1
|
Qian Q, Lyu H, Wang W, Wang Q, Li D, Liu X, He Y, Shen M. Combined transcriptomic and proteomic analyses reveal relevant myelin features in mice with ischemic stroke. Funct Integr Genomics 2025; 25:64. [PMID: 40085348 PMCID: PMC11909235 DOI: 10.1007/s10142-025-01573-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 02/24/2025] [Accepted: 03/05/2025] [Indexed: 03/16/2025]
Abstract
Ischemic stroke (IS), a leading cause of global disability and mortality, is characterized by white matter damage and demyelination. Despite advances, the molecular mechanisms driving post-IS myelin pathology remain poorly understood, limiting therapeutic development. This study investigates key myelin-related genes (MRGs) and their regulatory networks to identify novel therapeutic targets. A transient middle cerebral artery occlusion (MCAO) model was established in C57BL/6 mice, with brain tissues collected at four timepoints (Sham0D, MCAO0D, MCAO7D, MCAO14D). Transcriptomic and proteomic sequencing were performed, followed by soft clustering (Mfuzz), functional enrichment (GO/KEGG), and ROC analysis to identify key MRGs. Competing endogenous RNA (ceRNA) networks were constructed, and drug prediction was conducted using the Comparative Toxicogenomics Database (CTD) and molecular docking. Expression validation was performed via qRT-PCR and Western blot. Integrated multi-omics analysis identified Wasf3 and Slc25a5 as key MRGs, enriched in mitochondrial respiration, calcium metabolism, and cytoskeletal regulation. The AUC values of the one-to-one model scores were all greater than 0.7, suggesting that Wasf3 and Slc25a5 were able to effectively discriminate between samples from different time points. A ceRNA network revealed critical interactions, including the Wasf3-mmu-miR-423-5p-H19 axis, linking apoptosis and myelin dysfunction. Drug prediction highlighted valproic acid (VPA) as a high-affinity binder for both genes (binding energies: - 4.2 and - 4.7 kcal/mol), suggesting its potential as a therapeutic candidate for IS. Experimental validation confirmed significant downregulation of Wasf3 mRNA (p < 0.01) and protein (p = 0.069) post-IS, while Slc25a5 showed no significant changes, potentially due to sample size limitations. This study establishes Wasf3 and Slc25a5 as pivotal regulators of post-IS myelin pathology and proposes VPA as a promising therapeutic candidate to enhance remyelination. The findings underscore the utility of multi-omics approaches in bridging molecular mechanisms to clinical translation, offering new strategies for IS diagnosis and treatment.
Collapse
Affiliation(s)
- Qiuyang Qian
- Department of Rehabilitation Medicine, People's Hospital of Longhua, No 38 Jinglong Construction Road, Shenzhen, 518109, Longhua District, China
| | - Hao Lyu
- Department of Neurosurgery, Shenzhen Second People'S Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, 518000, China
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Wei Wang
- Department of Rehabilitation Medicine, People's Hospital of Longhua, No 38 Jinglong Construction Road, Shenzhen, 518109, Longhua District, China
| | - Qiwen Wang
- Department of Rehabilitation Medicine, People's Hospital of Longhua, No 38 Jinglong Construction Road, Shenzhen, 518109, Longhua District, China
| | - Desheng Li
- Department of Rehabilitation Medicine, People's Hospital of Longhua, No 38 Jinglong Construction Road, Shenzhen, 518109, Longhua District, China
| | - Xiaojia Liu
- Department of Neurosurgery, Shenzhen Second People'S Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, 518000, China
| | - Yi He
- Department of Neurosurgery, Shenzhen Second People'S Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, 518000, China
| | - Mei Shen
- Department of Rehabilitation Medicine, People's Hospital of Longhua, No 38 Jinglong Construction Road, Shenzhen, 518109, Longhua District, China.
| |
Collapse
|
2
|
Sun R, Zhang X, Gong T, Zhang Y, Wang Q, He C, Ju J, Jin C, Ding W, Gao J, Shen J, Li Q, Shan Z. Knockdown H19 Accelerated iPSCs Reprogramming through Epigenetic Modifications and Mesenchymal-to-Epithelial Transition. Biomolecules 2024; 14:509. [PMID: 38785917 PMCID: PMC11118134 DOI: 10.3390/biom14050509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/11/2024] [Accepted: 04/15/2024] [Indexed: 05/25/2024] Open
Abstract
H19 is an essential imprinted gene that is expressed to govern normal embryonic development. During reprogramming, the parental pronuclei have asymmetric reprogramming capacities and the critical reprogramming factors predominantly reside in the male pronucleus. After inhibiting the expression of H19 and Gtl2, androgenetic haploid ESCs (AG-haESCs) can efficiently and stably support the generation of healthy SC pups at a rate of ~20%, and double-knockout parthenogenetic haESCs can also produce efficiently. Induced pluripotent stem (iPS) cell reprogramming is thought to have a characteristic epigenetic pattern that is the reverse of its developmental potential; however, it is unclear how H19 participates in iPS cell reprogramming. Here, we showed that the expression of H19 was transiently increased during iPSC reprogramming. H19 knockdown resulted in greater reprogramming efficiency. The genes associated with pluripotency showed enhanced expression during the early reprogramming process, and the Oct4 promoter was demethylated by bisulfite genomic sequencing analysis. Moreover, expression analysis revealed that the mesenchymal master regulators associated with epithelial-to-mesenchymal transition (EMT) were downregulated during reprogramming in H19 knockdown. These findings provide functional insight into the role of H19 as a barrier to the early reprogramming process.
