1
|
Wang W, Asiru, Luo G, Chen Y, Cui Y, Ping S, Chen Y. A Novel Effect of Id2 in Microglia TNFα Regulation. Mol Neurobiol 2025; 62:304-321. [PMID: 38850351 DOI: 10.1007/s12035-024-04278-2] [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: 03/07/2024] [Accepted: 06/01/2024] [Indexed: 06/10/2024]
Abstract
Microglia are the most important immune cells in the central nervous system (CNS), which can defend against external pathogens and stimuli. Dysregulation of microglia releases excessive proinflammatory cytokines and leads to neuroinflammation, which is fundamental to the pathophysiology of multiple neurological diseases. However, the molecular mechanisms underlying the regulation of proinflammatory cytokines in microglia are still not well-understood. Here, we identified that inhibitor of DNA binding protein 2 (Id2) was a negative regulator of tumor necrosis factor-α (TNFα) in cultured microglia. Knockdown of Id2 significantly increased the expression of TNFα in microglia, while overexpression of Id2 inhibited TNFα expression. Furthermore, by interacting with the p65 subunit of nuclear factor kappa-B (NF-κB), Id2 suppressed the transcription activation of NF-κB and inhibited TNFα expression. Interestingly, in lipopolysaccharides (LPS)-treated microglia, Id2 increased and underwent a cytoplasmic relocation. Immunoprecipitation and immunostaining results showed that by binding to the LIM domain of Id2, a scaffold protein PDZ and LIM 5 (PDLIM5) involved in the Id2 cytoplasmic relocation, which inactivated Id2 and resulted in higher TNFα expression in LPS-treated microglia. Collectively, our data delineate a novel effect of Id2 on TNFα regulation in microglia, which may shed a light on the proinflammatory cytokines regulating in microglia associated neuroimmune disorders.
Collapse
Affiliation(s)
- Wenhui Wang
- Neurobiology Research Center, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, People's Republic of China
| | - Asiru
- Neurobiology Research Center, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, People's Republic of China
| | - Guoya Luo
- Neurobiology Research Center, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, People's Republic of China
| | - Yanmei Chen
- Neurobiology Research Center, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, People's Republic of China
| | - Yu Cui
- Neurobiology Research Center, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, People's Republic of China
| | - Suning Ping
- Neurobiology Research Center, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, People's Republic of China.
- Department of Histology and Embryology, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, People's Republic of China.
| | - Yuan Chen
- Neurobiology Research Center, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, People's Republic of China.
| |
Collapse
|
2
|
Fu R, Dou Z, Li N, Fan X, Amin S, Zhang J, Wang Y, Li Z, Li Z, Yang P. Avenanthramide A potentiates Bim-mediated antineoplastic properties of 5-fluorouracil via targeting KDM4C/ MIR17HG/GSK-3 β negative feedback loop in colorectal cancer. Acta Pharm Sin B 2024; 14:5321-5340. [PMID: 39807336 PMCID: PMC11725033 DOI: 10.1016/j.apsb.2024.07.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 05/24/2024] [Accepted: 07/10/2024] [Indexed: 01/16/2025] Open
Abstract
Chemoresistance to 5-fluorouracil (5-FU) is a significant challenge in treating colorectal cancer (CRC). Novel combined regimens to thwart chemoresistance are therefore urgently needed. Herein, we demonstrated that the combination of Avenanthramide A (AVN A) and 5-FU has significant therapeutic advantages against CRC. Mechanistically, AVN A directly binds to the S198 site of the histone lysine demethylase KDM4C to promote its degradation, which subsequently fosters H3K9me3 occupancy on the MIR17HG promoter to block its transcription and derepress Bim expression. AVN A enhanced the therapeutic efficacy of 5-FU via impairing the KDM4C/MIR17HG/GSK-3β negative feedback loop. Importantly, the clinical correlation of the KDM4C/MIR17HG/Bim signaling axis with 5-FU response was validated in the refractory CRC patients. We provide evidence for the enhanced effectiveness of 5-FU when combined with AVN A in chemoresistant xenografts, CRC organoids, and Apc Min/+ mouse model. Additionally, AVN A mitigated the systemic adverse effects of 5-FU. Overall, our findings demonstrate that combinatorial therapy with AVN A and 5-FU represents an appealing opportunity and highlights KDM4C/MIR17HG/GSK-3β negative feedback loop which confers therapeutically exploitable vulnerability to chemo-refractory CRC patients.
Collapse
Affiliation(s)
- Rong Fu
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, China
- Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Shanxi University, Taiyuan 030006, China
| | - Zhangfeng Dou
- Department of Gastroenterology, First Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Ning Li
- Department of Gastroenterology, First Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Xueyuan Fan
- School of Life Science, Shanxi University, Taiyuan 030006, China
| | - Sajid Amin
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples 80138, Italy
| | - Jinqi Zhang
- Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Shanxi University, Taiyuan 030006, China
| | - Yuqing Wang
- Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Shanxi University, Taiyuan 030006, China
| | - Zongwei Li
- School of Life Science, Anhui Medical University, Hefei 230032, China
| | - Zhuoyu Li
- Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Shanxi University, Taiyuan 030006, China
| | - Peng Yang
- Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Shanxi University, Taiyuan 030006, China
| |
Collapse
|
3
|
Fu Y, Li S, Nie J, Yan D, Zhang B, Hao X, Zhang H. Expression of PDLIM5 Spliceosomes and Regulatory Functions on Myogenesis in Pigs. Cells 2024; 13:720. [PMID: 38667334 PMCID: PMC11049100 DOI: 10.3390/cells13080720] [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: 03/14/2024] [Revised: 04/18/2024] [Accepted: 04/19/2024] [Indexed: 04/28/2024] Open
Abstract
Meat yield, determined by muscle growth and development, is an important economic trait for the swine industry and a focus of research in animal genetics and breeding. PDZ and LIM domain 5 (PDLIM5) are cytoskeleton-related proteins that play key roles in various tissues and cells. These proteins have multiple isoforms, primarily categorized as short (PDLIM5-short) and long (PDLIM5-long) types, distinguished by the absence and presence of an LIM domain, respectively. However, the expression patterns of swine PDLIM5 isoforms and their regulation during porcine skeletal muscle development remain largely unexplored. We observed that PDLIM5-long was expressed at very low levels in pig muscles and that PDLIM5-short and total PDLIM5 were highly expressed in the muscles of slow-growing pigs, suggesting that PDLIM5-short, the dominant transcript in pigs, is associated with a slow rate of muscle growth. PDLIM5-short suppressed myoblast proliferation and myogenic differentiation in vitro. We also identified two single nucleotide polymorphisms (-258 A > T and -191 T > G) in the 5' flanking region of PDLIM5, which influenced the activity of the promoter and were associated with muscle growth rate in pigs. In summary, we demonstrated that PDLIM5-short negatively regulates myoblast proliferation and differentiation, providing a theoretical basis for improving pig breeding programs.
Collapse
Affiliation(s)
- Yu Fu
- National Engineering Laboratory for Livestock and Poultry Breeding, Beijing Key Laboratory of Animal Genetic Engineering, China Agricultural University, Beijing 100193, China; (Y.F.); (S.L.); (J.N.); (B.Z.)
| | - Shixin Li
- National Engineering Laboratory for Livestock and Poultry Breeding, Beijing Key Laboratory of Animal Genetic Engineering, China Agricultural University, Beijing 100193, China; (Y.F.); (S.L.); (J.N.); (B.Z.)
| | - Jingru Nie
- National Engineering Laboratory for Livestock and Poultry Breeding, Beijing Key Laboratory of Animal Genetic Engineering, China Agricultural University, Beijing 100193, China; (Y.F.); (S.L.); (J.N.); (B.Z.)
| | - Dawei Yan
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China;
| | - Bo Zhang
- National Engineering Laboratory for Livestock and Poultry Breeding, Beijing Key Laboratory of Animal Genetic Engineering, China Agricultural University, Beijing 100193, China; (Y.F.); (S.L.); (J.N.); (B.Z.)
| | - Xin Hao
- National Engineering Laboratory for Livestock and Poultry Breeding, Beijing Key Laboratory of Animal Genetic Engineering, China Agricultural University, Beijing 100193, China; (Y.F.); (S.L.); (J.N.); (B.Z.)
| | - Hao Zhang
- National Engineering Laboratory for Livestock and Poultry Breeding, Beijing Key Laboratory of Animal Genetic Engineering, China Agricultural University, Beijing 100193, China; (Y.F.); (S.L.); (J.N.); (B.Z.)
| |
Collapse
|
4
|
Chen SL, Wu CC, Li N, Weng TH. Post-transcriptional regulation of myogenic transcription factors during muscle development and pathogenesis. J Muscle Res Cell Motil 2024; 45:21-39. [PMID: 38206489 DOI: 10.1007/s10974-023-09663-3] [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: 10/16/2023] [Accepted: 11/29/2023] [Indexed: 01/12/2024]
Abstract
The transcriptional regulation of skeletal muscle (SKM) development (myogenesis) has been documented for over 3 decades and served as a paradigm for tissue-specific cell type determination and differentiation. Myogenic stem cells (MuSC) in embryos and adult SKM are regulated by the transcription factors Pax3 and Pax7 for their stem cell characteristics, while their lineage determination and terminal differentiation are both dictated by the myogenic regulatory factors (MRF) that comprise Mrf4, Myf5, Myogenin, and MyoD. The myocyte enhancer factor Mef2c is activated by MRF during terminal differentiation and collaborates with them to promote myoblast fusion and differentiation. Recent studies have found critical regulation of these myogenic transcription factors at mRNA level, including subcellular localization, stability, and translational regulation. Therefore, the regulation of Pax3/7, MRFs and Mef2c mRNAs by RNA-binding factors and non-coding RNAs (ncRNA), including microRNAs and long non-coding RNAs (lncRNA), will be the focus of this review and the impact of this regulation on myogenesis will be further addressed. Interestingly, the stem cell characteristics of MuSC has been found to be critically regulated by ncRNAs, implying the involvement of ncRNAs in SKM homeostasis and regeneration. Current studies have further identified that some ncRNAs are implicated in the etiology of some SKM diseases and can serve as valuable tools/indicators for prediction of prognosis. The roles of ncRNAs in the MuSC biology and SKM disease etiology will also be discussed in this review.
Collapse
Affiliation(s)
- Shen-Liang Chen
- Department of Life Sciences, National Central University, 300 Jhongda Rd, Jhongli, 32001, Taiwan.
| | - Chuan-Che Wu
- Department of Life Sciences, National Central University, 300 Jhongda Rd, Jhongli, 32001, Taiwan
| | - Ning Li
- Department of Life Sciences, National Central University, 300 Jhongda Rd, Jhongli, 32001, Taiwan
| | - Tzu-Han Weng
- Department of Life Sciences, National Central University, 300 Jhongda Rd, Jhongli, 32001, Taiwan
| |
Collapse
|
5
|
Zhao L, Tang P, Lin Y, Du M, Li H, Jiang L, Xu H, Sun H, Han J, Sun Z, Xu R, Lou H, Chen Z, Kopylov P, Liu X, Zhang Y. MiR-203 improves cardiac dysfunction by targeting PARP1-NAD + axis in aging murine. Aging Cell 2024; 23:e14063. [PMID: 38098220 PMCID: PMC10928583 DOI: 10.1111/acel.14063] [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: 08/17/2023] [Revised: 11/08/2023] [Accepted: 11/26/2023] [Indexed: 03/13/2024] Open
Abstract
Heart aging is a prevalent cause of cardiovascular diseases among the elderly. NAD+ depletion is a hallmark feature of aging heart, however, the molecular mechanisms that affect NAD+ depletion remain unclear. In this study, we identified microRNA-203 (miR-203) as a senescence-associated microRNA that regulates NAD+ homeostasis. We found that the blood miR-203 level negatively correlated with human age and its expression significantly decreased in the hearts of aged mice and senescent cardiomyocytes. Transgenic mice with overexpressed miR-203 (TgN (miR-203)) showed resistance to aging-induced cardiac diastolic dysfunction, cardiac remodeling, and myocardial senescence. At the cellular level, overexpression of miR-203 significantly prevented D-gal-induced cardiomyocyte senescence and mitochondrial damage, while miR-203 knockdown aggravated these effects. Mechanistically, miR-203 inhibited PARP1 expression by targeting its 3'UTR, which helped to reduce NAD+ depletion and improve mitochondrial function and cell senescence. Overall, our study first identified miR-203 as a genetic tool for anti-heart aging by restoring NAD+ function in cardiomyocytes.
