1
|
You B, Zhang J, Yang C, Dun Y, Qi D, Long Y, Cheng J, Lin Y, Zhou N, Zeng T, Dong J, Liu S. Salidroside protects against myocardial infarction via activating MIF-mediated mitochondrial quality control. Chin Med 2025; 20:27. [PMID: 40016840 PMCID: PMC11869418 DOI: 10.1186/s13020-025-01076-3] [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: 09/14/2024] [Accepted: 02/11/2025] [Indexed: 03/01/2025] Open
Abstract
BACKGROUND Salidroside is a potential therapeutic agent for myocardial infarction (MI), exerting therapeutic effects on macrophage migration inhibitory factor (MIF)-regulated mitochondrial quality control. Our aim was to explore the mechanism through which the MIF pathway regulates salidroside-mediated resistance to hypoxia-induced cardiomyocyte apoptosis. METHODS Ligation surgery of the left anterior descending branch of the coronary artery was employed to establish a myocardial infarction mouse model. Salidroside at low and high doses was administered to the mice for 4 weeks after the surgery. Cardiac function was evaluated via echocardiography. Morphological changes, apoptosis, and mitochondrial damage in the myocardium were examined. For the cell experiments, cardiomyocytes were treated with salidroside under oxygen‒glucose deprivation (OGD) conditions and were either treated with recombinant MIF (rMIF) or transfected with Mif-siRNA. Subsequently, mitochondrial quality control and apoptosis were assessed. RESULTS Salidroside enhanced mitochondrial quality control in MI model mice, mitigated apoptosis and improved cardiac dysfunction. Transmission electron microscopy indicated that there were fewer damaged mitochondria in the salidroside-treated mice compared with the control mice. MIF and downstream mitochondrial quality control pathways were activated in the mice treated with salidroside. Consistently, the cell experiments demonstrated that salidroside and rMIF alleviated apoptosis, improved impaired mitochondrial quality control in OGD-induced cells and activated MIF signaling in OGD-induced cells. However, these effects of salidroside were partially blocked by Mif-siRNA transfection. CONCLUSION Salidroside alleviated myocardial apoptosis and ameliorated cardiac dysfunction in MI model mice through the MIF pathway and downstream mitochondrial quality control.
Collapse
Affiliation(s)
- Baiyang You
- Division of Cardiac Rehabilitation, Department of Physical Medicine and Rehabilitation, Xiangya Hospital Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, Hunan, China
- Department of Physical Medicine and Rehabilitation, Xiangya Hospital Central South University, Jiangxi (National Regional Center for Neurological Diseases), China
| | - Jie Zhang
- Division of Cardiac Rehabilitation, Department of Physical Medicine and Rehabilitation, Xiangya Hospital Central South University, Changsha, Hunan, China
| | - Chuyan Yang
- Department of Physical Medicine and Rehabilitation, Xiangya Hospital Central South University, Jiangxi (National Regional Center for Neurological Diseases), China
- Department of Rehabilitation Medicine, Jiangxi Provincial People's Hospital, Clinical College of Nanchang Medical College, First Affiliated Hospital of Nanchang Medical College, Jiangxi, China
| | - Yaoshan Dun
- Division of Cardiac Rehabilitation, Department of Physical Medicine and Rehabilitation, Xiangya Hospital Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, Hunan, China
- Department of Physical Medicine and Rehabilitation, Xiangya Hospital Central South University, Jiangxi (National Regional Center for Neurological Diseases), China
| | - Dake Qi
- College of Pharmacy, University of Manitoba, Winnipeg, MB, Canada
| | - Yuqiong Long
- Division of Cardiac Rehabilitation, Department of Physical Medicine and Rehabilitation, Xiangya Hospital Central South University, Changsha, Hunan, China
| | - Jing Cheng
- Department of Cardiovascular, Shenzhen District Yantian People's Hospital, Shenzhen, Guangdong, China
| | - Yuan Lin
- Division of Cardiac Rehabilitation, Department of Physical Medicine and Rehabilitation, Xiangya Hospital Central South University, Changsha, Hunan, China
- Department of Physical Medicine and Rehabilitation, Xiangya Hospital Central South University, Jiangxi (National Regional Center for Neurological Diseases), China
| | - Nanjiang Zhou
- Division of Cardiac Rehabilitation, Department of Physical Medicine and Rehabilitation, Xiangya Hospital Central South University, Changsha, Hunan, China
| | - Tanghao Zeng
- Division of Cardiac Rehabilitation, Department of Physical Medicine and Rehabilitation, Xiangya Hospital Central South University, Changsha, Hunan, China
| | - Jie Dong
- Division of Cardiac Rehabilitation, Department of Physical Medicine and Rehabilitation, Xiangya Hospital Central South University, Changsha, Hunan, China.