Collapse
Affiliation(s)
- Ruizhen Sun
- Department of Histology and Embryology, Harbin Medical University, Harbin 150081, China; (R.S.); (X.Z.); (T.G.); (Y.Z.); (Q.W.); (C.H.); (J.J.); (C.J.); (W.D.); (J.G.)
| | - Ximei Zhang
- Department of Histology and Embryology, Harbin Medical University, Harbin 150081, China; (R.S.); (X.Z.); (T.G.); (Y.Z.); (Q.W.); (C.H.); (J.J.); (C.J.); (W.D.); (J.G.)
| | - Tiantian Gong
- Department of Histology and Embryology, Harbin Medical University, Harbin 150081, China; (R.S.); (X.Z.); (T.G.); (Y.Z.); (Q.W.); (C.H.); (J.J.); (C.J.); (W.D.); (J.G.)
| | - Yue Zhang
- Department of Histology and Embryology, Harbin Medical University, Harbin 150081, China; (R.S.); (X.Z.); (T.G.); (Y.Z.); (Q.W.); (C.H.); (J.J.); (C.J.); (W.D.); (J.G.)
| | - Qi Wang
- Department of Histology and Embryology, Harbin Medical University, Harbin 150081, China; (R.S.); (X.Z.); (T.G.); (Y.Z.); (Q.W.); (C.H.); (J.J.); (C.J.); (W.D.); (J.G.)
| | - Chenyao He
- Department of Histology and Embryology, Harbin Medical University, Harbin 150081, China; (R.S.); (X.Z.); (T.G.); (Y.Z.); (Q.W.); (C.H.); (J.J.); (C.J.); (W.D.); (J.G.)
| | - Jielan Ju
- Department of Histology and Embryology, Harbin Medical University, Harbin 150081, China; (R.S.); (X.Z.); (T.G.); (Y.Z.); (Q.W.); (C.H.); (J.J.); (C.J.); (W.D.); (J.G.)
| | - Chunmiao Jin
- Department of Histology and Embryology, Harbin Medical University, Harbin 150081, China; (R.S.); (X.Z.); (T.G.); (Y.Z.); (Q.W.); (C.H.); (J.J.); (C.J.); (W.D.); (J.G.)
| | - Wenxin Ding
- Department of Histology and Embryology, Harbin Medical University, Harbin 150081, China; (R.S.); (X.Z.); (T.G.); (Y.Z.); (Q.W.); (C.H.); (J.J.); (C.J.); (W.D.); (J.G.)
| | - Jingnan Gao
- Department of Histology and Embryology, Harbin Medical University, Harbin 150081, China; (R.S.); (X.Z.); (T.G.); (Y.Z.); (Q.W.); (C.H.); (J.J.); (C.J.); (W.D.); (J.G.)
| | - Jingling Shen
- Institute of Life Sciences, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China;
| | - Qiuming Li
- Department of Histology and Embryology, Harbin Medical University, Harbin 150081, China; (R.S.); (X.Z.); (T.G.); (Y.Z.); (Q.W.); (C.H.); (J.J.); (C.J.); (W.D.); (J.G.)
| | - Zhiyan Shan
- Department of Histology and Embryology, Harbin Medical University, Harbin 150081, China; (R.S.); (X.Z.); (T.G.); (Y.Z.); (Q.W.); (C.H.); (J.J.); (C.J.); (W.D.); (J.G.)