Collapse
Affiliation(s)
- Limin Zhao
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine- Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Pingping Tang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine- Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Yuan Lin
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine- Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Menghan Du
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine- Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Huimin Li
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine- Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Lintong Jiang
- Department of Pharmacy, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Henghui Xu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine- Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Heyang Sun
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine- Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Jingjing Han
- Department of Pharmacy, Caoxian People's Hospital, Heze, China
| | - Zeqi Sun
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine- Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Run Xu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine- Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Han Lou
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine- Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Zhouxiu Chen
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine- Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Philipp Kopylov
- Department of Preventive and Emergency Cardiology, Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Xin Liu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine- Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone, Chinese Academy of Medical Sciences, Harbin, China
| | - Yong Zhang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine- Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone, Chinese Academy of Medical Sciences, Harbin, China
- Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Harbin, China
| |
Collapse
|
6
|
Song YL, Yang MH, Zhang S, Wang H, Kai KL, Yao CX, Dai FF, Zhou MJ, Li JB, Wei ZR, Yin Z, Zhu WG, Xue L, Zang MX. A GRIP-1-EZH2 switch binding to GATA-4 is linked to the genesis of rhabdomyosarcoma through miR-29a. Oncogene 2022; 41:5223-5237. [PMID: 36309571 DOI: 10.1038/s41388-022-02521-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 10/14/2022] [Accepted: 10/18/2022] [Indexed: 12/14/2022]
Abstract
Terminal differentiation failure is an important cause of rhabdomyosarcoma genesis, however, little is known about the epigenetic regulation of aberrant myogenic differentiation. Here, we show that GATA-4 recruits polycomb group proteins such as EZH2 to negatively regulate miR-29a in undifferentiated C2C12 myoblast cells, whereas recruitment of GRIP-1 to GATA-4 proteins displaces EZH2, resulting in the activation of miR-29a during myogenic differentiation of C2C12 cells. Moreover, in poorly differentiated rhabdomyosarcoma cells, EZH2 still binds to the miR-29a promoter with GATA-4 to mediate transcriptional repression of miR-29a. Interestingly, once re-differentiation of rhabdomyosarcoma cells toward skeletal muscle, EZH2 was dispelled from miR-29a promoter which is similar to that in myogenic differentiation of C2C12 cells. Eventually, this expression of miR-29a results in limited rhabdomyosarcoma cell proliferation and promotes myogenic differentiation. We thus establish that GATA-4 can function as a molecular switch in the up- and downregulation of miR-29a expression. We also demonstrate that GATA-4 acts as a tumor suppressor in rhabdomyosarcoma partly via miR-29a, which thus provides a potential therapeutic target for rhabdomyosarcoma.
Collapse
Affiliation(s)
- Yang-Liu Song
- Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Ming-Hui Yang
- Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Si Zhang
- Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Hao Wang
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing, 100191, China
| | - Kun-Lun Kai
- Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Chun-Xia Yao
- Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Fei-Fei Dai
- Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Meng-Jiao Zhou
- Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jin-Biao Li
- Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Zhi-Ru Wei
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhongnan Yin
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing, 100191, China
| | - Wei-Guo Zhu
- Department of Biochemistry and Molecular Biology, Shenzhen University School of Medicine, Shenzhen, 518055, China
| | - Lixiang Xue
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing, 100191, China.
- Cancer Center of Peking University Third Hospital, Peking University Third Hospital, Beijing, 100191, China.
| | - Ming-Xi Zang
- Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
7
|
Dang Y, He Q, Yang S, Sun H, Liu Y, Li W, Tang Y, Zheng Y, Wu T. FTH1- and SAT1-Induced Astrocytic Ferroptosis Is Involved in Alzheimer’s Disease: Evidence from Single-Cell Transcriptomic Analysis. Pharmaceuticals (Basel) 2022; 15:ph15101177. [PMID: 36297287 PMCID: PMC9610574 DOI: 10.3390/ph15101177] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 09/16/2022] [Accepted: 09/19/2022] [Indexed: 12/01/2022] Open
Abstract
Objectives: Despite significant advances in neuroscience, the mechanisms of AD are not fully understood. Single-cell RNA sequencing (scRNA-seq) techniques provide potential solutions to analyze cellular composition of complex brain tissue and explore cellular and molecular biological mechanisms of AD. Methods: We investigated cellular heterogeneity in AD via utilization of bioinformatic analysis of scRNA-seq in AD patients and healthy controls from the Gene Expression Omnibus (GEO) database. The “GOplot” package was applied to explore possible biological processes in oligodendrocytes, astrocytes, and oligodendrocyte progenitor cells (OPCs). Expression patterns and biological functions of differentially expressed genes (DEGs) from scRNA-seq data were validated in RNA sequencing data. DEGs in astrocytes interacted with ferroptosis-related genes in FerrDb. CCK-8 and EdU assays were performed to measure cell proliferation ability. ROS, Fe2+ level, mitochondrial membrane potentials, iron concentrations, and total iron binding capacity (TIBC) in serum were evaluated. Y-maze and elevated maze were used to measure anxiety-like behavior. Autonomous and exploration behaviors or learning and memory ability in mice were analyzed using open field test and novel object recognition test. Results: Multiple clusters were identified, including oligodendrocytes, astrocytes, OPCs, neurons, microglia, doublets, and endothelial cells. Astrocytes were significantly decreased in AD, while oligodendrocytes and OPCs increased. Cell-to-cell ligand–receptor interaction analysis revealed that astrocytes, neurons, and OPCs mainly established contacts with other cells via the NRG3–ERBB4 ligand–receptor pair. GO and KEGG analyses found that astrocytes were enriched in the ferroptosis pathway. FTH1 and SAT1 in astrocytes were identified as hub mRNAs associated with ferroptosis. Serum iron concentration of 5xFAD mice was higher than that of WT, and emotional and cognitive function were significantly impaired as compared to WT. Serum iron concentration was negatively correlated with number of astrocytes and percentage of time spent entering the novelty arm in the Y-maze test, while it was positively correlated with percentage of time spent in the central area. Meanwhile, number of astrocytes was negatively correlated with percentage of time spent in the central area, while it was positively correlated with percentage of time spent entering the novelty arm. Conclusions: Through scRNA-seq analysis, we found that ferroptosis was activated in astrocytes and may contribute to the pathophysiological process in the entorhinal cortex. FTH1 and SAT1 were identified to impact astrocyte ferroptosis. Emotional and cognitive impairment in AD was associated with astrocyte ferroptosis. Our findings provide clues to reveal the pathophysiological processes following AD at the cellular level and highlight potential drug targets for the treatment of AD.
Collapse
Affiliation(s)
- Yini Dang
- Department of Gastroenterology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Division of Gastroenterological Rehabilitation, Department of Gastroenterology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Qing He
- Department of Neurology, the Affiliated Hospital of China University of Mining and Technology, Xuzhou 221116, China
| | - Siyu Yang
- Division of Brain Rehabilitation, Department of Neurology, the First Affiliated Hospital of Nanjing Medical University, No.300 Guangzhou Road, Nanjing 210029, China
- Department of Neurology, the First Affiliated Hospital of Nanjing Medical University, No.300 Guangzhou Road, Nanjing 210029, China
| | - Huaiqing Sun
- Division of Brain Rehabilitation, Department of Neurology, the First Affiliated Hospital of Nanjing Medical University, No.300 Guangzhou Road, Nanjing 210029, China
- Department of Neurology, the First Affiliated Hospital of Nanjing Medical University, No.300 Guangzhou Road, Nanjing 210029, China
| | - Yin Liu
- Division of Brain Rehabilitation, Department of Neurology, the First Affiliated Hospital of Nanjing Medical University, No.300 Guangzhou Road, Nanjing 210029, China
- Department of Neurology, the First Affiliated Hospital of Nanjing Medical University, No.300 Guangzhou Road, Nanjing 210029, China
| | - Wanting Li
- Division of Brain Rehabilitation, Department of Neurology, the First Affiliated Hospital of Nanjing Medical University, No.300 Guangzhou Road, Nanjing 210029, China
- Department of Neurology, the First Affiliated Hospital of Nanjing Medical University, No.300 Guangzhou Road, Nanjing 210029, China
| | - Yi Tang
- Division of Brain Rehabilitation, Department of Neurology, the First Affiliated Hospital of Nanjing Medical University, No.300 Guangzhou Road, Nanjing 210029, China
- Department of Neurology, the First Affiliated Hospital of Nanjing Medical University, No.300 Guangzhou Road, Nanjing 210029, China
| | - Yu Zheng
- Department of Rehabilitation Medicine, the First Affiliated Hospital of Nanjing Medical University, No.300 Guangzhou Road, Nanjing 210029, China
- Correspondence: (Y.Z.); (T.W.)
| | - Ting Wu
- Division of Brain Rehabilitation, Department of Neurology, the First Affiliated Hospital of Nanjing Medical University, No.300 Guangzhou Road, Nanjing 210029, China
- Department of Neurology, the First Affiliated Hospital of Nanjing Medical University, No.300 Guangzhou Road, Nanjing 210029, China
- Correspondence: (Y.Z.); (T.W.)
| |
Collapse
|
8
|
Abstract
Recognition of viral RNAs by melanoma differentiation associated gene-5 (MDA5) initiates chicken antiviral response by producing type I interferons. Our previous studies showed that chicken microRNA-155-5p (gga-miR-155-5p) enhanced IFN-β expression and suppressed the replication of infectious burse disease virus (IBDV), a double-stranded RNA (dsRNA) virus causing infectious burse disease in chickens. However, the mechanism underlying IBDV-induced gga-miR-155-5p expression in host cells remains elusive. Here, we show that IBDV infection or poly(I:C) treatment of DF-1 cells markedly increased the expression of GATA-binding protein 3 (GATA3), a master regulator for TH2 cell differentiation, and that GATA3 promoted gga-miR-155-5p expression in IBDV-infected or poly(I:C)-treated cells by directly binding to its promoter. Surprisingly, ectopic expression of GATA3 significantly reduced IBDV replication in DF-1 cells, and this reduction could be completely abolished by treatment with gga-miR-155-5p inhibitors, whereas knockdown of GATA3 by RNA interference enhanced IBDV growth, and this enhancement could be blocked with gga-miR-155-5p mimics, indicating that GATA3 suppressed IBDV replication by gga-miR-155-5p. Furthermore, our data show that MDA5 is required for GATA3 expression in host cells with poly(I:C) treatment, so are the adaptor protein TBK1 and transcription factor IRF7, suggesting that induction of GATA3 expression in IBDV-infected cells relies on MDA5-TBK1-IRF7 signaling pathway. These results uncover a novel role for GATA3 as an antivirus transcription factor in innate immune response by promoting miR-155 expression, further our understandings of host response against pathogenic infection, and provide valuable clues to the development of antiviral reagents for public health. IMPORTANCE Gga-miR-155-5p acts as an important antivirus factor against IBDV infection, which causes a severe immunosuppressive disease in chicken. Elucidation of the mechanism regulating gga-miR-155-5p expression in IBDV-infected cells is essential to our understandings of the host response against pathogenic infection. This study shows that transcription factor GATA3 initiated gga-miR-155-5p expression in IBDV-infected cells by directly binding to its promoter, suppressing viral replication. Furthermore, induction of GATA3 expression was attributable to the recognition of dsRNA by MDA5, which initiates signal transduction via TBK1 and IRF7. Thus, it is clear that IBDV induces GATA3 expression via MDA5-TBK1-IRF7 signaling pathway, thereby suppressing IBDV replication by GATA3-mediated gga-miR-155-5p expression. This information remarkably expands our knowledge of the roles for GATA3 as an antivirus transcription factor in host innate immune response particularly at an RNA level and may prove valuable in the development of antiviral drugs for public health.
Collapse
|
9
|
Ye L, Zuo Y, Chen F, Peng Q, Lu X, Wang G, Shu X. miR-18a-3p and Its Target Protein HuR May Regulate Myogenic Differentiation in Immune-Mediated Necrotizing Myopathy. Front Immunol 2022; 12:780237. [PMID: 35069550 PMCID: PMC8766969 DOI: 10.3389/fimmu.2021.780237] [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: 09/20/2021] [Accepted: 12/13/2021] [Indexed: 11/18/2022] Open
Abstract
Immune-mediated necrotizing myopathy (IMNM) is characterized by manifestation of myonecrosis and regeneration of muscle fibers; however, the underlying pathogenesis remains unclear. This study aimed to investigate the role and mechanism of miR-18a-3p and its target RNA-binding protein HuR in IMNM. HuR and miR-18a-3p levels were detected in the skeletal muscles of 18 patients with IMNM using quantitative reverse-transcription real-time polymerase chain reaction (qRT-PCR) and western blotting analysis. Human myoblasts were transfected with small interfering RNA targeting HuR and miR-18a-3p mimic or inhibitor. Myogenic differentiation markers, myogenin and myosin heavy chain, were analyzed by qRT-PCR, western blotting analysis, and immunofluorescence staining. The results showed that miR-18a-3p was upregulated (p=0.0002), whereas HuR was downregulated (p=0.002) in the skeletal muscles of patients with IMNM. The expression of miR-18a-3p in patients with IMNM was negatively correlated with those of HuR (r = -0.512, p = 0.029). We also found that disease activity was positively correlated with HuR expression (r = 0.576, p = 0.012) but muscle activity was negatively correlated with miR-18a-3p expression (r = -0.550, p = 0.017). Besides, bioinformatics analysis and dual-luciferase reporter assays suggested that miR-18a-3p could directly target HuR. Cellular experiments showed that overexpression of miR-18a-3p inhibited myogenic differentiation by targeting HuR, whereas inhibition of miR-18a-3p led to opposite results. Therefore, miR-18a-3p and its target protein HuR may be responsible for modulating the myogenic process in IMNM and can thus be therapeutic targets for the same.
