast cancer. In 157 paired breast cancer and adjacent normal samples, tumour suppressor gene GPER1 and miR-339 are both downregulated in Luminal A/B and Triple Negative Breast Cancer subtypes. Mechanistic investigations revealed that miR-339 upregulates GPER1 expression in breast cancer cells by switching on the GPER1 enhancer, which can be blocked by enhancer deletion through the CRISPR/Cas9 system. Collectively, our findings reveal novel mechanistic insights into TSG dysfunction in cancer development, and provide evidence that reactivation of TSG by enhancer switching may be a promising alternative strategy for clinical breast cancer treatment.
Collapse
Affiliation(s)
- Ying Liang
- Shanghai Public Health Clinical Center and Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute and Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, P. R. China
| | - Qi Lu
- Department of Gynaecology, Jinshan Hospital of Fudan University, Shanghai 201508, P. R. China
| | - Wei Li
- Shanghai Public Health Clinical Center and Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute and Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, P. R. China
| | - Dapeng Zhang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, P. R. China
| | - Fanglin Zhang
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, P. R. China
| | - Qingping Zou
- Shanghai Public Health Clinical Center and Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute and Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, P. R. China
| | - Lu Chen
- Shanghai Public Health Clinical Center and Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute and Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, P. R. China
| | - Ying Tong
- Shanghai Public Health Clinical Center and Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute and Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, P. R. China
| | - Mengxing Liu
- Shanghai Public Health Clinical Center and Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute and Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, P. R. China
| | - Shaoxuan Wang
- Shanghai Public Health Clinical Center and Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute and Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, P. R. China
| | - Wenxuan Li
- Shanghai Public Health Clinical Center and Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute and Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, P. R. China
| | - Xiaoguang Ren
- Shanghai Public Health Clinical Center and Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute and Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, P. R. China
| | - Peng Xu
- Shanghai Public Health Clinical Center and Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute and Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, P. R. China
| | - Zhicong Yang
- Shanghai Public Health Clinical Center and Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute and Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, P. R. China
| | - Shihua Dong
- Shanghai Public Health Clinical Center and Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute and Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, P. R. China
| | - Baolong Zhang
- Shanghai Public Health Clinical Center and Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute and Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, P. R. China
| | - Yanni Huang
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, P. R. China
| | - Daqiang Li
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, P. R. China
| | - Hailin Wang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, P. R. China
| | - Wenqiang Yu
- Shanghai Public Health Clinical Center and Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute and Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, P. R. China
| |
Collapse
|
16
|
Zhong J, Han C, Zhang X, Chen P, Liu R. scGET: Predicting Cell Fate Transition During Early Embryonic Development by Single-cell Graph Entropy. GENOMICS, PROTEOMICS & BIOINFORMATICS 2021; 19:461-474. [PMID: 34954425 PMCID: PMC8864248 DOI: 10.1016/j.gpb.2020.11.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 11/08/2020] [Accepted: 01/02/2021] [Indexed: 01/26/2023]
Abstract
During early embryonic development, cell fate commitment represents a critical transition or "tipping point" of embryonic differentiation, at which there is a drastic and qualitative shift of the cell populations. In this study, we presented a computational approach, scGET, to explore the gene-gene associations based on single-cell RNA sequencing (scRNA-seq) data for critical transition prediction. Specifically, by transforming the gene expression data to the local network entropy, the single-cell graph entropy (SGE) value quantitatively characterizes the stability and criticality of gene regulatory networks among cell populations and thus can be employed to detect the critical signal of cell fate or lineage commitment at the single-cell level. Being applied to five scRNA-seq datasets of embryonic differentiation, scGET accurately predicts all the impending cell fate transitions. After identifying the "dark genes" that are non-differentially expressed genes but sensitive to the SGE value, the underlying signaling mechanisms were revealed, suggesting that the synergy of dark genes and their downstream targets may play a key role in various cell development processes.The application in all five datasets demonstrates the effectiveness of scGET in analyzing scRNA-seq data from a network perspective and its potential to track the dynamics of cell differentiation. The source code of scGET is accessible at https://github.com/zhongjiayuna/scGET_Project.