| |
Collapse
|
3
|
Xia Y, Pei T, Zhao J, Wang Z, Shen Y, Yang Y, Liang J. Long noncoding RNA H19: functions and mechanisms in regulating programmed cell death in cancer. Cell Death Discov 2024; 10:76. [PMID: 38355574 PMCID: PMC10866971 DOI: 10.1038/s41420-024-01832-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 01/16/2024] [Accepted: 01/25/2024] [Indexed: 02/16/2024] Open
Abstract
Long noncoding RNAs (lncRNAs) are a group of noncoding RNAs with transcript lengths of >200 nucleotides. Mounting evidence suggests that lncRNAs are closely associated with tumorigenesis. LncRNA H19 (H19) was the first lncRNA to function as an oncogene in many malignant tumors. Apart from the established role of H19 in promoting cell growth, proliferation, invasion, migration, epithelial-mesenchymal transition (EMT), and metastasis, it has been recently discovered that H19 also inhibits programmed cell death (PCD) of cancer cells. In this review, we summarize the mechanisms by which H19 regulates PCD in cancer cells through various signaling pathways, molecular mechanisms, and epigenetic modifications. H19 regulates PCD through the Wnt/β-catenin pathway and the PI3K-Akt-mTOR pathway. It also acts as a competitive endogenous RNA (ceRNA) in PCD regulation. The interaction between H19 and RNA-binding proteins (RBP) regulates apoptosis in cancer. Moreover, epigenetic modifications, including DNA and RNA methylation and histone modifications, are also involved in H19-associated PCD regulation. In conclusion, we summarize the role of H19 signaling via PCD in cancer chemoresistance, highlighting the promising research significance of H19 as a therapeutic target. We hope that our study will contribute to a broader understanding of H19 in cancer development and treatment.
Collapse
Affiliation(s)
- Yuyang Xia
- Department of Urology, Institute of Urology, West China Hospital, West China School of Medicine, Sichuan University, 610041, Chengdu, China
| | - Tianjiao Pei
- Department of Reproductive Medicine, West China Second University Hospital of Sichuan University, Chengdu, China.
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital of Sichuan University, Chengdu, China.
| | - Junjie Zhao
- Department of Urology, Institute of Urology, West China Hospital, West China School of Medicine, Sichuan University, 610041, Chengdu, China
| | - Zilin Wang
- Department of Urology, Institute of Urology, West China Hospital, West China School of Medicine, Sichuan University, 610041, Chengdu, China
| | - Yu Shen
- Department of Urology, Institute of Urology, West China Hospital, West China School of Medicine, Sichuan University, 610041, Chengdu, China
| | - Yang Yang
- Department of Urology, Institute of Urology, West China Hospital, West China School of Medicine, Sichuan University, 610041, Chengdu, China
| | - Jiayu Liang
- Department of Urology, Institute of Urology, West China Hospital, West China School of Medicine, Sichuan University, 610041, Chengdu, China.
| |
Collapse
|
4
|
Jin J, Zhong XB. Epigenetic Mechanisms Contribute to Intraindividual Variations of Drug Metabolism Mediated by Cytochrome P450 Enzymes. Drug Metab Dispos 2023; 51:672-684. [PMID: 36973001 PMCID: PMC10197210 DOI: 10.1124/dmd.122.001007] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 02/24/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023] Open
Abstract
Significant interindividual and intraindividual variations on cytochrome P450 (CYP)-mediated drug metabolism exist in the general population globally. Genetic polymorphisms are one of the major contribution factors for interindividual variations, but epigenetic mechanisms mainly contribute to intraindividual variations, including DNA methylation, histone modifications, microRNAs, and long non-coding RNAs. The current review provides analysis of advanced knowledge in the last decade on contributions of epigenetic mechanisms to intraindividual variations on CYP-mediated drug metabolism in several situations, including (1) ontogeny, the developmental changes of CYP expression in individuals from neonates to adults; (2) increased activities of CYP enzymes induced by drug treatment; (3) increased activities of CYP enzymes in adult ages induced by drug treatment at neonate ages; and (4) decreased activities of CYP enzymes in individuals with drug-induced liver injury (DILI). Furthermore, current challenges, knowledge gaps, and future perspective of the epigenetic mechanisms in development of CYP pharmacoepigenetics are discussed. In conclusion, epigenetic mechanisms have been proven to contribute to intraindividual variations of drug metabolism mediated by CYP enzymes in age development, drug induction, and DILI conditions. The knowledge has helped understanding how intraindividual variation are generated. Future studies are needed to develop CYP-based pharmacoepigenetics to guide clinical applications for precision medicine with improved therapeutic efficacy and reduced risk of adverse drug reactions and toxicity. SIGNIFICANCE STATEMENT: Understanding epigenetic mechanisms in contribution to intraindividual variations of CYP-mediated drug metabolism may help to develop CYP-based pharmacoepigenetics for precision medicine to improve therapeutic efficacy and reduce adverse drug reactions and toxicity for drugs metabolized by CYP enzymes.