Collapse
Affiliation(s)
- Lifang Ye
- Department of Rheumatology, Key Laboratory of Myositis, China-Japan Friendship Hospital, Beijing, China.,Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yu Zuo
- Department of Rheumatology, Key Laboratory of Myositis, China-Japan Friendship Hospital, Beijing, China
| | - Fang Chen
- Department of Rheumatology, Key Laboratory of Myositis, China-Japan Friendship Hospital, Beijing, China
| | - Qinglin Peng
- Department of Rheumatology, Key Laboratory of Myositis, China-Japan Friendship Hospital, Beijing, China
| | - Xin Lu
- Department of Rheumatology, Key Laboratory of Myositis, China-Japan Friendship Hospital, Beijing, China
| | - Guochun Wang
- Department of Rheumatology, Key Laboratory of Myositis, China-Japan Friendship Hospital, Beijing, China.,Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaoming Shu
- Department of Rheumatology, Key Laboratory of Myositis, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
10
|
Song C, Fang X, Yang Z, Wang Q, Meng F, Chen Y, Chen J, Zhao B, Wang Y, Fang X, Gu L, Zhang C. miR-152 Regulates Bovine Myoblast Proliferation by Targeting KLF6. Animals (Basel) 2021; 11:ani11103001. [PMID: 34680020 PMCID: PMC8532817 DOI: 10.3390/ani11103001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/13/2021] [Accepted: 10/16/2021] [Indexed: 01/03/2023] Open
Abstract
Though miRNAs have been reported to regulate bovine myoblast proliferation, but many miRNAs still need to be further explored. Specifically, miR-152 is a highly expressed miRNA in cattle skeletal muscle tissues, but its function in skeletal muscle development is unknown. Herein, we aimed to investigate the role of miR-152 in regulating bovine myoblast proliferation. Functionally, RT-qPCR, Western blotting, EdU assay, and flow cytometry detection results showed that miR-152 inhibited bovine myoblast proliferation. Mechanistically, we demonstrated transcription factor KLF6 was a target gene of miR-152 by means of bioinformatics software prediction and dual-luciferase report analysis, which had been demonstrated to be favorable for myoblast proliferation. Collectively, our research suggested that miR-152 inhibits bovine myoblast proliferation via targeting KLF6.
Collapse
Affiliation(s)
- Chengchuang Song
- Institute of Cellular and Molecular Biology, School of Life Science, Jiangsu Normal University, Xuzhou 221116, China; (C.S.); (X.F.); (Q.W.); (F.M.); (Y.C.); (J.C.); (B.Z.); (Y.W.); (X.F.)
| | - Xue Fang
- Institute of Cellular and Molecular Biology, School of Life Science, Jiangsu Normal University, Xuzhou 221116, China; (C.S.); (X.F.); (Q.W.); (F.M.); (Y.C.); (J.C.); (B.Z.); (Y.W.); (X.F.)
| | - Zhaoxin Yang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China;
| | - Qi Wang
- Institute of Cellular and Molecular Biology, School of Life Science, Jiangsu Normal University, Xuzhou 221116, China; (C.S.); (X.F.); (Q.W.); (F.M.); (Y.C.); (J.C.); (B.Z.); (Y.W.); (X.F.)
| | - Fantong Meng
- Institute of Cellular and Molecular Biology, School of Life Science, Jiangsu Normal University, Xuzhou 221116, China; (C.S.); (X.F.); (Q.W.); (F.M.); (Y.C.); (J.C.); (B.Z.); (Y.W.); (X.F.)
| | - Yaqi Chen
- Institute of Cellular and Molecular Biology, School of Life Science, Jiangsu Normal University, Xuzhou 221116, China; (C.S.); (X.F.); (Q.W.); (F.M.); (Y.C.); (J.C.); (B.Z.); (Y.W.); (X.F.)
| | - Junhao Chen
- Institute of Cellular and Molecular Biology, School of Life Science, Jiangsu Normal University, Xuzhou 221116, China; (C.S.); (X.F.); (Q.W.); (F.M.); (Y.C.); (J.C.); (B.Z.); (Y.W.); (X.F.)
| | - Bei Zhao
- Institute of Cellular and Molecular Biology, School of Life Science, Jiangsu Normal University, Xuzhou 221116, China; (C.S.); (X.F.); (Q.W.); (F.M.); (Y.C.); (J.C.); (B.Z.); (Y.W.); (X.F.)
| | - Yanhong Wang
- Institute of Cellular and Molecular Biology, School of Life Science, Jiangsu Normal University, Xuzhou 221116, China; (C.S.); (X.F.); (Q.W.); (F.M.); (Y.C.); (J.C.); (B.Z.); (Y.W.); (X.F.)
| | - Xingtang Fang
- Institute of Cellular and Molecular Biology, School of Life Science, Jiangsu Normal University, Xuzhou 221116, China; (C.S.); (X.F.); (Q.W.); (F.M.); (Y.C.); (J.C.); (B.Z.); (Y.W.); (X.F.)
| | - Lihong Gu
- Institute of Animal Science & Veterinary Medicine, Hainan Academy of Agricultural Sciences, Haikou 571100, China;
| | - Chunlei Zhang
- Institute of Cellular and Molecular Biology, School of Life Science, Jiangsu Normal University, Xuzhou 221116, China; (C.S.); (X.F.); (Q.W.); (F.M.); (Y.C.); (J.C.); (B.Z.); (Y.W.); (X.F.)
- Correspondence:
| |
Collapse
|
11
|
Huang X, Qu R, Peng Y, Yang Y, Fan T, Sun B, Khan AU, Wu S, Wei K, Xu C, Dai J, Ouyang J, Zhong S. Mechanical Sensing Element PDLIM5 Promotes Osteogenesis of Human Fibroblasts by Affecting the Activity of Microfilaments. Biomolecules 2021; 11:biom11050759. [PMID: 34069539 PMCID: PMC8161207 DOI: 10.3390/biom11050759] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/15/2021] [Accepted: 05/17/2021] [Indexed: 12/23/2022] Open
Abstract
Human skin fibroblasts (HSFs) approximate the multidirectional differentiation potential of mesenchymal stem cells, so they are often used in differentiation, cell cultures, and injury repair. They are an important seed source in the field of bone tissue engineering. However, there are a few studies describing the mechanism of osteogenic differentiation of HSFs. Here, osteogenic induction medium was used to induce fibroblasts to differentiate into osteoblasts, and the role of the mechanical sensitive element PDLIM5 in microfilament-mediated osteogenic differentiation of human fibroblasts was evaluated. The depolymerization of microfilaments inhibited the expression of osteogenesis-related proteins and alkaline phosphatase activity of HSFs, while the polymerization of microfilaments enhanced the osteogenic differentiation of HSFs. The evaluation of potential protein molecules affecting changes in microfilaments showed that during the osteogenic differentiation of HSFs, the expression of PDLIM5 increased with increasing induction time, and decreased under the state of microfilament depolymerization. Lentivirus-mediated PDLIM5 knockdown by shRNA weakened the osteogenic differentiation ability of HSFs and inhibited the expression and morphological changes of microfilament protein. The inhibitory effect of knocking down PDLIM5 on HSF osteogenic differentiation was reversed by a microfilament stabilizer. Taken together, these data suggest that PDLIM5 can mediate the osteogenic differentiation of fibroblasts by affecting the formation and polymerization of microfilaments.
Collapse
Affiliation(s)
- Xiaolan Huang
- Guangdong Provincial Key Laboratory of Medical Biomechanics & Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China; (X.H.); (R.Q.); (Y.P.); (Y.Y.); (T.F.); (B.S.); (A.U.K.); (S.W.)
| | - Rongmei Qu
- Guangdong Provincial Key Laboratory of Medical Biomechanics & Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China; (X.H.); (R.Q.); (Y.P.); (Y.Y.); (T.F.); (B.S.); (A.U.K.); (S.W.)
| | - Yan Peng
- Guangdong Provincial Key Laboratory of Medical Biomechanics & Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China; (X.H.); (R.Q.); (Y.P.); (Y.Y.); (T.F.); (B.S.); (A.U.K.); (S.W.)
| | - Yuchao Yang
- Guangdong Provincial Key Laboratory of Medical Biomechanics & Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China; (X.H.); (R.Q.); (Y.P.); (Y.Y.); (T.F.); (B.S.); (A.U.K.); (S.W.)
| | - Tingyu Fan
- Guangdong Provincial Key Laboratory of Medical Biomechanics & Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China; (X.H.); (R.Q.); (Y.P.); (Y.Y.); (T.F.); (B.S.); (A.U.K.); (S.W.)
| | - Bing Sun
- Guangdong Provincial Key Laboratory of Medical Biomechanics & Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China; (X.H.); (R.Q.); (Y.P.); (Y.Y.); (T.F.); (B.S.); (A.U.K.); (S.W.)
| | - Asmat Ullah Khan
- Guangdong Provincial Key Laboratory of Medical Biomechanics & Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China; (X.H.); (R.Q.); (Y.P.); (Y.Y.); (T.F.); (B.S.); (A.U.K.); (S.W.)
| | - Shutong Wu
- Guangdong Provincial Key Laboratory of Medical Biomechanics & Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China; (X.H.); (R.Q.); (Y.P.); (Y.Y.); (T.F.); (B.S.); (A.U.K.); (S.W.)
| | - Kuanhai Wei
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Guangdong Provincial Key Laboratory of Bone and Cartilage Regeneration Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China;
| | - Chujiang Xu
- Department of Orthopedics, TCM-Integrated Hospital, Southern Medical University, Guangzhou 510000, China;
| | - Jingxing Dai
- Guangdong Provincial Key Laboratory of Medical Biomechanics & Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China; (X.H.); (R.Q.); (Y.P.); (Y.Y.); (T.F.); (B.S.); (A.U.K.); (S.W.)
- Correspondence: (J.D.); (J.O.); (S.Z.); Tel.: +86-(20)-6164-8842 (J.D.); +86-(20)-6164-8199 (J.O.); +86-(20)-6164-8200 (S.Z.)
| | - Jun Ouyang
- Guangdong Provincial Key Laboratory of Medical Biomechanics & Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China; (X.H.); (R.Q.); (Y.P.); (Y.Y.); (T.F.); (B.S.); (A.U.K.); (S.W.)
- Correspondence: (J.D.); (J.O.); (S.Z.); Tel.: +86-(20)-6164-8842 (J.D.); +86-(20)-6164-8199 (J.O.); +86-(20)-6164-8200 (S.Z.)
| | - Shizhen Zhong
- Guangdong Provincial Key Laboratory of Medical Biomechanics & Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China; (X.H.); (R.Q.); (Y.P.); (Y.Y.); (T.F.); (B.S.); (A.U.K.); (S.W.)
- Correspondence: (J.D.); (J.O.); (S.Z.); Tel.: +86-(20)-6164-8842 (J.D.); +86-(20)-6164-8199 (J.O.); +86-(20)-6164-8200 (S.Z.)
| |
Collapse
|
12
|
Aránega AE, Lozano-Velasco E, Rodriguez-Outeiriño L, Ramírez de Acuña F, Franco D, Hernández-Torres F. MiRNAs and Muscle Regeneration: Therapeutic Targets in Duchenne Muscular Dystrophy. Int J Mol Sci 2021; 22:ijms22084236. [PMID: 33921834 PMCID: PMC8072594 DOI: 10.3390/ijms22084236] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 04/15/2021] [Indexed: 12/17/2022] Open
Abstract
microRNAs (miRNAs) are small non-coding RNAs required for the post-transcriptional control of gene expression. MicroRNAs play a critical role in modulating muscle regeneration and stem cell behavior. Muscle regeneration is affected in muscular dystrophies, and a critical point for the development of effective strategies for treating muscle disorders is optimizing approaches to target muscle stem cells in order to increase the ability to regenerate lost tissue. Within this framework, miRNAs are emerging as implicated in muscle stem cell response in neuromuscular disorders and new methodologies to regulate the expression of key microRNAs are coming up. In this review, we summarize recent advances highlighting the potential of miRNAs to be used in conjunction with gene replacement therapies, in order to improve muscle regeneration in the context of Duchenne Muscular Dystrophy (DMD).
Collapse
Affiliation(s)
- Amelia Eva Aránega
- Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaen, Paraje Las Lagunillas s/n, 23009 Jaen, Spain; (E.L.-V.); (L.R.-O.); (F.R.d.A.); (D.F.); (F.H.-T.)
- Medina Foundation, Technology Park of Health Sciences, Av. del Conocimiento 34, 18016 Granada, Spain
- Correspondence:
| | - Estefanía Lozano-Velasco
- Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaen, Paraje Las Lagunillas s/n, 23009 Jaen, Spain; (E.L.-V.); (L.R.-O.); (F.R.d.A.); (D.F.); (F.H.-T.)
- Medina Foundation, Technology Park of Health Sciences, Av. del Conocimiento 34, 18016 Granada, Spain
| | - Lara Rodriguez-Outeiriño
- Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaen, Paraje Las Lagunillas s/n, 23009 Jaen, Spain; (E.L.-V.); (L.R.-O.); (F.R.d.A.); (D.F.); (F.H.-T.)
- Medina Foundation, Technology Park of Health Sciences, Av. del Conocimiento 34, 18016 Granada, Spain
| | - Felicitas Ramírez de Acuña
- Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaen, Paraje Las Lagunillas s/n, 23009 Jaen, Spain; (E.L.-V.); (L.R.-O.); (F.R.d.A.); (D.F.); (F.H.-T.)
- Medina Foundation, Technology Park of Health Sciences, Av. del Conocimiento 34, 18016 Granada, Spain
| | - Diego Franco
- Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaen, Paraje Las Lagunillas s/n, 23009 Jaen, Spain; (E.L.-V.); (L.R.-O.); (F.R.d.A.); (D.F.); (F.H.-T.)
- Medina Foundation, Technology Park of Health Sciences, Av. del Conocimiento 34, 18016 Granada, Spain
| | - Francisco Hernández-Torres
- Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaen, Paraje Las Lagunillas s/n, 23009 Jaen, Spain; (E.L.-V.); (L.R.-O.); (F.R.d.A.); (D.F.); (F.H.-T.)
