1
|
Li F, Liu T, Dong Y, Gao Q, Lu R, Deng Z. 5-Methylcytosine RNA modification and its roles in cancer and cancer chemotherapy resistance. J Transl Med 2025; 23:390. [PMID: 40181461 PMCID: PMC11966802 DOI: 10.1186/s12967-025-06217-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 02/10/2025] [Indexed: 04/05/2025] Open
Abstract
Recent advancements in cancer therapies have improved clinical outcomes, yet therapeutic resistance remains a significant challenge because of its complex mechanisms. Among epigenetic factors, m5C RNA modification is emerging as a key player in cancer drug resistance, similar to the well-known m6A modification. m5C affects RNA metabolism processes, including splicing, export, translation, and stability, thereby influencing drug efficacy. This review highlights the critical roles of m5C in modulating resistance to chemotherapy, targeted therapy, radiotherapy, and immunotherapy. This review also discusses the functions of key regulators, including methyltransferases, demethylases, and m5C-binding proteins, as essential modulators of the m5C epigenetic landscape that contribute to its dynamic and complex regulatory network. Targeting these regulatory components offers a promising strategy to overcome resistance. We highlight the need for further research to elucidate the specific mechanisms by which m5C contributes to resistance and to develop precise m5C-targeted therapies, presenting m5C-focused strategies as potential novel anticancer treatments.
Collapse
Affiliation(s)
- Fang Li
- Science and technology department, Suzhou Key Laboratory of Neuro-Oncology and Nano-Bionics, Affiliated Kunshan Hospital of Jiangsu University, Suzhou, 215300, Jiangsu, China
| | - Tingting Liu
- Science and technology department, Suzhou Key Laboratory of Neuro-Oncology and Nano-Bionics, Affiliated Kunshan Hospital of Jiangsu University, Suzhou, 215300, Jiangsu, China
| | - Yajing Dong
- School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Qianqian Gao
- Science and technology department, Suzhou Key Laboratory of Neuro-Oncology and Nano-Bionics, Affiliated Kunshan Hospital of Jiangsu University, Suzhou, 215300, Jiangsu, China
| | - Rongzhu Lu
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China.
- Center for Experimental Research, Kunshan Hospital Affiliated to Jiangsu University, Kunshan, 215130, Jiangsu, China.
| | - Zhiyong Deng
- Science and technology department, Suzhou Key Laboratory of Neuro-Oncology and Nano-Bionics, Affiliated Kunshan Hospital of Jiangsu University, Suzhou, 215300, Jiangsu, China.
| |
Collapse
|
2
|
Lu J, Liu Y, Li H. Identification of key lncRNAs and mRNAs in muscle development pathways of Tan sheep. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2024; 52:101336. [PMID: 39378789 DOI: 10.1016/j.cbd.2024.101336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 10/10/2024]
Abstract
The study aimed to identify the long noncoding RNA (lncRNA) responsible for regulating muscle development in Tan sheep. RNA-seq analysis was conducted on longissimus dorsi samples from 1-day-old and 60-day-old Tan sheep to investigate the molecular processes involved in muscle development. A total of 5517 lncRNAs and 2885 mRNAs were found to be differentially expressed in the 60-day-old Tan sheep. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis revealed that these differentially expressed lncRNAs and mRNAs were linked to pathways crucial for muscle development, such as MAPK, cAMP, and calcium-mediated signaling pathways. Key genes like CDKN1A, MAPK14, TGFB1, MEF2C, MYOD1, and CD53 were identified as significant players in muscle development. The study validated the RNA-seq results through RT-qPCR, confirming the consistency of expression levels of differentially expressed lncRNAs and mRNAs. These findings indicate that lncRNA-mRNA networks produce a remarked effect on modulating muscle development in Tan sheep, such as lncRNAs (MSTRG.12808.1/MSTRG.22662.3/MSTRG.18310.1) and mRNAs (MSTRG.10027/MSTRG.10029/MSTRG.10258/MSTRG.11011/MSTRG.10354), laying the groundwork for future research in this area.