Collapse
Affiliation(s)
- Jing Jin
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut
| | - Xiao-Bo Zhong
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut
| |
Collapse
|
5
|
Abdelsattar S, Sweed D, Kamel HFM, Kasemy ZA, Gameel AM, Elzohry H, Ameen O, Elgizawy EI, Sallam A, Mosbeh A, Abdallah MS, Khalil FO, Al-Amodi HS, El-Hefnway SM. The Potential Utility of Circulating Oncofetal H19 Derived miR-675 Expression versus Tissue lncRNA-H19 Expression in Diagnosis and Prognosis of HCC in Egyptian Patients. Biomolecules 2022; 13:biom13010003. [PMID: 36671388 PMCID: PMC9856163 DOI: 10.3390/biom13010003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/28/2022] [Accepted: 12/06/2022] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common cancers worldwide. Interestingly, lncRNA-H19 acts independently in HCC and influences miR-675 expressions. We aimed to assess the potential utility of tissue lncRNA-H19 versus miR-675 expressions as a non-invasive biomarker for HCC diagnosis and prognosis in Egyptian patients. Ninety-one HCC patients and 91 controls included in this study were investigated for expression of lncRNA-H19 and miR675 using RT-qPCR. Our results showed that the expression of lncRNA-H19 and microRNA-675 were higher in patients than in controls (p < 0.001 for both). Additionally, lncRNA-H19 expression was higher in tumorous than in non-tumorous tissue (p < 0.001). Linear regression revealed that miR-675 expression was a significantly higher positive predictor than lncRNA-H19 for tumor size, pathologic grade, and AFP level; similarly, for cyclin D1 and VEGF protein expression. By using the ROC curve, the sensitivity of miR-675 was higher than lncRNA-H19 for discriminating HCC from controls (95-89%, respectively) and the sensitivity of lncRNA-H19 was higher in tumorous than in non-tumorous tissues (76%). The high expressions of both were associated with low OS (p < 0.001, 0.001, respectively). Oncofetal H19-derived miR-675 expression could be considered a potential noninvasive diagnostic and prognostic biomarker, outstanding the performance of the expression of tissue lncRNA-H19 for HCC.
Collapse
Affiliation(s)
- Shimaa Abdelsattar
- Clinical Biochemistry and Molecular Diagnostics Department, National Liver Institute, Menofia University, Shebin El-Kom 32511, Egypt
- Correspondence:
| | - Dina Sweed
- Pathology Department, National Liver Institute, Menofia University, Shebin El-Kom 32511, Egypt
| | - Hala F. M. Kamel
- Department of Biochemistry, Faculty of Medicine, Umm Al Qura University, Makka 21955, Saudi Arabia
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Ain Shams University, Cairo 11566, Egypt
| | - Zeinab A. Kasemy
- Public Health and Community Medicine Department, Faculty of Medicine, Menofia University, Shebin El-Kom 32511, Egypt
| | - Abdallah M. Gameel
- Clinical Pathology Department, National Cancer Institute, Cairo University, Cairo 11796, Egypt
| | - Hassan Elzohry
- Hepatology and Gastroenterology Department, National Liver Institute, Menofia University, Shebin El-Kom 32511, Egypt
| | - Omnia Ameen
- Physiology Department, Faculty of Medicine, Menofia University, Shebin El-Kom 32511, Egypt
| | - Eman Ibrahim Elgizawy
- Physiology Department, Faculty of Medicine, Menofia University, Shebin El-Kom 32511, Egypt
| | - Ahmed Sallam
- Department of Hepatobiliary and Pancreatic Surgery, National Liver Institute, Menofia University, Shebin El-Kom 32511, Egypt
| | - Asmaa Mosbeh
- Pathology Department, National Liver Institute, Menofia University, Shebin El-Kom 32511, Egypt
| | - Mahmoud S. Abdallah
- Clinical Pharmacy Department, Faculty of Pharmacy, University of Sadat City (USC), Sadat City 32897, Egypt
| | - Fatma O. Khalil
- Clinical and Molecular Microbiology and Immunology Department, National Liver Institute, Menoufia University, Shebin El-Kom 32511, Egypt
| | - Hiba S. Al-Amodi
- Department of Biochemistry, Faculty of Medicine, Umm Al Qura University, Makka 21955, Saudi Arabia
| | - Sally M. El-Hefnway
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Menofia University, Shebin El-Kom 32511, Egypt
| |
Collapse
|
6
|
Xu J, Hao S, Shi Q, Deng Q, Jiang Y, Guo P, Yuan Y, Shi X, Shangguan S, Zheng H, Lai G, Huang Y, Wang Y, Song Y, Liu Y, Wu L, Wang Z, Cheng J, Wei X, Cheng M, Lai Y, Volpe G, Esteban MA, Hou Y, Liu C, Liu L. Transcriptomic Profile of the Mouse Postnatal Liver Development by Single-Nucleus RNA Sequencing. Front Cell Dev Biol 2022; 10:833392. [PMID: 35465320 PMCID: PMC9019599 DOI: 10.3389/fcell.2022.833392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 03/11/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- Jiangshan Xu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- BGI-Shenzhen, Shenzhen, China
| | - Shijie Hao
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- BGI-Shenzhen, Shenzhen, China
| | - Quan Shi
- BGI-Shenzhen, Shenzhen, China
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Qiuting Deng
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- BGI-Shenzhen, Shenzhen, China
| | - Yujia Jiang
- BGI-Shenzhen, Shenzhen, China
- BGI College and Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Pengcheng Guo
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