- Department of Physical Medicine and Rehabilitation, Xiangya Hospital Central South University, Jiangxi (National Regional Center for Neurological Diseases), China.
| | - Suixin Liu
- Division of Cardiac Rehabilitation, Department of Physical Medicine and Rehabilitation, Xiangya Hospital Central South University, Changsha, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, Hunan, China.
- Department of Physical Medicine and Rehabilitation, Xiangya Hospital Central South University, Jiangxi (National Regional Center for Neurological Diseases), China.
| |
Collapse
|
2
|
Caño-Carrillo S, Garcia-Padilla C, Aranega AE, Lozano-Velasco E, Franco D. Mef2c- and Nkx2-5-Divergent Transcriptional Regulation of Chick WT1_76127 and Mouse Gm14014 lncRNAs and Their Implication in Epicardial Cell Migration. Int J Mol Sci 2024; 25:12904. [PMID: 39684625 DOI: 10.3390/ijms252312904] [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: 10/17/2024] [Revised: 11/25/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
Cardiac development is a complex developmental process. The early cardiac straight tube is composed of an external myocardial layer and an internal endocardial lining. Soon after rightward looping, the embryonic heart becomes externally covered by a new epithelial lining, the embryonic epicardium. A subset of these embryonic epicardial cells migrate and colonize the embryonic myocardium, contributing to the formation of distinct cell types. In recent years, our understanding of the molecular mechanisms that govern proepicardium and embryonic epicardium formation has greatly increased. We have recently witnessed the discovery of a novel layer of complexity governing gene regulation with the discovery of non-coding RNAs. Our laboratory recently identified three distinct lncRNAs, adjacent to the Wt1, Bmp4 and Fgf8 chicken gene loci, with enhanced expression in the proepicardium that are distinctly regulated by Bmp, Fgf and thymosin β4, providing support for their plausible implication in epicardial formation. The expression of lncRNAs was analyzed in different chicken and mouse tissues as well as their subcellular distribution in chicken proepicardial, epicardial, ventricle explants and in different murine cardiac cell types. lncRNA transcriptional regulation was analyzed by using siRNAs and expression vectors of different transcription factors in chicken and mouse models, whereas antisense oligonucleotides were used to inhibit Gm14014 expression. Furthermore, RT-qPCR, immunocytochemistry, RNA pulldown, Western blot, viability and cell migration assays were conducted to investigate the biological functions of Wt1_76127 and Gm14014. We demonstrated that Wt1_76127 in chicken and its putative conserved homologue Gm14014 in mice are widely distributed in different embryonic and adult tissues and distinctly regulated by cardiac-enriched transcription factors, particularly Mef2c and Nkx2.5. Furthermore, silencing assays demonstrated that mouse Gm14014, but not chicken Wt1_76127, is essential for epicardial, but not endocardial or myocardial, cell migration. Such processes are governed by partnering with Myl9, promoting cytoskeletal remodeling. Our data show that Gm14014 plays a pivotal role in epicardial cell migration essential for heart regeneration under these experimental conditions.