- Medina Foundation, Technology Park of Health Sciences, Av. del Conocimiento 34, 18016 Granada, Spain
- Department of Biochemistry and Molecular Biology III and Immunology, Faculty of Medicine, University of Granada, Avda. de la Investigación 11, 18016 Granada, Spain
| |
Collapse
|
13
|
PDLIM5 Affects Chicken Skeletal Muscle Satellite Cell Proliferation and Differentiation via the p38-MAPK Pathway. Animals (Basel) 2021; 11:ani11041016. [PMID: 33916517 PMCID: PMC8065989 DOI: 10.3390/ani11041016] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 03/29/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary PDZ and LIM domain 5 (PDLIM5) can increase C2C12 cell differentiation; however, the role of PDLIM5 in chicken skeletal muscle satellite cells (SMSCs) is unclear. In this study, the effect of PDLIM5 was verified on SMSCs in vitro, and then the molecular mechanism was determined by transcriptome sequencing. We demonstrated that PDLIM5 can positively affect chicken SMSC proliferation and differentiation via the p38-MAPK (mitogen activated kinase-like protein) pathway. These results indicate that PDLIM5 may be involved in chicken skeletal muscle growth and development. Abstract Skeletal muscle satellite cell growth and development is a complicated process driven by multiple genes. The PDZ and LIM domain 5 (PDLIM5) gene has been proven to function in C2C12 myoblast differentiation and is involved in the regulation of skeletal muscle development. The role of PDLIM5 in chicken skeletal muscle satellite cells, however, is unclear. In this study, in order to determine whether the PDLIM5 gene has a function in chicken skeletal muscle satellite cells, we examined the changes in proliferation and differentiation of chicken skeletal muscle satellite cells (SMSCs) after interfering and overexpressing PDLIM5 in cells. In addition, the molecular pathways of the PDLIM5 gene regulating SMSC proliferation and differentiation were analyzed by transcriptome sequencing. Our results show that PDLIM5 can promote the proliferation and differentiation of SMSCs; furthermore, through transcriptome sequencing, it can be found that the differential genes are enriched in the MAPK signaling pathway after knocking down PDLIM5. Finally, it was verified that PDLIM5 played an active role in the proliferation and differentiation of chicken SMSCs by activating the p38-MAPK signaling pathway. These results indicate that PDLIM5 may be involved in the growth and development of chicken skeletal muscle.
Collapse
|
14
|
Yang L, Qi Q, Wang J, Song C, Wang Y, Chen X, Chen H, Zhang C, Hu L, Fang X. MiR-452 Regulates C2C12 Myoblast Proliferation and Differentiation via Targeting ANGPT1. Front Genet 2021; 12:640807. [PMID: 33777108 PMCID: PMC7994525 DOI: 10.3389/fgene.2021.640807] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 02/02/2021] [Indexed: 11/26/2022] Open
Abstract
microRNAs are a kind of endogenous, non-coding, single-strand small RNA. They have been reported as an important regulatory factor in skeletal myogenesis. In this study, miR-452 was selected from RNA high-throughput sequencing data to explore its regulatory role in myogenesis. Functionally, miR-452 overexpression could promote C2C12 myoblast proliferation while inhibiting myogenic differentiation. On the contrary, inhibition of miR-452 could suppress C2C12 myoblast proliferation but accelerate myogenic differentiation. Bioinformatics analysis and dual luciferase report assays showed that Angiopoietin 1 (ANGPT1), RB1, and CACNB4 were the potential target genes of miR-452. To further confirm the target relationship between ANGPT1, RB1, and CACNB4 with miR-452, the mRNA level and protein level of these genes were detected by using RT-qPCR and Western blot, respectively. Result analysis indicated that ANGPT1 was a target gene of miR-452. In addition, knockdown of ANGPT1 could obviously promote C2C12 myoblast proliferation but block their differentiation. In summary, these results demonstrated that miR-452 promoted C2C12 myoblast proliferation and inhibited their differentiation via targeting ANGPT1.
Collapse
Affiliation(s)
- Lingzhi Yang
- Institute of Cellular and Molecular Biology, Jiangsu Normal University, Xuzhou, China
| | - Qi Qi
- Institute of Cellular and Molecular Biology, Jiangsu Normal University, Xuzhou, China
| | - Jian Wang
- Institute of Cellular and Molecular Biology, Jiangsu Normal University, Xuzhou, China
| | - Chengchuang Song
- Institute of Cellular and Molecular Biology, Jiangsu Normal University, Xuzhou, China
| | - Yanhong Wang
- Institute of Cellular and Molecular Biology, Jiangsu Normal University, Xuzhou, China
| | - Xi Chen
- Institute of Cellular and Molecular Biology, Jiangsu Normal University, Xuzhou, China
| | - Hong Chen
- Institute of Cellular and Molecular Biology, Jiangsu Normal University, Xuzhou, China
| | - Chunlei Zhang
- Institute of Cellular and Molecular Biology, Jiangsu Normal University, Xuzhou, China
| | - Linyong Hu
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, China
| | - Xingtang Fang
- Institute of Cellular and Molecular Biology, Jiangsu Normal University, Xuzhou, China
| |
Collapse
|
15
|
Dong X, Luo Y, Lu S, Ma H, Zhang W, Zhu Y, Sun G, Sun X. Ursodesoxycholic acid alleviates liver fibrosis via proregeneration by activation of the ID1-WNT2/HGF signaling pathway. Clin Transl Med 2021; 11:e296. [PMID: 33635004 PMCID: PMC7828260 DOI: 10.1002/ctm2.296] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 01/07/2021] [Accepted: 01/10/2021] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND The human liver possesses a remarkable capacity for self-repair. However, liver fibrosis remains a serious medical concern, potentially progressing to end-stage liver cirrhosis and even death. Liver fibrosis is characterized by excess accumulation of extracellular matrix in response to chronic injury. Liver regenerative ability, a strong indicator of liver health, is important in resisting fibrosis. In this study, we provide evidence that ursodesoxycholic acid (UDCA) can alleviate liver fibrosis by promoting liver regeneration via activation of the ID1-WNT2/hepatocyte growth factor (HGF) pathway. METHODS Bile duct ligation (BDL) and partial hepatectomy (PH) mouse models were used to verify the effects of UDCA on liver fibrosis, regeneration, and the ID1-WNT2/HGF pathway. An Id1 knockdown mouse model was also used to assess the role of Id1 in UDCA alleviation of liver fibrosis. RESULTS Our results demonstrate that UDCA can alleviate liver fibrosis in the BDL mice and promote liver regeneration via the ID1-WNT2/HGF pathway in PH mice. In addition, Id1 knockdown abolished the protection afforded by UDCA in BDL mice. CONCLUSIONS We conclude that UDCA protects against liver fibrosis by proregeneration via activation of the ID1-WNT2/HGF pathway.
Collapse
Affiliation(s)
- Xi Dong
- Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational MedicineInstitute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical SciencesBeijing100193P. R. China
- Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine PrescriptionChinese Academy of Medical SciencesBeijing100193P. R. China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant DevelopmentChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100193P. R. China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant DevelopmentPeking Union Medical College and Chinese Academy of Medical SciencesBeijing100193P. R. China
| | - Yun Luo
- Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational MedicineInstitute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical SciencesBeijing100193P. R. China
- Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine PrescriptionChinese Academy of Medical SciencesBeijing100193P. R. China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant DevelopmentChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100193P. R. China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant DevelopmentPeking Union Medical College and Chinese Academy of Medical SciencesBeijing100193P. R. China
| | - Shan Lu
- Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational MedicineInstitute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical SciencesBeijing100193P. R. China
- Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine PrescriptionChinese Academy of Medical SciencesBeijing100193P. R. China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant DevelopmentChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100193P. R. China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant DevelopmentPeking Union Medical College and Chinese Academy of Medical SciencesBeijing100193P. R. China
| | - Han Ma
- School of Traditional Chinese MedicineCapital Medical UniversityBeijingP. R. China
| | - Wenchao Zhang
- College of Life Science and TechnologyBeijing University of Chemical TechnologyBeijingP. R. China
| | - Yue Zhu
- Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational MedicineInstitute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical SciencesBeijing100193P. R. China
- Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine PrescriptionChinese Academy of Medical SciencesBeijing100193P. R. China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant DevelopmentChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100193P. R. China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant DevelopmentPeking Union Medical College and Chinese Academy of Medical SciencesBeijing100193P. R. China
| | - Guibo Sun
- Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational MedicineInstitute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical SciencesBeijing100193P. R. China
- Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine PrescriptionChinese Academy of Medical SciencesBeijing100193P. R. China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant DevelopmentChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100193P. R. China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant DevelopmentPeking Union Medical College and Chinese Academy of Medical SciencesBeijing100193P. R. China
| | - Xiaobo Sun
- Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational MedicineInstitute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical SciencesBeijing100193P. R. China
- Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine PrescriptionChinese Academy of Medical SciencesBeijing100193P. R. China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant DevelopmentChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100193P. R. China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant DevelopmentPeking Union Medical College and Chinese Academy of Medical SciencesBeijing100193P. R. China
| |
Collapse
|
16
|
Luo H, Lv W, Tong Q, Jin J, Xu Z, Zuo B. Functional Non-coding RNA During Embryonic Myogenesis and Postnatal Muscle Development and Disease. Front Cell Dev Biol 2021; 9:628339. [PMID: 33585483 PMCID: PMC7876409 DOI: 10.3389/fcell.2021.628339] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 01/06/2021] [Indexed: 12/19/2022] Open
Abstract
Skeletal muscle is a highly heterogeneous tissue that plays a crucial role in mammalian metabolism and motion maintenance. Myogenesis is a complex biological process that includes embryonic and postnatal development, which is regulated by specific signaling pathways and transcription factors. Various non-coding RNAs (ncRNAs) account for the majority of total RNA in cells and have an important regulatory role in myogenesis. In this review, we introduced the research progress in miRNAs, circRNAs, and lncRNAs related to embryonic and postnatal muscle development. We mainly focused on ncRNAs that regulate myoblast proliferation, differentiation, and postnatal muscle development through multiple mechanisms. Finally, challenges and future perspectives related to the identification and verification of functional ncRNAs are discussed. The identification and elucidation of ncRNAs related to myogenesis will enrich the myogenic regulatory network, and the effective application of ncRNAs will enhance the function of skeletal muscle.
Collapse
Affiliation(s)
- Hongmei Luo
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan, China
| | - Wei Lv
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan, China
| | - Qian Tong
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan, China
| | - Jianjun Jin
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan, China
| | - Zaiyan Xu
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, China.,Department of Basic Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Bo Zuo
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| |
Collapse
|
17
|
He C, He W, Hou J, Chen K, Huang M, Yang M, Luo X, Li C. Bone and Muscle Crosstalk in Aging. Front Cell Dev Biol 2020; 8:585644. [PMID: 33363144 PMCID: PMC7758235 DOI: 10.3389/fcell.2020.585644] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 11/09/2020] [Indexed: 12/12/2022] Open
Abstract
Osteoporosis and sarcopenia are two age-related diseases that affect the quality of life in the elderly. Initially, they were thought to be two independent diseases; however, recently, increasing basic and clinical data suggest that skeletal muscle and bone are both spatially and metabolically connected. The term "osteosarcopenia" is used to define a condition of synergy of low bone mineral density with muscle atrophy and hypofunction. Bone and muscle cells secrete several factors, such as cytokines, myokines, and osteokines, into the circulation to influence the biological and pathological activities in local and distant organs and cells. Recent studies reveal that extracellular vesicles containing microRNAs derived from senescent skeletal muscle and bone cells can also be transported and aid in regulating bone-muscle crosstalk. In this review, we summarize the age-related changes in the secretome and extracellular vesicle-microRNAs secreted by the muscle and bone, and discuss their interactions between muscle and bone cells during aging.
Collapse
Affiliation(s)
- Chen He
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Wenzhen He
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Jing Hou
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Kaixuan Chen
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Mei Huang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Mi Yang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Xianghang Luo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital of Central South University, Changsha, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
| | - Changjun Li
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital of Central South University, Changsha, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
| |
Collapse
|
18
|
Singh GB, Cowan DB, Wang DZ. Tiny Regulators of Massive Tissue: MicroRNAs in Skeletal Muscle Development, Myopathies, and Cancer Cachexia. Front Oncol 2020; 10:598964. [PMID: 33330096 PMCID: PMC7719840 DOI: 10.3389/fonc.2020.598964] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 10/29/2020] [Indexed: 12/13/2022] Open
Abstract
Skeletal muscles are the largest tissues in our body and the physiological function of muscle is essential to every aspect of life. The regulation of development, homeostasis, and metabolism is critical for the proper functioning of skeletal muscle. Consequently, understanding the processes involved in the regulation of myogenesis is of great interest. Non-coding RNAs especially microRNAs (miRNAs) are important regulators of gene expression and function. MiRNAs are small (~22 nucleotides long) noncoding RNAs known to negatively regulate target gene expression post-transcriptionally and are abundantly expressed in skeletal muscle. Gain- and loss-of function studies have revealed important roles of this class of small molecules in muscle biology and disease. In this review, we summarize the latest research that explores the role of miRNAs in skeletal muscle development, gene expression, and function as well as in muscle disorders like sarcopenia and Duchenne muscular dystrophy (DMD). Continuing with the theme of the current review series, we also briefly discuss the role of miRNAs in cancer cachexia.