- Laboratory of Integrative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Yue Yuan
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- BGI-Shenzhen, Shenzhen, China
| | - Xuyang Shi
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- BGI-Shenzhen, Shenzhen, China
| | - Shuncheng Shangguan
- Laboratory of Integrative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Joint School of Life Sciences, Guangzhou Medical University and Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Huiwen Zheng
- BGI-Shenzhen, Shenzhen, China
- BGI College and Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Guangyao Lai
- Laboratory of Integrative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Joint School of Life Sciences, Guangzhou Medical University and Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | | | | | | | | | - Liang Wu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- BGI-Shenzhen, Shenzhen, China
| | | | - Jiehui Cheng
- Guangdong Hospital of Traditional Chinese Medicine, Zhuhai, China
| | | | - Mengnan Cheng
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- BGI-Shenzhen, Shenzhen, China
| | - Yiwei Lai
- Laboratory of Integrative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Giacomo Volpe
- Hematology and Cell Therapy Unit, IRCCS-Istituto Tumori‘Giovanni Paolo II’, Bari, Italy
| | - Miguel A. Esteban
- BGI-Shenzhen, Shenzhen, China
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
- Laboratory of Integrative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | | | | | - Longqi Liu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- BGI-Shenzhen, Shenzhen, China
| |
Collapse
|
7
|
Khan H, Ni Z, Feng H, Xing Y, Wu X, Huang D, Chen L, Niu Y, Shi G. Combination of curcumin with N-n-butyl haloperidol iodide inhibits hepatocellular carcinoma malignant proliferation by downregulating enhancer of zeste homolog 2 (EZH2) - lncRNA H19 to silence Wnt/β-catenin signaling. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 91:153706. [PMID: 34517264 DOI: 10.1016/j.phymed.2021.153706] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 08/09/2021] [Accepted: 08/10/2021] [Indexed: 02/05/2023]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is one of the most common cause of cancer-related death worldwide. Curcumin (C) has been extensively investigated in different types of malignancies, including hepatocellular carcinoma, but its physicochemical properties have significantly influenced its clinical use. Several approaches are being explored to enhance curcumin's therapeutic response, including its combination with various drugs. PURPOSE This study aimed to evaluate the anti-tumor effect of curcumin (C) in combination with F2 (N-n-butyl haloperidol iodide) on hepatocellular carcinoma and its potential underlying mechanism in vitro and in vivo. METHODS Cell proliferation was evaluated by CCK-8 and colony formation assays, and apoptosis was measured by flow cytometry. The migratory and invasive abilities of Hep3B and SMMC-7721 cells were measured by wound-healing and matrigel transwell assays. In order to investigate the molecular pathways, various experiments such as western blotting, qPCR, RNA-seq, immunostaining and transfection were performed. To evaluate the anti-HCC effects in vivo, a xenograft tumor model was used. RESULTS Our findings showed that the combination of curcumin (C) & F2 (F2C) strongly inhibited malignant proliferation and migration in SMMC-7721 and Hep3B cells. The F2C treatment downregulates enhancer of zeste homolog 2 (EZH2) transcription and protein expression, which is key epigenetic regulator responsible for HCC development. Moreover, the inhibition of EZH2 by F2C led to Wnt/β-catenin signaling inhibition by decreasing tri-methylation of histone H3 at lysine 27 (H3K27me3) and long non-coding RNA H19 expression. The inhibition of F2C was associated with the suppression of tumorigenicity in xenograft HCC models. CONCLUSION These findings suggested that, F2C inhibited HCC formation, migration and its modulatory mechanism seemed to be associated with downregulation of EZH2, silencing Wnt/β-catenin signaling by interacting with H19, suggesting that F2C may be a promising drug in the clinical treatment of HCC.
Collapse
Affiliation(s)
- Hanif Khan
- Department of Pharmacology, Shantou University Medical College, No. 22 Xinling Road, Shantou 515041, Guangdong, China
| | - Zhengzhong Ni
- Department of Pharmacology, Shantou University Medical College, No. 22 Xinling Road, Shantou 515041, Guangdong, China
| | - Hai Feng
- Shanghai Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yaqi Xing
- Department of Pharmacology, Shantou University Medical College, No. 22 Xinling Road, Shantou 515041, Guangdong, China
| | - Xuejun Wu
- Department of Pharmacology, Shantou University Medical College, No. 22 Xinling Road, Shantou 515041, Guangdong, China
| | - Danmei Huang
- Department of Pharmacology, Shantou University Medical College, No. 22 Xinling Road, Shantou 515041, Guangdong, China
| | - Ling Chen
- Department of Pharmacology, Shantou University Medical College, No. 22 Xinling Road, Shantou 515041, Guangdong, China
| | - Yongdong Niu
- Department of Pharmacology, Shantou University Medical College, No. 22 Xinling Road, Shantou 515041, Guangdong, China.