Collapse
Affiliation(s)
- Sheila Caño-Carrillo
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain
| | - Carlos Garcia-Padilla
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain
| | - Amelia E Aranega
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain
- Fundación Medina, 18016 Granada, Spain
| | - Estefania Lozano-Velasco
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain
- Fundación Medina, 18016 Granada, Spain
| | - Diego Franco
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain
- Fundación Medina, 18016 Granada, Spain
| |
Collapse
|
3
|
Zhao Y, Han S, Zeng Z, Zheng H, Li Y, Wang F, Huang Y, Zhao Y, Zhuo W, Lv G, Wang H, Zhao G, Zhao E, Hu Y, Hu P, Zhao G. Decreased lncRNA HNF4A-AS1 facilitates resistance to sorafenib-induced ferroptosis of hepatocellular carcinoma by reprogramming lipid metabolism. Theranostics 2024; 14:7088-7110. [PMID: 39629121 PMCID: PMC11610135 DOI: 10.7150/thno.99197] [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: 06/01/2024] [Accepted: 10/12/2024] [Indexed: 12/06/2024] Open
Abstract
Background: Resistance to sorafenib remains a major challenge in the systemic therapy of liver cancer. However, the involvement of lipid metabolism-related lncRNAs in this process remains unclear. Methods: Different expression levels of lipid metabolism-related lncRNAs in HCC were compared by analysis of Gene Expression Omnibus and The Cancer Genome Atlas databases. The influence of HNF4A-AS1 on sorafenib response was evaluated through analysis of public biobanks, cell cytotoxicity and colony formation assays. The effect of HNF4A-AS1 on sorafenib-induced ferroptosis was measured using lipid peroxidation, glutathione, malondialdehyde, and ROS levels. Furthermore, bioinformatic analyses and lipidomic profiling were conducted to study HNF4A-AS1 involvement in lipid metabolic reprogramming. Mechanistic experiments, including the luciferase reporter assay, RNA pulldown, RNA immunoprecipitation (RIP), methylated RNA immunoprecipitation (MeRIP), and RNA remaining assays, were employed to uncover the downstream targets and regulatory mechanisms of HNF4A-AS1 in sorafenib resistance in HCC. Xenograft and organoid experiments were carried out to assess the impact of HNF4A-AS1 on sorafenib response. Results: Bioinformatics analysis revealed that HNF4A-AS1, a lipid metabolism-related lncRNA, is specifically high-expressed in the normal liver and associated with sorafenib resistance in HCC. We further confirmed that HNF4A-AS1 was downregulated in HCC cells and organoids that resistant to sorafenib. Moreover, both in vitro and in vivo studies demonstrated that HNF4A-AS1 overexpression reversed sorafenib resistance in HCC cells, which was further enhanced by polyunsaturated fatty acids (PUFA) supplementation. Mechanistically, HNF4A-AS1 interacted with METTL3, leading to m6A modification of DECR1 mRNA, which subsequently decreased DECR1 expression via YTHDF3-dependent mRNA degradation. Consequently, decreased HNF4A-AS1 levels caused DECR1 overexpression, leading to decreased intracellular PUFA content and promoting resistance to sorafenib-induced ferroptosis in HCC. Conclusions: Our results indicated the pivotal role of lipid metabolism-related and liver-specific HNF4A-AS1 in inhibiting sorafenib resistance by promoting ferroptosis and suggesting that HNF4A-AS1 might be a potential target for HCC.
Collapse
MESH Headings
- Sorafenib/pharmacology
- Sorafenib/therapeutic use
- Humans
- Ferroptosis/drug effects
- Ferroptosis/genetics
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Liver Neoplasms/drug therapy
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Drug Resistance, Neoplasm/genetics
- Lipid Metabolism/drug effects
- Lipid Metabolism/genetics
- Animals
- Hepatocyte Nuclear Factor 4/metabolism
- Hepatocyte Nuclear Factor 4/genetics
- Mice
- Cell Line, Tumor
- Gene Expression Regulation, Neoplastic/drug effects
- Mice, Nude
- Antineoplastic Agents/pharmacology
- Xenograft Model Antitumor Assays
- Male
- Mice, Inbred BALB C
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Ping Hu
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gang Zhao
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
4
|
Qin C, Wang D, Han H, Cao Y, Wang X, Xuan Z, Wei M, Li Z, Liu Q. Expression patterns of housekeeping genes and tissue-specific genes in black goats across multiple tissues. Sci Rep 2024; 14:21896. [PMID: 39300207 DOI: 10.1038/s41598-024-72844-8] [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: 06/25/2024] [Accepted: 09/11/2024] [Indexed: 09/22/2024] Open
Abstract
Black goats are a significant meat breed in southern China. To investigate the expression patterns and biological functions of genes in various tissues of black goats, we analyzed housekeeping genes (HKGs), tissue-specific genes (TSGs), and hub genes (HUBGs) across 23 tissues. Additionally, we analyzed HKGs in 13 tissues under different feeding conditions. We identified 2968 HKGs, including six important ones. Interestingly, HKGs in grazing black goats demonstrated higher and more stable expression levels. We discovered a total of 9912 TSGs, including 134 newly identified ones. The number of TSGs for mRNA and lncRNA were nearly equal, with 127 mRNA TSGs expressed solely in one tissue. Additionally, the predicted functions of tissue-specific long non-coding RNAs (lncRNAs) targeting mRNAs corresponded with the physiological functions of the tissues.Weighted gene co-expression network analysis (WGCNA) constructed 30 modules, where the dark grey module consists almost entirely of HKGs, and TSGs are located in modules most correlated with their respective tissues. Additionally, we identified 289 HUBGs, which are involved in regulating the physiological functions of their respective tissues. Overall, these identified HKGs, TSGs, and HUBGs provide a foundation for studying the molecular mechanisms affecting the genetic and biological processes of complex traits in black goats.