Collapse
Affiliation(s)
- Gurinder Bir Singh
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Douglas B Cowan
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Da-Zhi Wang
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
19
|
Huang X, Qu R, Ouyang J, Zhong S, Dai J. An Overview of the Cytoskeleton-Associated Role of PDLIM5. Front Physiol 2020; 11:975. [PMID: 32848888 PMCID: PMC7426503 DOI: 10.3389/fphys.2020.00975] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 07/16/2020] [Indexed: 01/08/2023] Open
Abstract
Regenerative medicine represented by stem cell technology has become one of the pillar medical technologies for human disease treatment. Cytoskeleton plays important roles in maintaining cell morphology, bearing external forces, and maintaining the effectiveness of cell internal structure, among which cytoskeleton related proteins are involved in and play an indispensable role in the changes of cytoskeleton. PDLIM5 is a cytoskeleton-related protein that, like other cytoskeletal proteins, acts as a binding protein. PDZ and LIM domain 5 (PDLIM5), also known as ENH (Enigma homolog), is a cytoplasmic protein with a molecular mass of about 63 KDa that consists of a PDZ domain at the N-terminus and three LIM domains at the C-terminus. PDLIM5 binds to the cytoskeleton and membrane proteins through its PDZ domain and interacts with various signaling molecules, including protein kinases and transcription factors, through its LIM domain. As a cytoskeleton-related protein, PDLIM5 plays an important role in regulating cell proliferation, differentiation and cell fate decision in multiple tissues and cell types. In this review, we briefly summarize the state of knowledge on the PDLIM5 gene, structural properties, and molecular functional mechanisms of the PDLIM5 protein, and its role in cells, tissues, and organ systems, and describe the possible underlying molecular signaling pathways. In the last part of this review, we will focus on discussing the limitations of existing research and the future prospects of PDLIM5 research in turn.
Collapse
Affiliation(s)
- Xiaolan Huang
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Rongmei Qu
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jun Ouyang
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Shizhen Zhong
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jingxing Dai
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
20
|
Chen X, Zhao C, Dou M, Sun Y, Yu T, Pang W, Yang G. Deciphering the miRNA transcriptome of Rongchang pig longissimus dorsi at weaning and slaughter time points. J Anim Physiol Anim Nutr (Berl) 2020; 104:954-964. [PMID: 32056287 DOI: 10.1111/jpn.13314] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 12/26/2019] [Accepted: 12/28/2019] [Indexed: 11/28/2022]
Abstract
MicroRNA (miRNA) is essential for the process of gene posttranscriptional regulation in skeletal muscle of many species, such as mice, cattle and so on. However, a little number of miRNAs have been reported in the muscle development of Chinese native pig breeds. In this study, the longissimus dorsi transcripts of Chinese native Rongchang pig at weaning and slaughter time points were analysed for miRNA-seq. The results showed that 19 novel and 186 known miRNAs involved in the Rongchang pig skeletal muscle development were identified. Based on these findings, we further confirmed that porcine miR-127, miR-299 and miR-432-5p were obviously down-expressed in adult pig (287 days of age), while miR-7134-3p and 664-5p were significantly up-expressed in weaning pig (35 days of age). In other words, these miRNAs could be the potential molecular markers and play vital roles in the muscle development process. Moreover, we found miR-127 could inhibit the proliferation and myogenesis of porcine satellite cells in longissimus dorsi muscle. Our findings will provide deep insight into miRNA function for pork quality research with Chinese indigenous pig breeds.
Collapse
Affiliation(s)
- Xiaochang Chen
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China.,Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Chen Zhao
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China.,Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Mingle Dou
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China.,Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Yunmei Sun
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China.,Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Taiyong Yu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China.,Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Weijun Pang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China.,Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Gongshe Yang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China.,Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| |
Collapse
|
21
|
Li F, Liu WC, Wang Q, Sun Y, Wang H, Jin X. NG2-glia cell proliferation and differentiation by glial growth factor 2 (GGF2), a strategy to promote functional recovery after ischemic stroke. Biochem Pharmacol 2019; 171:113720. [PMID: 31751533 DOI: 10.1016/j.bcp.2019.113720] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Accepted: 11/14/2019] [Indexed: 12/16/2022]
Abstract
Stroke is the leading cause of adult disability. Spontaneous functional recovery occurs after ischemic stroke, but it is very limited. Therefore, it is urgent to find a strategy to promote functional recovery after stroke in clinical setting. Gray matter damage has received extensive attention owing to the important roles of the gray matter in synaptic plasticity, cognitive, and motor function. However, stroke also causes white matter damage, which accounts for half of the infarct volume and can be aggravated by blood brain barrier damage. Disruption of white matter integrity, which is characterized by death of oligodendrocytes (OLs), loss of myelin, and axonal injury, greatly contributes to impaired neurological function. Impaired proliferation and differentiation of OL precursor cell (OPC, NG2-glia cells) play an important role in limited functional recovery after ischemic stroke and inhibitor of differentiation 2 (ID2) is a key factor controlling NG2-glia cells differentiation. It has been reported that the number of NG2-glia cells in the peri-infarction area significantly increases after ischemic stroke and glial growth factor (GGF2) administration promotes the proliferation and differentiation of NG2-glia cells as well as functional recovery after spinal cord injury. On the basis of the important roles of GGF2 in functional recovery and those of ID2 in NG2-glia cell proliferation and differentiation, we propose that after binding with the ErBb receptor on the surface of NG2-glia cells, GGF2 promotes NG2-glia cell proliferation and differentiation, thereby repairing BBB and white matter integrity and promoting neural functional recovery after ischemic stroke.
Collapse
Affiliation(s)
- Fei Li
- School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan 442000, China; Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan 442000, China
| | - Wen-Cao Liu
- Shanxi Provincial People's Hospital, Taiyuan 030001, China
| | - Qi Wang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Yanyun Sun
- Jiangsu Key Laboratory of Neuro-Psychiatry Research and Institute of Neuroscience, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China.
| | - Hongbo Wang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation, School of Pharmacy, Yantai University, Yantai, China.
| | - Xinchun Jin
- Jiangsu Key Laboratory of Neuro-Psychiatry Research and Institute of Neuroscience, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China; Key Laboratory of Molecular Pharmacology and Drug Evaluation, School of Pharmacy, Yantai University, Yantai, China.
| |
Collapse
|
22
|
Conditional Deletion of Dicer in Adult Mice Impairs Skeletal Muscle Regeneration. Int J Mol Sci 2019; 20:ijms20225686. [PMID: 31766249 PMCID: PMC6887938 DOI: 10.3390/ijms20225686] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 11/08/2019] [Accepted: 11/12/2019] [Indexed: 12/14/2022] Open
Abstract
Skeletal muscle has a remarkable regenerative capacity, which is orchestrated by multiple processes, including the proliferation, fusion, and differentiation of the resident stem cells in muscle. MicroRNAs (miRNAs) are small noncoding RNAs that mediate the translational repression or degradation of mRNA to regulate diverse biological functions. Previous studies have suggested that several miRNAs play important roles in myoblast proliferation and differentiation in vitro. However, their potential roles in skeletal muscle regeneration in vivo have not been fully established. In this study, we generated a mouse in which the Dicer gene, which encodes an enzyme essential in miRNA processing, was knocked out in a tamoxifen-inducible way (iDicer KO mouse) and determined its regenerative potential after cardiotoxin-induced acute muscle injury. Dicer mRNA expression was significantly reduced in the tibialis anterior muscle of the iDicer KO mice, whereas the expression of muscle-enriched miRNAs was only slightly reduced in the Dicer-deficient muscles. After cardiotoxin injection, the iDicer KO mice showed impaired muscle regeneration. We also demonstrated that the number of PAX7+ cells, cell proliferation, and the myogenic differentiation capacity of the primary myoblasts did not differ between the wild-type and the iDicer KO mice. Taken together, these data demonstrate that Dicer is a critical factor for muscle regeneration in vivo.
Collapse
|
23
|
Robinson KA, Baker LA, Graham-Brown MPM, Watson EL. Skeletal muscle wasting in chronic kidney disease: the emerging role of microRNAs. Nephrol Dial Transplant 2019; 35:1469-1478. [DOI: 10.1093/ndt/gfz193] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 08/27/2019] [Indexed: 12/17/2022] Open
Abstract
Abstract
Skeletal muscle wasting is a common complication of chronic kidney disease (CKD), characterized by the loss of muscle mass, strength and function, which significantly increases the risk of morbidity and mortality in this population. Numerous complications associated with declining renal function and lifestyle activate catabolic pathways and impair muscle regeneration, resulting in substantial protein wasting. Evidence suggests that increasing skeletal muscle mass improves outcomes in CKD, making this a clinically important research focus. Despite extensive research, the pathogenesis of skeletal muscle wasting is not completely understood. It is widely recognized that microRNAs (miRNAs), a family of short non-coding RNAs, are pivotal in the regulation of skeletal muscle homoeostasis, with significant roles in regulating muscle growth, regeneration and metabolism. The abnormal expression of miRNAs in skeletal muscle during disease has been well described in cellular and animal models of muscle atrophy, and in recent years, the involvement of miRNAs in the regulation of muscle atrophy in CKD has been demonstrated. As this exciting field evolves, there is emerging evidence for the involvement of miRNAs in a beneficial crosstalk system between skeletal muscle and other organs that may potentially limit the progression of CKD. In this article, we describe the pathophysiological mechanisms of muscle wasting and explore the contribution of miRNAs to the development of muscle wasting in CKD. We also discuss advances in our understanding of miRNAs in muscle–organ crosstalk and summarize miRNA-based therapeutics currently in clinical trials.
Collapse
Affiliation(s)
- Kate A Robinson
- Department of Infection Immunity and Inflammation, College of Life Sciences, University of Leicester, Leicester, UK
| | - Luke A Baker
- Department of Health Sciences, College of Life Sciences, George Davies Centre, University of Leicester, Leicester, UK
| | - Matthew P M Graham-Brown
- Department of Cardiovascular Sciences, University of Leicester and NIHR Leicester Cardiovascular Biomedical Research Centre, Glenfield Hospital Leicester, Leicester, UK
| | - Emma L Watson
- Department of Cardiovascular Sciences, University of Leicester and NIHR Leicester Cardiovascular Biomedical Research Centre, Glenfield Hospital Leicester, Leicester, UK
| |
Collapse
|
24
|
Bonnet S, Boucherat O, Paulin R, Wu D, Hindmarch CCT, Archer SL, Song R, Moore JB, Provencher S, Zhang L, Uchida S. Clinical value of non-coding RNAs in cardiovascular, pulmonary, and muscle diseases. Am J Physiol Cell Physiol 2019; 318:C1-C28. [PMID: 31483703 DOI: 10.1152/ajpcell.00078.2019] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Although a majority of the mammalian genome is transcribed to RNA, mounting evidence indicates that only a minor proportion of these transcriptional products are actually translated into proteins. Since the discovery of the first non-coding RNA (ncRNA) in the 1980s, the field has gone on to recognize ncRNAs as important molecular regulators of RNA activity and protein function, knowledge of which has stimulated the expansion of a scientific field that quests to understand the role of ncRNAs in cellular physiology, tissue homeostasis, and human disease. Although our knowledge of these molecules has significantly improved over the years, we have limited understanding of their precise functions, protein interacting partners, and tissue-specific activities. Adding to this complexity, it remains unknown exactly how many ncRNAs there are in existence. The increased use of high-throughput transcriptomics techniques has rapidly expanded the list of ncRNAs, which now includes classical ncRNAs (e.g., ribosomal RNAs and transfer RNAs), microRNAs, and long ncRNAs. In addition, splicing by-products of protein-coding genes and ncRNAs, so-called circular RNAs, are now being investigated. Because there is substantial heterogeneity in the functions of ncRNAs, we have summarized the present state of knowledge regarding the functions of ncRNAs in heart, lungs, and skeletal muscle. This review highlights the pathophysiologic relevance of these ncRNAs in the context of human cardiovascular, pulmonary, and muscle diseases.
Collapse
Affiliation(s)
- Sébastien Bonnet
- Pulmonary Hypertension and Vascular Biology Research Group, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Department of Medicine, Université Laval, Quebec City, Quebec, Canada.,Department of Medicine, Université Laval, Quebec City, Quebec, Canada
| | - Olivier Boucherat
- Pulmonary Hypertension and Vascular Biology Research Group, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Department of Medicine, Université Laval, Quebec City, Quebec, Canada.,Department of Medicine, Université Laval, Quebec City, Quebec, Canada
| | - Roxane Paulin
- Pulmonary Hypertension and Vascular Biology Research Group, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Department of Medicine, Université Laval, Quebec City, Quebec, Canada.,Department of Medicine, Université Laval, Quebec City, Quebec, Canada
| | - Danchen Wu
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Charles C T Hindmarch
- Queen's Cardiopulmonary Unit, Translational Institute of Medicine, Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Stephen L Archer
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Rui Song
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Joseph B Moore
- Diabetes and Obesity Center, University of Louisville, Louisville, Kentucky.,The Christina Lee Brown Envirome Institute, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Steeve Provencher
- Pulmonary Hypertension and Vascular Biology Research Group, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Department of Medicine, Université Laval, Quebec City, Quebec, Canada.,Department of Medicine, Université Laval, Quebec City, Quebec, Canada
| | - Lubo Zhang
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Shizuka Uchida
- Diabetes and Obesity Center, University of Louisville, Louisville, Kentucky.,The Christina Lee Brown Envirome Institute, Department of Medicine, University of Louisville, Louisville, Kentucky.,Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky
| |
Collapse
|
25
|
A novel mitochondrial micropeptide MPM enhances mitochondrial respiratory activity and promotes myogenic differentiation. Cell Death Dis 2019; 10:528. [PMID: 31296841 PMCID: PMC6624212 DOI: 10.1038/s41419-019-1767-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 05/25/2019] [Accepted: 06/26/2019] [Indexed: 12/14/2022]
Abstract
Micropeptides belong to a class of newly identified small molecules with <100 amino acids in length, and their functions remain largely unknown. Here, we identified a novel muscle-enriched micropeptide that was localized to mitochondria (named MPM, micropeptide in mitochondria) and upregulated during in vitro differentiation of C2C12 myoblasts and in vivo early postnatal skeletal muscle development, and muscle regeneration after cardiotoxin (CTX) damage. Downregulation of MPM was observed in the muscular tissues of tibial muscular dystrophy and Duchenne muscular dystrophy patients. Furthermore, MPM silencing inhibited the differentiation of C2C12 myoblasts into myotubes, whereas MPM overexpression stimulated it. MPM−/− mice exhibited smaller skeletal muscle fibers and worse muscle performance, such as decrease in the maximum grip force of limbs, the latency to fall off rotarod, and the exhausting swimming time. Muscle regeneration was also impaired in MPM−/− mice, as evidenced by lower expression of Pax7, MyoD, and MyoG after CTX injection and smaller regenerated myofibers, compared with wild-type mice. Mechanistical investigations based on both gain- and loss-of function studies revealed that MPM increased oxygen consumption and ATP production of mitochondria. Moreover, ectopic expression of PGC-1α, which can enhance mitochondrial respiration, attenuated the inhibitory effect of siMPM on myogenic differentiation. These results imply that MPM may promote myogenic differentiation and muscle fiber growth by enhancing mitochondrial respiratory activity, which highlights the importance of micropeptides in the elaborate regulatory network of both myogenesis and mitochondrial activity and implicates MPM as a potential target for muscular dystrophy therapy.