| | - Ganggang Shi
- Department of Pharmacology, Shantou University Medical College, No. 22 Xinling Road, Shantou 515041, Guangdong, China.
| |
Collapse
|
8
|
Gamaev L, Mizrahi L, Friehmann T, Rosenberg N, Pappo O, Olam D, Zeira E, Bahar Halpern K, Caruso S, Zucman-Rossi J, Axelrod JH, Galun E, Goldenberg DS. The pro-oncogenic effect of the lncRNA H19 in the development of chronic inflammation-mediated hepatocellular carcinoma. Oncogene 2021; 40:127-139. [PMID: 33093654 DOI: 10.1038/s41388-020-01513-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 10/01/2020] [Accepted: 10/05/2020] [Indexed: 01/09/2023]
Abstract
The oncofetal long noncoding RNA (lncRNA) H19 is postnatally repressed in most tissues, and re-expressed in many cancers, including hepatocellular carcinoma (HCC). The role of H19 in carcinogenesis is a subject of controversy. We aimed to examine the role of H19 in chronic inflammation-mediated hepatocarcinogenesis using the Mdr2/Abcb4 knockout (Mdr2-KO) mouse, a well-established HCC model. For this goal, we have generated Mdr2-KO/H19-KO double knockout (dKO) mice and followed spontaneous tumor development in the dKO and control Mdr2-KO mice. Cellular localization of H19 and effects of H19 loss in the liver were determined in young and old Mdr2-KO mice. Tumor incidence and tumor load were both significantly decreased in the liver of dKO versus Mdr2-KO females. The expression levels of H19 and Igf2 were variable in nontumor liver tissues of Mdr2-KO females and were significantly downregulated in most matched tumors. In nontumor liver tissue of aged Mdr2-KO females, H19 was expressed mainly in hepatocytes, and hepatocyte proliferation was increased compared to dKO females. At an early age, dKO females displayed lower levels of liver injury and B-cell infiltration, with higher percentage of binuclear hepatocytes. In human samples, H19 expression was higher in females, positively correlated with cirrhosis (in nontumor liver samples) and negatively correlated with CTNNB1 (beta-catenin) mutations and patients' survival (in tumors). Our data demonstrate that the lncRNA H19 is pro-oncogenic during the development of chronic inflammation-mediated HCC in the Mdr2-KO mouse model, mainly by increasing liver injury and decreasing hepatocyte polyploidy in young mice.
Collapse
Affiliation(s)
- Lika Gamaev
- The Goldyne Savad Institute of Gene and Cell Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Lina Mizrahi
- The Goldyne Savad Institute of Gene and Cell Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Tomer Friehmann
- The Goldyne Savad Institute of Gene and Cell Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Nofar Rosenberg
- The Goldyne Savad Institute of Gene and Cell Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Orit Pappo
- Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Devorah Olam
- The Goldyne Savad Institute of Gene and Cell Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Evelyne Zeira
- The Goldyne Savad Institute of Gene and Cell Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Keren Bahar Halpern
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Stefano Caruso
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Functional Genomics of Solid Tumors Laboratory, F-75006, Paris, France
| | - Jessica Zucman-Rossi
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Functional Genomics of Solid Tumors Laboratory, F-75006, Paris, France
- Hôpital Européen Georges Pompidou, AP-HP, F-75015, Paris, France
| | - Jonathan H Axelrod
- The Goldyne Savad Institute of Gene and Cell Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Eithan Galun
- The Goldyne Savad Institute of Gene and Cell Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Daniel S Goldenberg
- The Goldyne Savad Institute of Gene and Cell Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel.