Collapse
Affiliation(s)
- Chaobin Qin
- School of Animal Science and Technology, Guangxi University, Nanning, 530004, China
| | - Dong Wang
- School of Animal Science and Technology, Guangxi University, Nanning, 530004, China
| | - Hongbing Han
- Beijing Key Laboratory of Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Yanhong Cao
- Guangxi Vocational University of Agriculture, Nanning, 530007, Guangxi, China
| | - Xiaobo Wang
- Henan Academy of Crops Molecular Breeding/The Shennong Laboratory, Zhengzhou, 450099, Henan, China
| | - Zeyi Xuan
- Guangxi Vocational University of Agriculture, Nanning, 530007, Guangxi, China
| | - Mingsong Wei
- Guangxi Vocational University of Agriculture, Nanning, 530007, Guangxi, China.
| | - Zhipeng Li
- School of Animal Science and Technology, Guangxi University, Nanning, 530004, China.
| | - Qingyou Liu
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan, 528225, China.
| |
Collapse
|
5
|
Shiu PKT, DiStefano JK, Alahari SK, Enguita FJ, Feinberg MW, Sideris N, Bayraktar S, Castellano L, Buitrago DL, Caporali A, Mannucci A, Goel A. The Non-Coding RNA Journal Club: Highlights on Recent Papers-13. Noncoding RNA 2023; 9:76. [PMID: 38133210 PMCID: PMC10745700 DOI: 10.3390/ncrna9060076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 12/06/2023] [Indexed: 12/23/2023] Open
Abstract
We are delighted to share with you our thirteenth Journal Club and highlight some of the most interesting papers published recently [...].
Collapse
Affiliation(s)
- Patrick K. T. Shiu
- Division of Biological Sciences, University of Missouri, Columbia, MO 65211, USA
| | - Johanna K. DiStefano
- Metabolic Disease Research Unit, Translational Genomics Research Institute, Phoenix, AZ 85004, USA
| | - Suresh K. Alahari
- Department of Biochemistry and Molecular Biology, LSUHSC School of Medicine, New Orleans, LA 70112, USA
| | - Francisco J. Enguita
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Mark W. Feinberg
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Nikolaos Sideris
- School of Life Sciences, University of Sussex, Falmer, Brighton BN1 9QG, UK (S.B.)
| | - Salih Bayraktar
- School of Life Sciences, University of Sussex, Falmer, Brighton BN1 9QG, UK (S.B.)
| | - Leandro Castellano
- School of Life Sciences, University of Sussex, Falmer, Brighton BN1 9QG, UK (S.B.)
- Department of Surgery and Cancer, Imperial Centre for Translational and Experimental Medicine (ICTEM), Imperial College London, London W12 0NN, UK
| | - Diana Luna Buitrago
- BHF Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK;
| | - Andrea Caporali
- BHF Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK;
| | - Alessandro Mannucci
- Gastroenterology and Gastrointestinal Endoscopy Unit, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, 20132 Milan, Italy;
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Biomedical Research Center, Monrovia, CA 91016, USA
| | - Ajay Goel
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Biomedical Research Center, Monrovia, CA 91016, USA
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| |
Collapse
|