Collapse
|
26
|
Xu M, Chen X, Chen D, Yu B, Li M, He J, Huang Z. Regulation of skeletal myogenesis by microRNAs. J Cell Physiol 2019; 235:87-104. [DOI: 10.1002/jcp.28986] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 05/31/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Meng Xu
- Key Laboratory for Animal Disease‐Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition Sichuan Agricultural University Chengdu Sichuan China
| | - Xiaoling Chen
- Key Laboratory for Animal Disease‐Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition Sichuan Agricultural University Chengdu Sichuan China
| | - Daiwen Chen
- Key Laboratory for Animal Disease‐Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition Sichuan Agricultural University Chengdu Sichuan China
| | - Bing Yu
- Key Laboratory for Animal Disease‐Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition Sichuan Agricultural University Chengdu Sichuan China
| | - Mingzhou Li
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology Sichuan Agricultural University Chengdu Sichuan China
| | - Jun He
- Key Laboratory for Animal Disease‐Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition Sichuan Agricultural University Chengdu Sichuan China
| | - Zhiqing Huang
- Key Laboratory for Animal Disease‐Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition Sichuan Agricultural University Chengdu Sichuan China
| |
Collapse
|
27
|
Kong D, He M, Yang L, Zhou R, Yan YQ, Liang Y, Teng CB. MiR-17 and miR-19 cooperatively promote skeletal muscle cell differentiation. Cell Mol Life Sci 2019; 76:5041-5054. [PMID: 31214725 PMCID: PMC6881278 DOI: 10.1007/s00018-019-03165-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 05/15/2019] [Accepted: 05/28/2019] [Indexed: 12/11/2022]
Abstract
Skeletal myogenesis is a highly coordinated process that involves cell proliferation, differentiation and fusion controlled by a complex gene regulatory network. The microRNA gene cluster miR-17–92 has been shown to be related to this process; however, the exact role of each cluster member remains unclear. Here, we show that miR-17 and miR-20a could effectively promote the differentiation of both C2C12 myoblasts and primary bovine satellite cells. In contrast, miR-18a might play a negative role in C2C12 cell differentiation, while miR-19 and miR-92a had little influence. Transcriptome and target analyses revealed that miR-17 could act on Ccnd2, Jak1 and Rhoc genes that are critical for cell proliferation and/or fusion. Notably, the addition of miR-19 could reverse the lethal effect of miR-17 and could thus facilitate the maturation of myotubes. Furthermore, by co-injecting the lentiviral shRNAs of miR-17 and miR-19 into mouse tibialis anterior muscles, we demonstrated the wound healing abilities of the two miRNAs. Our findings indicate that in combination with miR-19, miR-17 is a potent inducer of skeletal muscle differentiation.
Collapse
Affiliation(s)
- Delin Kong
- College of Life Science, Northeast Forestry University, Harbin, 150040, China
| | - Mei He
- College of Life Science, Northeast Forestry University, Harbin, 150040, China
| | - Lin Yang
- College of Life Science, Northeast Forestry University, Harbin, 150040, China
| | - Rongtao Zhou
- College of Life Science, Northeast Forestry University, Harbin, 150040, China
| | - Yun-Qin Yan
- The Laboratory of Cell and Developmental Biology, Northeast Agricultural University, Harbin, China
| | - Yang Liang
- College of Life Science, Northeast Forestry University, Harbin, 150040, China.
| | - Chun-Bo Teng
- College of Life Science, Northeast Forestry University, Harbin, 150040, China.
| |
Collapse
|
28
|
Jang YJ, Ahn J, Son HJ, Jung CH, Ahn J, Ha TY. Hydrangea serrata
Tea Enhances Running Endurance and Skeletal Muscle Mass. Mol Nutr Food Res 2019; 63:e1801149. [DOI: 10.1002/mnfr.201801149] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 04/29/2019] [Indexed: 11/11/2022]
Affiliation(s)
- Young Jin Jang
- Research Group of Natural Materials and MetabolismKorea Food Research Institute Wanju‐gun 55365 Republic of Korea
| | - Jisong Ahn
- Research Group of Natural Materials and MetabolismKorea Food Research Institute Wanju‐gun 55365 Republic of Korea
- Department of Food Science and TechnologyChonbuk National University Jeonju‐si 54896 Republic of Korea
| | - Hyo Jeong Son
- Research Group of Natural Materials and MetabolismKorea Food Research Institute Wanju‐gun 55365 Republic of Korea
| | - Chang Hwa Jung
- Research Group of Natural Materials and MetabolismKorea Food Research Institute Wanju‐gun 55365 Republic of Korea
- Division of Food BiotechnologyUniversity of Science and Technology Daejeon 34113 Republic of Korea
| | - Jiyun Ahn
- Research Group of Natural Materials and MetabolismKorea Food Research Institute Wanju‐gun 55365 Republic of Korea
- Division of Food BiotechnologyUniversity of Science and Technology Daejeon 34113 Republic of Korea
| | - Tae Youl Ha
- Research Group of Natural Materials and MetabolismKorea Food Research Institute Wanju‐gun 55365 Republic of Korea
- Division of Food BiotechnologyUniversity of Science and Technology Daejeon 34113 Republic of Korea
| |
Collapse
|
29
|
Pan WL, Chopp M, Fan B, Zhang R, Wang X, Hu J, Zhang XM, Zhang ZG, Liu XS. Ablation of the microRNA-17-92 cluster in neural stem cells diminishes adult hippocampal neurogenesis and cognitive function. FASEB J 2019; 33:5257-5267. [PMID: 30668139 DOI: 10.1096/fj.201801019r] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Impairment of adult neurogenesis in the hippocampus causes cognitive deficits; however, the underlying molecular mechanisms have not been fully elucidated. microRNAs (miRNAs) regulate neural stem cell (NSC) function. With the use of a transgenic mouse line with conditional ablation of the miR-17-92 cluster in nestin lineage NSCs, we tested the hypothesis that the miR-17-92 cluster regulates adult neurogenesis and cognitive function in vivo. Compared with wild-type mice, ablation of the miR-17-92 cluster significantly reduced the number of proliferating NSCs and neuroblasts and neuronal differentiation in the dentate gyrus (DG) of the hippocampus and significantly impaired hippocampal-dependent learning and memory, as assayed by social recognition memory, novel object recognition, and Morris water-maze tests. Statistical analysis showed a highly significant correlation between newly generated neuroblasts in the DG and cognition deficits in miR-17-92 knockout (KO) mice. Western blot analysis showed that conditional KO of the miR-17-92 cluster significantly increased and reduced a cytoskeleton-associated protein, Enigma homolog 1 (ENH1), and its downstream transcription factor, inhibitor of differentiation 1 (ID1), respectively, as well as increased phosphatase and tensin homolog gene. These proteins are related to neuronal differentiation. Our study demonstrates that the miR-17-92 cluster in NSCs is critical for cognitive and behavioral function and regulates neurogenesis and that the miR-17-92 cluster may target ENH1/ID1 signaling.-Pan, W. L., Chopp, M., Fan, B., Zhang, R., Wang, X., Hu, J., Zhang, X. M., Zhang, Z. G., Liu, X. S. Ablation of the microRNA-17-92 cluster in neural stem cells diminishes adult hippocampal neurogenesis and cognitive function.
Collapse
Affiliation(s)
- Wan Long Pan
- Department of Microbiology and Immunology, Sichuan Key Laboratory of Medical Imaging, Affiliated Hospital of North Sichuan Medical College, Nanchong, China.,Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA.,Department of Physics, Oakland University, Rochester, Michigan, USA
| | - Baoyan Fan
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
| | - Ruilan Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
| | - Xinli Wang
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
| | - Jiani Hu
- Department of Radiology, Wayne State University, Detroit, Michigan, USA
| | - Xiao Ming Zhang
- Department of Radiology, Sichuan Key Laboratory of Medical Imaging, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Zheng Gang Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
| | - Xian Shuang Liu
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
| |
Collapse
|
30
|
Huang D, Peng Y, Ma K, Deng X, Tang L, Jing D, Shao Z. MiR-20a, a novel promising biomarker to predict prognosis in human cancer: a meta-analysis. BMC Cancer 2018; 18:1189. [PMID: 30497428 PMCID: PMC6267918 DOI: 10.1186/s12885-018-4907-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Accepted: 10/08/2018] [Indexed: 02/09/2023] Open
Abstract
Background Recently, microRNA-20a (miR-20a) has been reported to influence the clinical features and may have prognostic value in human cancers. The present meta-analysis assessed the prognostic role of miR-20a in various carcinomas. Methods Literature searches of seven electronic databases were performed for eligible articles of the prognostic role of miR-20a in human cancers. Hazard ratios (HR) for overall survival (OS), disease free survival (DFS), progression-free survival (PFS) as well as their 95% confidence intervals (95%CIs) were used to assess the influence of miR-20a expression on patient prognosis. Odds ratio (OR) and 95%CIs were applied to evaluate the correlation between miR-20a expression and clinicopathological characteristics. Results Based on the OS analyzed by log rank tests, there was a significant association between miR-20a levels and OS by fixed effects model. By subgroup analyses, the significance was also observed in the studies of specimen derived from blood and gastrointestinal cancer group. The independent prognostic role of miR-20a expression for the OS was observed significantly by fixed effects model. In addition, we observed significant association between miR-20a expression levels and DFS of log rank tests, DFS of cox regression. Significant relation of gender/differentiation and the expression level of miR-20a was identified. Conclusions Base on the findings, the elevated miR-20a expression level is related to poor prognosis of gastrointestinal cancer patients. As for other types of carcinomas, the results are still not stable and more studies are required to further identify miR-20a prognostic values. In addition, miR-20a expression level is relatively higher in women than that in men, and increased miR-20a expression level is linked to poor tumor differentiation.
Collapse
Affiliation(s)
- Donghua Huang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yizhong Peng
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Kaige Ma
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiangyu Deng
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Lu Tang
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Doudou Jing
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zengwu Shao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
31
|
Zhou S, Li S, Zhang W, Tong H, Li S, Yan Y. MiR-139 promotes differentiation of bovine skeletal muscle-derived satellite cells by regulating DHFR gene expression. J Cell Physiol 2018; 234:632-641. [PMID: 30078180 DOI: 10.1002/jcp.26817] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Accepted: 04/30/2018] [Indexed: 11/09/2022]
Abstract
MicroRNAs play an important regulatory role in the proliferation and differentiation of skeletal muscle-derived satellite cells (MDSCs). In particular, miR-139 can inhibit tumor cell proliferation and invasion, and its expression is down-regulated during C2C12 myoblast differentiation. The aim of this study was thus to examine the effect and potential mechanism of miR-139 in bovine MDSCs. The expression of miR-139 was found to be significantly increased during bovine MDSC differentiation by stem-loop reverse transcription-polymerase chain reaction amplification. Statistical analysis of the myotube fusion rate was done through immunofluorescence detection of desmin, and western blotting was used to measure the change in protein expression of the muscle differentiation marker genes MYOG and MYH3. The results showed that the miR-139 mimic could enhance the differentiation of bovine MDSCs, whereas the inhibitor had the opposite effect. By using the dual-luciferase reporter system, miR-139 was found to target the 3'-untranslated region of the dihydrofolate reductase (DHFR) gene and regulate its expression. In addition, the expression of miR-139 was found to be regulated by its host gene phosphodiesterase 2A (PDE2A) via inhibition of the latter by CRISPR interference (CRISPRi). Overall, our findings indicate that miR-139 plays an important role in regulating the differentiation of bovine MDSCs.