| |
Collapse
|
9
|
Lau-Corona D, Bae WK, Hennighausen L, Waxman DJ. Sex-biased genetic programs in liver metabolism and liver fibrosis are controlled by EZH1 and EZH2. PLoS Genet 2020; 16:e1008796. [PMID: 32428001 PMCID: PMC7263639 DOI: 10.1371/journal.pgen.1008796] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 06/01/2020] [Accepted: 04/24/2020] [Indexed: 12/15/2022] Open
Abstract
Sex differences in the incidence and progression of many liver diseases, including liver fibrosis and hepatocellular carcinoma, are associated with sex-biased hepatic expression of hundreds of genes. This sexual dimorphism is largely determined by the sex-specific pattern of pituitary growth hormone secretion, which controls a transcriptional regulatory network operative in the context of sex-biased and growth hormone-regulated chromatin states. Histone H3K27-trimethylation yields a major sex-biased repressive chromatin mark deposited at many strongly female-biased genes in male mouse liver, but not at male-biased genes in female liver, and is catalyzed by polycomb repressive complex-2 through its homologous catalytic subunits, Ezh1 and Ezh2. Here, we used Ezh1-knockout mice with a hepatocyte-specific knockout of Ezh2 to investigate the sex bias of liver H3K27-trimethylation and its functional role in regulating sex-differences in the liver. Combined hepatic Ezh1/Ezh2 deficiency led to a significant loss of sex-biased gene expression, particularly in male liver, where many female-biased genes were increased in expression while male-biased genes showed decreased expression. The associated loss of H3K27me3 marks, and increases in the active enhancer marks H3K27ac and H3K4me1, were also more pronounced in male liver. Further, Ezh1/Ezh2 deficiency in male liver, and to a lesser extent in female liver, led to up regulation of many genes linked to liver fibrosis and liver cancer, which may contribute to the observed liver pathologies and the increased sensitivity of these mice to hepatotoxin exposure. Thus, Ezh1/Ezh2-catalyzed H3K27-trimethyation regulates sex-dependent genetic programs in liver metabolism and liver fibrosis through its sex-dependent effects on the epigenome, and may thereby determine the sex-bias in liver disease susceptibility. Sex-differences in the expression of genes in liver have a direct impact on liver diseases whose incidence and severity is sex-biased, and is controlled by hormones that regulate chemical alterations to histone proteins used to package chromosomal DNA. However, a direct demonstration of the functional importance of such sex differences in histone protein modifications has been elusive. Here, we address this question using a mouse model deficient in two enzymes, Ezh1/Ezh2, which generate the histone repressive mark H3K27me3. Remarkably, although H3K27me3 marks are formed by Ezh1/Ezh2 throughout the genome, loss of liver Ezh1/Ezh2 preferentially disrupts the control of sex-biased genes, with expression increasing in male mouse liver for many female-biased genes and decreasing for many male-biased genes. Sex-biased H3K27me3 repressive marks were abolished, and there was a gain of active histone marks at gene enhancers. We also found increased expression of many genes associated with liver fibrosis and hepatocellular carcinoma, which may help explain the increased sensitivity of Ezh1/Ezh2-deficient livers to hepatotoxic chemicals whose exposure may lead to sex differences in liver disease incidence and susceptibility. Thus, our findings highlight the potential role of sex differences in histone modifications catalyzed by Ezh1/Ezh2 in widespread sex differences in liver diseases.
Collapse
Affiliation(s)
- Dana Lau-Corona
- Department of Biology and Bioinformatics Program, Boston University, Boston, Massachusetts, United States of America
| | - Woo Kyun Bae
- Laboratory of Genetics and Physiology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Korea
| | - Lothar Hennighausen
- Laboratory of Genetics and Physiology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - David J. Waxman
- Department of Biology and Bioinformatics Program, Boston University, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
10
|
Abstract
Long non-coding RNAs (lncRNAs) represent a major fraction of the transcriptome in multicellular organisms. Although a handful of well-studied lncRNAs are broadly recognized as biologically meaningful, the fraction of such transcripts out of the entire collection of lncRNAs remains a subject of vigorous debate. Here we review the evidence for and against biological functionalities of lncRNAs and attempt to arrive at potential modes of lncRNA functionality that would reconcile the contradictory conclusions. Finally, we discuss different strategies of phenotypic analyses that could be used to investigate such modes of lncRNA functionality.
Collapse
Affiliation(s)
- Fan Gao
- Institute of Genomics, School of Biomedical Sciences, Huaqiao University, 201 Pan-Chinese S & T Building, 668 Jimei Road, Xiamen, 361021, China
| | - Ye Cai
- Institute of Genomics, School of Biomedical Sciences, Huaqiao University, 201 Pan-Chinese S & T Building, 668 Jimei Road, Xiamen, 361021, China
| | - Philipp Kapranov
- Institute of Genomics, School of Biomedical Sciences, Huaqiao University, 201 Pan-Chinese S & T Building, 668 Jimei Road, Xiamen, 361021, China.
| | - Dongyang Xu
- Institute of Genomics, School of Biomedical Sciences, Huaqiao University, 201 Pan-Chinese S & T Building, 668 Jimei Road, Xiamen, 361021, China.