Collapse
Affiliation(s)
- Jiayuan Zhong
- School of Mathematics, South China University of Technology, Guangzhou 510640, PR China
| | - Chongyin Han
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510640, PR China
| | - Xuhang Zhang
- School of Computer Science and Engineering, South China University of Technology, Guangzhou 510640, PR China
| | - Pei Chen
- School of Mathematics, South China University of Technology, Guangzhou 510640, PR China.
| | - Rui Liu
- School of Mathematics, South China University of Technology, Guangzhou 510640, PR China; Pazhou Lab, Guangzhou 510330, PR China.
| |
Collapse
|
17
|
Liang Y, Xu P, Zou Q, Luo H, Yu W. An epigenetic perspective on tumorigenesis: Loss of cell identity, enhancer switching, and NamiRNA network. Semin Cancer Biol 2018; 57:1-9. [PMID: 30213688 DOI: 10.1016/j.semcancer.2018.09.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 08/26/2018] [Accepted: 09/06/2018] [Indexed: 02/09/2023]
Abstract
Various tumorigenic theories have been proposed in the past century, which contribute to the prevention and treatment of cancer clinically. However, the underlying mechanisms of the initiation of cancer, drug resistance, neoplasm relapse, and metastasis are still challenging to be panoramically addressed. Based on the abundant evidence provided by others and us, we postulate that Tumor Initiated by Loss of Cell Identity (LOCI), which is an inevitable initiating event of tumorigenesis. As a result, normal cells are transformed into the cancerous cell. In this process, epigenetic regulatory program, especially NamiRNA (Nuclear activating miRNA)-enhancer-gene activation network, is vital for the cell identity. The disorganization of NamiRNA-enhancer-gene activation network is a causal predisposition to the cell identity loss, and the altered cell identity is stabilized by genetic variations of the NamiRNA-enhancer-gene activation network. Furthermore, the additional genetic or epigenetic abnormities confer those cells to carcinogenic characteristics, such as growth advantage over normal cells, and finally yield cancer. In this review, we literally explain our tumor initiation hypothesis based on the corresponding evidence, which will not only help to refresh our understanding of tumorigenesis but also bring benefits to developing "cell identity reversing" based therapies.
Collapse
Affiliation(s)
- Ying Liang
- Shanghai Public Health Clinical Center & Laboratory of RNA Epigenetics, Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 201508, China; Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai 200032, China; Department of Biochemistry and Molecular Biology, Shanghai Medical College, MOE Key Laboratory of Metabolism and Molecular Medicine, Department of Molecular Biology, Fudan University, Shanghai, 200032, China; Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, 200433, China
| | - Peng Xu
- Shanghai Public Health Clinical Center & Laboratory of RNA Epigenetics, Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 201508, China; Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai 200032, China; Department of Biochemistry and Molecular Biology, Shanghai Medical College, MOE Key Laboratory of Metabolism and Molecular Medicine, Department of Molecular Biology, Fudan University, Shanghai, 200032, China; Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, 200433, China
| | - Qingping Zou
- Shanghai Public Health Clinical Center & Laboratory of RNA Epigenetics, Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 201508, China; Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai 200032, China; Department of Biochemistry and Molecular Biology, Shanghai Medical College, MOE Key Laboratory of Metabolism and Molecular Medicine, Department of Molecular Biology, Fudan University, Shanghai, 200032, China; Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, 200433, China
| | - Huaibing Luo
- Shanghai Public Health Clinical Center & Laboratory of RNA Epigenetics, Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 201508, China; Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai 200032, China; Department of Biochemistry and Molecular Biology, Shanghai Medical College, MOE Key Laboratory of Metabolism and Molecular Medicine, Department of Molecular Biology, Fudan University, Shanghai, 200032, China; Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, 200433, China
| | - Wenqiang Yu
- Shanghai Public Health Clinical Center & Laboratory of RNA Epigenetics, Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 201508, China; Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai 200032, China; Department of Biochemistry and Molecular Biology, Shanghai Medical College, MOE Key Laboratory of Metabolism and Molecular Medicine, Department of Molecular Biology, Fudan University, Shanghai, 200032, China; Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, 200433, China.