Collapse
Affiliation(s)
- Shuang Zhou
- The Laboratory of Cell and Development, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Shuang Li
- The Laboratory of Cell and Development, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Weiwei Zhang
- Department of Life Science and Agroforestry, Qiqihar University, Quqihar, Heilongjiang, China
| | - Huili Tong
- The Laboratory of Cell and Development, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Shufeng Li
- The Laboratory of Cell and Development, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Yunqin Yan
- The Laboratory of Cell and Development, Northeast Agricultural University, Harbin, Heilongjiang, China
| |
Collapse
|
32
|
Zhang W, Xu Y, Zhang L, Wang S, Yin B, Zhao S, Li X. Synergistic effects of TGFβ2, WNT9a, and FGFR4 signals attenuate satellite cell differentiation during skeletal muscle development. Aging Cell 2018; 17:e12788. [PMID: 29869452 PMCID: PMC6052404 DOI: 10.1111/acel.12788] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2018] [Indexed: 11/28/2022] Open
Abstract
Satellite cells play a key role in the aging, generation, and damage repair of skeletal muscle. The molecular mechanism of satellite cells in these processes remains largely unknown. This study systematically investigated for the first time the characteristics of mouse satellite cells at ten different ages. Results indicated that the number and differentiation capacity of satellite cells decreased with age during skeletal muscle development. Transcriptome analysis revealed that 2,907 genes were differentially expressed at six time points at postnatal stage. WGCNA and GO analysis indicated that 1,739 of the 2,907 DEGs were mainly involved in skeletal muscle development processes. Moreover, the results of WGCNA and protein interaction analysis demonstrated that Tgfβ2, Wnt9a, and Fgfr4 were the key genes responsible for the differentiation of satellite cells. Functional analysis showed that TGFβ2 and WNT9a inhibited, whereas FGFR4 promoted the differentiation of satellite cells. Furthermore, each two of them had a regulatory relationship at the protein level. In vivo study also confirmed that TGFβ2 could regulate the regeneration of skeletal muscle, as well as the expression of WNT9a and FGFR4. Therefore, we concluded that the synergistic effects of TGFβ2, WNT9a, and FGFR4 were responsible for attenuating of the differentiation of aging satellite cells during skeletal muscle development. This study provided new insights into the molecular mechanism of satellite cell development. The target genes and signaling pathways investigated in this study would be useful for improving the muscle growth of livestock or treating muscle diseases in clinical settings.
Collapse
Affiliation(s)
- Weiya Zhang
- Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of the Ministry of Education & Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture; Huazhong Agricultural University; Wuhan China
| | - Yueyuan Xu
- Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of the Ministry of Education & Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture; Huazhong Agricultural University; Wuhan China
| | - Lu Zhang
- Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of the Ministry of Education & Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture; Huazhong Agricultural University; Wuhan China
| | - Sheng Wang
- Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of the Ministry of Education & Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture; Huazhong Agricultural University; Wuhan China
| | - Binxu Yin
- Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of the Ministry of Education & Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture; Huazhong Agricultural University; Wuhan China
| | - Shuhong Zhao
- Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of the Ministry of Education & Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture; Huazhong Agricultural University; Wuhan China
- The Cooperative Innovation Center for Sustainable Pig Production; Wuhan China
| | - Xinyun Li
- Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of the Ministry of Education & Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture; Huazhong Agricultural University; Wuhan China
- The Cooperative Innovation Center for Sustainable Pig Production; Wuhan China
| |
Collapse
|
33
|
MicroRNA-106a-5p Inhibited C2C12 Myogenesis via Targeting PIK3R1 and Modulating the PI3K/AKT Signaling. Genes (Basel) 2018; 9:genes9070333. [PMID: 30004470 PMCID: PMC6070835 DOI: 10.3390/genes9070333] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Accepted: 06/28/2018] [Indexed: 12/13/2022] Open
Abstract
The microRNA (miR)-17 family is widely expressed in mammalian tissues and play important roles in various physiological and pathological processes. Here, the functions of miR-106a-5p, a member of miR-17 family, were explored during myogenic differentiation in C2C12 cell line. First, miR-106a-5p was found to be relatively lower expressed in two-month skeletal muscle tissues and gradually decreased upon myogenic stimuli. Forced expression of miR-106a-5p significantly reduced the differentiation index, fusion index as well as the expression of myogenic markers (MyoD, MyoG, MyHC, Myomixer, Myomarker). Meanwhile, the levels of phosphorylated AKT were reduced by overexpression of miR-106a-5p, and administration of insulin-like growth factor 1 (IGF1), a booster of myogenic differentiation, could recover all the inhibitory effects above of miR-106a-5p. Furthermore, miR-106a-5p was elevated in aged muscles and dexamethasone (DEX)-treated myotubes, and up-regulation of miR-106a-5p significantly reduced the diameters of myotubes accompanied with increased levels of muscular atrophy genes and decreased PI3K/AKT activities. Finally, miR-106a-5p was demonstrated to directly bind to the 3'-UTR of PIK3R1, thus, repress the PI3K/AKT signaling.
Collapse
|
34
|
The regulation of skeletal muscle fiber-type composition by betaine is associated with NFATc1/MyoD. J Mol Med (Berl) 2018; 96:685-700. [PMID: 29876588 DOI: 10.1007/s00109-018-1657-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Revised: 05/16/2018] [Accepted: 05/17/2018] [Indexed: 01/03/2023]
Abstract
Increasing evidence indicates that muscular dysfunction or alterations in skeletal muscle fiber-type composition not only are involved in muscle metabolism and function but also can limit functional capacity. Therefore, understanding the mechanisms regulating key events during skeletal myogenesis is necessary. Betaine is a naturally occurring component of commonly eaten foods. Here, we showed that 10 mM betaine supplementation in vitro significantly repressed myoblast proliferation and enhanced myoblast differentiation. This effect can be mediated by regulation of miR-29b-3p. Further analysis showed that betaine supplementation in vitro regulated skeletal muscle fiber-type composition through the induction of NFATc1 and the negative regulation of MyoD expression. Furthermore, mice fed with 10 mM betaine in water for 133 days showed no impairment in overall health. Consistently, betaine supplementation increased muscle mass, promoted muscle formation, and modulated the ratio of fiber types in skeletal muscle in vivo. These findings shed light on the diverse biological functions of betaine and indicate that betaine supplementation may lead to new therapies for diseases such as muscular dystrophy or other diseases related to muscle dysfunction. KEY MESSAGES: Betaine supplementation inhibits proliferation and promotes differentiation of C2C12 myoblasts. Betaine supplementation regulates fast to slow muscle fiber-type conversion and is associated with NFATc1/MyoD. Betaine supplementation enhances skeletal myogenesis in vivo. Betaine supplementation does not impair health of mice.
Collapse
|
35
|
Wang H, Zhang Q, Wang B, Wu W, Wei J, Li P, Huang R. miR-22 regulates C2C12 myoblast proliferation and differentiation by targeting TGFBR1. Eur J Cell Biol 2018; 97:257-268. [PMID: 29588073 DOI: 10.1016/j.ejcb.2018.03.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 03/20/2018] [Accepted: 03/20/2018] [Indexed: 12/22/2022] Open
Abstract
Recently, miR-22 was found to be differentially expressed in different skeletal muscle growth period, indicated that it might have function in skeletal muscle myogenesis. In this study, we found that the expression of miR-22 was the most in skeletal muscle and was gradually up-regulated during mouse myoblast cell (C2C12 myoblast cell line) differentiation. Overexpression of miR-22 repressed C2C12 myoblast proliferation and promoted myoblast differentiation into myotubes, whereas inhibition of miR-22 showed the opposite results. During myogenesis, we predicted and verified transforming growth factor beta receptor 1 (TGFBR1), a key receptor of the TGF-β/Smad signaling pathway, was a target gene of miR-22. Then, we found miR-22 could regulate the expression of TGFBR1 and down-regulate the Smad3 signaling pathway. Knockdown of TGFBR1 by siRNA suppressed the proliferation of C2C12 cells but induced its differentiation. Conversely, overexpression of TGFBR1 significantly promoted proliferation but inhibited differentiation of the myoblast. Additionally, when C2C12 cells were treated with different concentrations of transforming growth factor beta 1 (TGF-β1), the level of miR-22 in C2C12 cells was reduced. The TGFBR1 protein level was significantly elevated in C2C12 cells treated with TGF-β1. Moreover, miR-22 was able to inhibit TGF-β1-induced TGFBR1 expression in C2C12 cells. Altogether, we demonstrated that TGF-β1 inhibited miR-22 expression in C2C12 cells and miR-22 regulated C2C12 cell myogenesis by targeting TGFBR1.
Collapse
Affiliation(s)
- Han Wang
- Institute of Swine Science, Nanjing Agricultural University, Nanjing, 210095, China; College of Animal Science and Technology, Zhejiang A&F University, Lin'an, 311300, China
| | - Qian Zhang
- Institute of Swine Science, Nanjing Agricultural University, Nanjing, 210095, China
| | - BinBin Wang
- Institute of Swine Science, Nanjing Agricultural University, Nanjing, 210095, China
| | - WangJun Wu
- Institute of Swine Science, Nanjing Agricultural University, Nanjing, 210095, China
| | - Julong Wei
- Institute of Swine Science, Nanjing Agricultural University, Nanjing, 210095, China
| | - Pinghua Li
- Institute of Swine Science, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Ruihua Huang
- Institute of Swine Science, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
36
|
Gan M, Du J, Shen L, Yang D, Jiang A, Li Q, Jiang Y, Tang G, Li M, Wang J, Li X, Zhang S, Zhu L. miR-152 regulates the proliferation and differentiation of C2C12 myoblasts by targeting E2F3. In Vitro Cell Dev Biol Anim 2018; 54:304-310. [PMID: 29508126 DOI: 10.1007/s11626-017-0219-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 11/27/2017] [Indexed: 01/19/2023]
Abstract
The development of skeletal muscle is a complex process involving the proliferation, differentiation, apoptosis, and changing of muscle fiber types in myoblasts. Many reports have described the involvement of microRNAs in the myogenesis of myoblasts. In this study, we found that the expression of miR-152 was gradually down-regulated during myoblast proliferation, but gradually up-regulated during the differentiation of myoblasts. Transfection with miR-152 mimics restrained cell proliferation and decreased the expression levels of cyclin E, CDK4, and cyclin D1, but promoted myotube formation and significantly increased the mRNA expression levels of MyHC, MyoD, MRF4, and MyoG in C2C12 myoblasts. However, treatment with miR-152 inhibitors promoted cell proliferation and restrained differentiation. Moreover, over-expression of miR-152 significantly decreased E2F3 production in C2C12 myoblasts. A luciferase assay confirmed that miR-152 could bind to the 3' UTR of E2F3. In conclusion, this study showed that miR-152 inhibited proliferation and promoted myoblast differentiation by targeting E2F3.
Collapse
Affiliation(s)
- Mailin Gan
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Jingjing Du
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Linyuan Shen
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Dongli Yang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Anan Jiang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Qiang Li
- Sichuan Province General Station of Animal Husbandry, Chengdu, 611130, China
| | - Yanzhi Jiang
- College of Life and Science, Sichuan Agricultural University, Chengdu, 611130, China
| | - Guoqing Tang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Mingzhou Li
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Jinyong Wang
- Chongqing Academy of Animal Sciences, Chongqing, 402460, China
| | - Xuewei Li
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Shunhua Zhang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China.
| | - Li Zhu
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China.
| |
Collapse
|
37
|
Tang Z, Qiu H, Luo L, Liu N, Zhong J, Kang K, Gou D. miR-34b Modulates Skeletal Muscle Cell Proliferation and Differentiation. J Cell Biochem 2017; 118:4285-4295. [PMID: 28422320 DOI: 10.1002/jcb.26079] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 04/18/2017] [Indexed: 12/22/2022]
Abstract
Myogenesis involves myoblast proliferation and differentiation to myocytes, followed by fusion and hypertrophy to form myotubes during muscle development. Increasing evidence showed that microRNAs (miRNAs) play important roles in the regulation of myogenesis. We have previously revealed that miR-34b is steadily increased during this process. This miRNA regulates differentiation in various cell types, though its function in myogenesis remains to be elucidated. In this study, we show that miR-34b represses muscle cell proliferation and promotes myotube formation. Our quantitative iTRAQ-based proteomic analysis reveals 97 proteins are regulated by miR-34b in mouse myoblast C2C12. We identified that miR-34b targets 14-3-3 protein gamma, adenosylhomocysteinase and nucleolin by binding to their 3'UTR. Further analysis of these proteins expression patterns show that nucleolin is a cognate target of miR-34b during myogenic differentiation. Here, we proved that a moderate reduction of nucleolin in cells enhanced the myotube formation. However, nucleolin is required for myogenesis, as cells with low levels of nucleolin reduced cell proliferation rate and are unable to differentiate. Our data demonstrated that nucleolin regulates myogenesis in a protein-abundance-dependent manner. J. Cell. Biochem. 118: 4285-4295, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Zhixiong Tang
- Shenzhen Key Laboratory of Microbial Genetic Engineering, College of Life Sciences, Shenzhen University, Shenzhen, Guangdong, 518060, China
| | - Huiling Qiu
- Shenzhen Key Laboratory of Microbial Genetic Engineering, College of Life Sciences, Shenzhen University, Shenzhen, Guangdong, 518060, China
- Biomedical Engineering, Health and Environmental Engineering, Shenzhen Technology University, Shenzhen, Guangdong, 518000, China
| | - Lan Luo
- Shenzhen Key Laboratory of Microbial Genetic Engineering, College of Life Sciences, Shenzhen University, Shenzhen, Guangdong, 518060, China
| | - Nian Liu
- Shenzhen Key Laboratory of Microbial Genetic Engineering, College of Life Sciences, Shenzhen University, Shenzhen, Guangdong, 518060, China
| | - Jiasheng Zhong
- Shenzhen Key Laboratory of Microbial Genetic Engineering, College of Life Sciences, Shenzhen University, Shenzhen, Guangdong, 518060, China
| | - Kang Kang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shenzhen University Health Sciences Center, Shenzhen, 518000, China
| | - Deming Gou
- Shenzhen Key Laboratory of Microbial Genetic Engineering, College of Life Sciences, Shenzhen University, Shenzhen, Guangdong, 518060, China
| |
Collapse
|
38
|
Zhang S, Chen X, Huang Z, Chen D, Yu B, He J, Zheng P, Yu J, Luo J, Luo Y, Chen H. Effects of MicroRNA-27a on Myogenin Expression and Akt/FoxO1 Signal Pathway during Porcine Myoblast Differentiation. Anim Biotechnol 2017; 29:183-189. [DOI: 10.1080/10495398.2017.1348357] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Shurun Zhang
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
| | - Xiaoling Chen
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
| | - Zhiqing Huang
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
| | - Daiwen Chen
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
| | - Bing Yu
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
| | - Jun He
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
| | - Ping Zheng
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
| | - Jie Yu
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
| | - Junqiu Luo
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
| | - Yuheng Luo
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
| | - Hong Chen
- College of Food Science, Sichuan Agricultural University, Yaan, Sichuan, P. R. China
| |
Collapse
|
39
|
Tang Z, Liu N, Luo L, Kang K, Li L, Ni R, Qiu H, Gou D. MicroRNA-17-92 Regulates the Transcription Factor E2F3b during Myogenesis In Vitro and In Vivo. Int J Mol Sci 2017; 18:ijms18040727. [PMID: 28362317 PMCID: PMC5412313 DOI: 10.3390/ijms18040727] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Revised: 03/11/2017] [Accepted: 03/24/2017] [Indexed: 01/09/2023] Open
Abstract
Myogenic differentiation, which occurs during muscle development, is a highly ordered process that can be regulated by E2F transcription factors. Available data show that E2F3b, but not E2F3a, is upregulated and required for myogenic differentiation. However, the regulation of E2F3b expression in myogenic differentiation is not well understood. To investigate whether E2Fb expression is controlled by miRNAs, we used bioinformatics to combine the database of microRNAs downregulated during myogenesis and those predicted to target E2F3. This identified miR-17 and miR-20a as miRNAs potentially involved in E2F3 regulation. We found that miR-17-92 controls the expression of E2F3b in C2C12 cells during myogenic differentiation. Moreover, we confirmed that miR-20a regulates the expression of E2F3b proteins in vivo using a muscle regeneration model.