| |
Collapse
|
11
|
Wang H, Cao Y, Shu L, Zhu Y, Peng Q, Ran L, Wu J, Luo Y, Zuo G, Luo J, Zhou L, Shi Q, Weng Y, Huang A, He TC, Fan J. Long non-coding RNA (lncRNA) H19 induces hepatic steatosis through activating MLXIPL and mTORC1 networks in hepatocytes. J Cell Mol Med 2020; 24:1399-1412. [PMID: 31809000 PMCID: PMC6991647 DOI: 10.1111/jcmm.14818] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 09/04/2019] [Accepted: 09/16/2019] [Indexed: 12/11/2022] Open
Abstract
Liver plays an essential role in regulating lipid metabolism, and chronically disturbed hepatic metabolism may cause obesity and metabolic syndrome, which may lead to non-alcoholic fatty liver disease (NAFLD). Increasing evidence indicates long non-coding RNAs (lncRNAs) play an important role in energy metabolism. Here, we investigated the role of lncRNA H19 in hepatic lipid metabolism and its potential association with NAFLD. We found that H19 was up-regulated in oleic acid-induced steatosis and during the development of high-fat diet (HFD)-induced NAFLD. Exogenous overexpression of H19 in hepatocytes induced lipid accumulation and up-regulated the expression of numerous genes involved in lipid synthesis, storage and breakdown, while silencing endogenous H19 led to a decreased lipid accumulation in hepatocytes. Mechanistically, H19 was shown to promote hepatic steatosis by up-regulating lipogenic transcription factor MLXIPL. Silencing Mlxipl diminished H19-induced lipid accumulation in hepatocytes. Furthermore, H19-induced lipid accumulation was effectively inhibited by PI3K/mTOR inhibitor PF-04691502. Accordingly, H19 overexpression in hepatocytes up-regulated most components of the mTORC1 signalling axis, which were inhibited by silencing endogenous H19. In vivo hepatocyte implantation studies further confirm that H19 promoted hepatic steatosis by up-regulating both mTORC1 signalling axis and MLXIPL transcriptional network. Collectively, these findings strongly suggest that H19 may play an important role in regulating hepatic lipid metabolism and may serve as a potential therapeutic target for NAFLD.
Collapse
Affiliation(s)
- Hao Wang
- Ministry of Education Key Laboratory of Diagnostic Medicine, and School of Laboratory Medicine, Chongqing Medical University, Chongqing, China.,Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
| | - Youde Cao
- Department of Pathology, Chongqing Medical University, Chongqing, China
| | - Liqing Shu
- Department of Pathology, Chongqing Medical University, Chongqing, China
| | - Ying Zhu
- Ministry of Education Key Laboratory of Diagnostic Medicine, and School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Qi Peng
- Ministry of Education Key Laboratory of Diagnostic Medicine, and School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Longke Ran
- Department of Bioinformatics, Chongqing Medical University, Chongqing, China
| | - Jinghong Wu
- Ministry of Education Key Laboratory of Diagnostic Medicine, and School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Yetao Luo
- Department of Biostatistics, School of Public Health and Management, Chongqing Medical University, Chongqing, China
| | - Guowei Zuo
- Ministry of Education Key Laboratory of Diagnostic Medicine, and School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Jinyong Luo
- Ministry of Education Key Laboratory of Diagnostic Medicine, and School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Lan Zhou
- Ministry of Education Key Laboratory of Diagnostic Medicine, and School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Qiong Shi
- Ministry of Education Key Laboratory of Diagnostic Medicine, and School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Yaguang Weng
- Ministry of Education Key Laboratory of Diagnostic Medicine, and School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Ailong Huang
- Key Laboratory of Molecular Biology for Infectious Diseases of The Ministry of Education of China, Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
| | - Jiaming Fan
- Ministry of Education Key Laboratory of Diagnostic Medicine, and School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| |
Collapse
|
12
|
Wendt A, Esguerra JL, Eliasson L. Islet microRNAs in health and type-2 diabetes. Curr Opin Pharmacol 2018; 43:46-52. [PMID: 30144686 DOI: 10.1016/j.coph.2018.08.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 08/02/2018] [Indexed: 12/24/2022]
Abstract
Failure of the β-cell to secrete enough insulin is a major contributing factor in the pathogenesis of type-2 diabetes (T2D). MicroRNAs provide an extra layer in the regulation of protein expression, and are thus involved in β-cell compensation during development of the disease. In this review, we discuss how microRNAs can regulate their target protein expression and phenotypic output, present the status of nutritional regulation of microRNA expression, and summarize work on microRNA expression in human islets. In conclusion, current data lend support to microRNAs being essential regulators of insulin secretion. Future work will describe microRNAs in α-cell function, details of the microRNA-mRNA network, and possibilities to use microRNAs as biomarkers and in therapeutic treatment of T2D and complications.
Collapse
Affiliation(s)
- Anna Wendt
- Islet Cell Exocytosis, Lund University Diabetes Centre, Department of Clinical Sciences Malmö, Lund University and Clinical Research Centre, SUS, Malmö, Sweden
| | - Jonathan Ls Esguerra
- Islet Cell Exocytosis, Lund University Diabetes Centre, Department of Clinical Sciences Malmö, Lund University and Clinical Research Centre, SUS, Malmö, Sweden
| | - Lena Eliasson
- Islet Cell Exocytosis, Lund University Diabetes Centre, Department of Clinical Sciences Malmö, Lund University and Clinical Research Centre, SUS, Malmö, Sweden.
| |
Collapse
|