| |
Collapse
|
18
|
Liang Y, Xu P, Zou Q, Luo H, Yu W. An epigenetic perspective on tumorigenesis: Loss of cell identity, enhancer switching, and NamiRNA network. Semin Cancer Biol 2018; 83:596-604. [PMID: 30208341 DOI: 10.1016/j.semcancer.2018.09.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 09/07/2018] [Indexed: 02/09/2023]
Abstract
Various tumorigenic theories have been proposed in the past century, which contribute to the prevention and treatment of cancer clinically. However, the underlying mechanisms of the initiation of cancer, drug resistance, neoplasm relapse, and metastasis are still challenging to be panoramically addressed. Based on the abundant evidence provided by others and us, we postulate that Tumor Initiated by Loss of Cell Identity (LOCI), which is an inevitable initiating event of tumorigenesis. As a result, normal cells are transformed into the cancerous cell. In this process, epigenetic regulatory program, especially NamiRNA (Nuclear activating miRNA)-enhancer-gene activation network, is vital for the cell identity. The disorganization of NamiRNA-enhancer-gene activation network is a causal predisposition to the cell identity loss, and the altered cell identity is stabilized by genetic variations of the NamiRNA-enhancer-gene activation network. Furthermore, the additional genetic or epigenetic abnormities confer those cells to carcinogenic characteristics, such as growth advantage over normal cells, and finally yield cancer. In this review, we literally explain our tumor imitation hypothesis based on the corresponding evidence, which will not only help to refresh our understanding of tumorigenesis but also bring benefits to developing "cell identity reversing" based therapies.
Collapse
Affiliation(s)
- Ying Liang
- Shanghai Public Health Clinical Center & Laboratory of RNA Epigenetics, Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 201508, China; Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai 200032, China; Department of Biochemistry and Molecular Biology, Shanghai Medical College, MOE Key Laboratory of Metabolism and Molecular Medicine, Department of Molecular Biology, Fudan University, Shanghai, 200032, China; Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, 200433, China
| | - Peng Xu
- Shanghai Public Health Clinical Center & Laboratory of RNA Epigenetics, Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 201508, China; Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai 200032, China; Department of Biochemistry and Molecular Biology, Shanghai Medical College, MOE Key Laboratory of Metabolism and Molecular Medicine, Department of Molecular Biology, Fudan University, Shanghai, 200032, China; Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, 200433, China
| | - Qingping Zou
- Shanghai Public Health Clinical Center & Laboratory of RNA Epigenetics, Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 201508, China; Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai 200032, China; Department of Biochemistry and Molecular Biology, Shanghai Medical College, MOE Key Laboratory of Metabolism and Molecular Medicine, Department of Molecular Biology, Fudan University, Shanghai, 200032, China; Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, 200433, China
| | - Huaibing Luo
- Shanghai Public Health Clinical Center & Laboratory of RNA Epigenetics, Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 201508, China; Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai 200032, China; Department of Biochemistry and Molecular Biology, Shanghai Medical College, MOE Key Laboratory of Metabolism and Molecular Medicine, Department of Molecular Biology, Fudan University, Shanghai, 200032, China; Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, 200433, China
| | - Wenqiang Yu
- Shanghai Public Health Clinical Center & Laboratory of RNA Epigenetics, Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 201508, China; Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai 200032, China; Department of Biochemistry and Molecular Biology, Shanghai Medical College, MOE Key Laboratory of Metabolism and Molecular Medicine, Department of Molecular Biology, Fudan University, Shanghai, 200032, China; Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, 200433, China.
| |
Collapse
|