Collapse
Affiliation(s)
- Zhixiong Tang
- Shenzhen Key Laboratory of Microbial Genetic Engineering, College of Life Sciences, Shenzhen University, Shenzhen 518060, China.
| | - Nian Liu
- Shenzhen Key Laboratory of Microbial Genetic Engineering, College of Life Sciences, Shenzhen University, Shenzhen 518060, China.
| | - Lan Luo
- Shenzhen Key Laboratory of Microbial Genetic Engineering, College of Life Sciences, Shenzhen University, Shenzhen 518060, China.
| | - Kang Kang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shenzhen University Health Sciences Center, Shenzhen 518000, China.
| | - Li Li
- Shenzhen Key Laboratory of Microbial Genetic Engineering, College of Life Sciences, Shenzhen University, Shenzhen 518060, China.
| | - Ruiyang Ni
- School of Life Sciences, Peking University, Beijing 100871, China.
| | - Huiling Qiu
- Shenzhen Key Laboratory of Microbial Genetic Engineering, College of Life Sciences, Shenzhen University, Shenzhen 518060, China.
- Biomedical Engineering, College of Health and Environmental Engineering, Shenzhen Technology University, Shenzhen 51000, China.
| | - Deming Gou
- Shenzhen Key Laboratory of Microbial Genetic Engineering, College of Life Sciences, Shenzhen University, Shenzhen 518060, China.
| |
Collapse
|
40
|
Tong HL, Jiang RY, Zhang WW, Yan YQ. MiR-2425-5p targets RAD9A and MYOG to regulate the proliferation and differentiation of bovine skeletal muscle-derived satellite cells. Sci Rep 2017; 7:418. [PMID: 28341832 PMCID: PMC5428422 DOI: 10.1038/s41598-017-00470-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 02/28/2017] [Indexed: 12/18/2022] Open
Abstract
Our group previously identified miR-2425-5p, a unique bovine miRNA; however, its biological function and regulation in muscle-derived satellite cells (MDSCs) remain unclear. Herein, stem-loop RT-PCR results showed that miR-2425-5p increased during MDSCs proliferation, but decreased during differentiation. Cell proliferation was examined using EdU assays, cyclin B1 (CCNB1) and proliferating cell nuclear antigen (PCNA) western blot (WB) and flow cytometry analysis. These results showed that miR-2425-5p mimics (miR-2425-M) enhanced MDSCs proliferation, whereas, miR-2425-5p inhibitor (miR-2425-I) had opposite effect. Conversely, cell differentiation studies by desmin (DES) immunofluorescence, myotubes formation, and myosin heavy chain 3 (MYH3) WB analyses revealed that miR-2425-M and miR-2425-I blocked and promoted MDSCs differentiation, respectively. Moreover, luciferase reporter, RT-PCR, and WB assays showed that miR-2425-5p directly targeted the 3′-UTR of RAD9 homolog A (RAD9A) and myogenin (MYOG) to regulate their expression. Rescue experiment showed RAD9A inhibited the proliferation of MDSCs through miR-2425-5p. In addition, we found that miR-2425-5p expression was regulated by its host gene NCK associated protein 5-like (NCKAP5L) rather than being transcribed independently as a separate small RNA. Collectively, these data indicate that miR-2425-5p is a novel regulator of bovine MDSCs proliferation and differentiation and provides further insight into the biological functions of miRNA in this species.
Collapse
Affiliation(s)
- Hui Li Tong
- The Laboratory of Cell and Developmental Biology, Northeast Agricultural University, Harbin, Heilongjiang, 150030, China
| | - Run Ying Jiang
- The Laboratory of Cell and Developmental Biology, Northeast Agricultural University, Harbin, Heilongjiang, 150030, China
| | - Wei Wei Zhang
- College of Life Sciences and Agriculture & Forestry, Qiqihar University, Qiqihar, Heilongjiang, 161006, China
| | - Yun Qin Yan
- The Laboratory of Cell and Developmental Biology, Northeast Agricultural University, Harbin, Heilongjiang, 150030, China.
| |
Collapse
|
41
|
Gonçalves TJ, Armand AS. Non-coding RNAs in skeletal muscle regeneration. Noncoding RNA Res 2017; 2:56-67. [PMID: 30159421 PMCID: PMC6096429 DOI: 10.1016/j.ncrna.2017.03.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 03/23/2017] [Accepted: 03/23/2017] [Indexed: 01/09/2023] Open
Abstract
Following injury, skeletal muscles can regenerate from muscle specific stem cells, called satellite cells. Quiescent in uninjured muscles, satellite cells become activated, proliferate and differentiate into myotubes. Muscle regeneration occurs following distinct main overlapping phases, including inflammation, regeneration and maturation of the regenerated myofibers. Each step of muscle regeneration is orchestrated through complex signaling networks and gene regulatory networks, leading to the expression of specific set of genes in each concerned cell type. Apart from the well-established transcriptional mechanisms involving the myogenic regulatory factors of the MyoD family, increasing data indicate that each step of muscle regeneration is controlled by a wide range of non-coding RNAs. In this review, we discuss the role of two classes of non-coding RNAs (microRNAs and long non-coding RNAs) in the inflammatory, regeneration and maturation steps of muscle regeneration.
Collapse
Affiliation(s)
- Tristan J.M. Gonçalves
- Institut Necker-Enfants Malades, Inserm, U1151, 14 rue Maria Helena Vieira Da Silva, CS 61431, Paris, F-75014, France
- INSERM UMRS 1124, 45 rue des Saints-Pères, F-75270 Paris cedex 06, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Anne-Sophie Armand
- Institut Necker-Enfants Malades, Inserm, U1151, 14 rue Maria Helena Vieira Da Silva, CS 61431, Paris, F-75014, France
- INSERM UMRS 1124, 45 rue des Saints-Pères, F-75270 Paris cedex 06, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
42
|
Blondelle J, Shapiro P, Domenighetti AA, Lange S. Cullin E3 Ligase Activity Is Required for Myoblast Differentiation. J Mol Biol 2017; 429:1045-1066. [PMID: 28238764 DOI: 10.1016/j.jmb.2017.02.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 02/17/2017] [Accepted: 02/18/2017] [Indexed: 01/06/2023]
Abstract
The role of cullin E3-ubiquitin ligases for muscle homeostasis is best known during muscle atrophy, as the cullin-1 substrate adaptor atrogin-1 is among the most well-characterized muscle atrogins. We investigated whether cullin activity was also crucial during terminal myoblast differentiation and aggregation of acetylcholine receptors for the establishment of neuromuscular junctions in vitro. The activity of cullin E3-ligases is modulated through post-translational modification with the small ubiquitin-like modifier nedd8. Using either the Nae1 inhibitor MLN4924 (Pevonedistat) or siRNA against nedd8 in early or late stages of differentiation on C2C12 myoblasts, and primary satellite cells from mouse and human, we show that cullin E3-ligase activity is necessary for each step of the muscle cell differentiation program in vitro. We further investigate known transcriptional repressors for terminal muscle differentiation, namely ZBTB38, Bhlhe41, and Id1. Due to their identified roles for terminal muscle differentiation, we hypothesize that the accumulation of these potential cullin E3-ligase substrates may be partially responsible for the observed phenotype. MLN4924 is currently undergoing clinical trials in cancer patients, and our experiments highlight concerns on the homeostasis and regenerative capacity of muscles in these patients who often experience cachexia.
Collapse
Affiliation(s)
- Jordan Blondelle
- Division of Cardiology, University of California San Diego, La Jolla, CA-92093 USA
| | - Paige Shapiro
- Division of Cardiology, University of California San Diego, La Jolla, CA-92093 USA
| | - Andrea A Domenighetti
- Rehabilitation Institute of Chicago, Chicago, IL-60611 USA; Department of Physical Medicine and Rehabilitation, Northwestern University, Chicago, IL-60611, USA
| | - Stephan Lange
- Division of Cardiology, University of California San Diego, La Jolla, CA-92093 USA.
| |
Collapse
|
43
|
Yang P, Cai L, Zhang G, Bian Z, Han G. The role of the miR-17-92 cluster in neurogenesis and angiogenesis in the central nervous system of adults. J Neurosci Res 2016; 95:1574-1581. [PMID: 27869313 DOI: 10.1002/jnr.23991] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 10/23/2016] [Accepted: 10/24/2016] [Indexed: 02/03/2023]
Abstract
It is well known that neurogenesis is not the only concern for the fully functional recovery after brain or spinal cord injury, as it has been shed light on the critical role of angiogenesis in improving neurological functional recovery. Angiogenesis and neurogenesis coordinately interact with each other in the developing and adult brain, during which they may respond to similar mediators and receptors, in which they share a common posttranscriptional regulator: the miR-17-92 cluster. The miR-17-92 cluster was initially described as an oncogene and was later demonstrated to drive key physiological and pathological responses during development and diseases respectively. It has been reported that the miR-17-92 cluster regulates both neurogenesis and angiogenesis. The miR-17-92 cluster modulates neural progenitor cells proliferation not only during development but also during neurological disorders such as stroke. It has also been shown that the endothelial miR-17-92 cluster regulates angiogenesis during embryonic stage and adulthood. In this review, we have discussed the actions of the miR-17-92 cluster in neuronal and vascular plasticity, and its potential as a novel therapeutic strategy for CNS injury. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Ping Yang
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing, 400038, PR China
| | - Linghu Cai
- Cadet Brigade, Third Military Medical University, Chongqing, 400038, PR China
| | - Guan Zhang
- Cadet Brigade, Third Military Medical University, Chongqing, 400038, PR China
| | - Zhiqun Bian
- Cadet Brigade, Third Military Medical University, Chongqing, 400038, PR China
| | - Gaofeng Han
- Cadet Brigade, Third Military Medical University, Chongqing, 400038, PR China
| |
Collapse
|
44
|
Zhang Y, Yu B, He J, Chen D. From Nutrient to MicroRNA: a Novel Insight into Cell Signaling Involved in Skeletal Muscle Development and Disease. Int J Biol Sci 2016; 12:1247-1261. [PMID: 27766039 PMCID: PMC5069446 DOI: 10.7150/ijbs.16463] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 08/19/2016] [Indexed: 12/17/2022] Open
Abstract
Skeletal muscle is a remarkably complicated organ comprising many different cell types, and it plays an important role in lifelong metabolic health. Nutrients, as an external regulator, potently regulate skeletal muscle development through various internal regulatory factors, such as mammalian target of rapamycin (mTOR) and microRNAs (miRNAs). As a nutrient sensor, mTOR, integrates nutrient availability to regulate myogenesis and directly or indirectly influences microRNA expression. MiRNAs, a class of small non-coding RNAs mediating gene silencing, are implicated in myogenesis and muscle-related diseases. Meanwhile, growing evidence has emerged supporting the notion that the expression of myogenic miRNAs could be regulated by nutrients in an epigenetic mechanism. Therefore, this review presents a novel insight into the cell signaling network underlying nutrient-mTOR-miRNA pathway regulation of skeletal myogenesis and summarizes the epigenetic modifications in myogenic differentiation, which will provide valuable information for potential therapeutic intervention.
Collapse
Affiliation(s)
- Yong Zhang
- Institute of Animal Nutrition, Sichuan Agricultural University, Ya'an, Sichuan 625014, P. R. China.; Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, China
| | - Bing Yu
- Institute of Animal Nutrition, Sichuan Agricultural University, Ya'an, Sichuan 625014, P. R. China.; Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, China
| | - Jun He
- Institute of Animal Nutrition, Sichuan Agricultural University, Ya'an, Sichuan 625014, P. R. China.; Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, China
| | - Daiwen Chen
- Institute of Animal Nutrition, Sichuan Agricultural University, Ya'an, Sichuan 625014, P. R. China.; Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, China
| |
Collapse
|