Collapse
Affiliation(s)
- Jiawei Lu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Yilan Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Huixia Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
3
|
Chen M, Peng J, Zhu G, Qian C, Xiao Z, Song X, Yu H, Huang R, Wang W, Zheng H, Yu Y. Long noncoding RNA MALAT1 as a ceRNA drives mouse fibroblast activation via the miR-335-3p/P2ry2 axis. PLoS One 2024; 19:e0308723. [PMID: 39133718 PMCID: PMC11318857 DOI: 10.1371/journal.pone.0308723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 07/29/2024] [Indexed: 08/15/2024] Open
Abstract
Fibrosis is a complex pathological process that can lead to the permanent loss of biological function, with P2ry2 playing a crucial role in this process. Long non-coding RNAs (lncRNAs) have been reported to play an critically important role in the fibrotic process. However, it remains unclear whether lncRNAs can regulate fibrosis through P2ry2. In this study, we detected the expression of the long non-coding RNA metastasis-associated lung adenocarcinoma transcript 1 (lnc-MALAT1). We investigated the expression patterns of lnc-MALAT1 and P2ry2 in denervated skeletal muscle, a classical model of fibrosis. Additionally, we utilized a TGF-β-mediated fibrosis model in NIH/3T3 cells to examine the effects of lnc-MALAT1 and P2ry2 on fibroblast activation and the underlying regulatory mechanisms in vitro. Our results demonstrated that the expression levels of lnc-MALAT1 and P2ry2 were consistently elevated in denervated skeletal muscle, correlating with the degree of fibrosis. In vitro experiments confirmed the regulatory effect of lnc-MALAT1 on P2ry2. Furthermore, we identified miR-335-3p as a potential key molecule in the regulatory relationship of lnc-MALAT1/P2ry2. Dual luciferase reporter assays and AGO2-RIP verified the molecular sponging effect of lnc-MALAT1 on miR-335-3p. Additionally, we validated the regulation of the lnc-MALAT1/miR-335-3p/P2ry2 axis through experimental approaches. In conclusion, our study identified a crucial role of lnc-MALAT1/miR-335-3p/P2ry2 axis in fibroblast activation, providing a promising treatment option against the fibrosis.
Collapse
Affiliation(s)
- Mengjie Chen
- Department of Otolaryngology Head & Neck Surgery, The First Afflilated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Otolaryngology Head & Neck Surgery, Changhai Hospital of Navy Medical University, Shanghai, Shanghai, China
| | - Jieying Peng
- Department of Otolaryngology Head & Neck Surgery, Changhai Hospital of Navy Medical University, Shanghai, Shanghai, China
| | - Guanghao Zhu
- Department of Otolaryngology Head & Neck Surgery, Changhai Hospital of Navy Medical University, Shanghai, Shanghai, China
| | - Cunhui Qian
- Department of Otolaryngology Head & Neck Surgery, The First Afflilated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Zhi Xiao
- Department of Otolaryngology Head & Neck Surgery, The First Afflilated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xianmin Song
- Department of Otolaryngology Head & Neck Surgery, Changhai Hospital of Navy Medical University, Shanghai, Shanghai, China
| | - Haojun Yu
- Department of Otolaryngology Head & Neck Surgery, Changhai Hospital of Navy Medical University, Shanghai, Shanghai, China
| | - Rushi Huang
- Department of Otolaryngology Head & Neck Surgery, Changhai Hospital of Navy Medical University, Shanghai, Shanghai, China
| | - Wei Wang
- Department of Otolaryngology Head & Neck Surgery, Changhai Hospital of Navy Medical University, Shanghai, Shanghai, China
| | - Hongliang Zheng
- Department of Otolaryngology Head & Neck Surgery, Changhai Hospital of Navy Medical University, Shanghai, Shanghai, China
| | - Yafeng Yu
- Department of Otolaryngology Head & Neck Surgery, The First Afflilated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
4
|
Luo Z, Zhu J, Fang Z, Xu R, Wan R, He Y, Chen Y, Chen S, Wang Q, Liu Q, Chen S. Exercise-augmented THSD7B exhibited a positive prognostic implication and tumor-suppressed functionality in pan-cancer. Front Immunol 2024; 15:1440226. [PMID: 39161765 PMCID: PMC11330788 DOI: 10.3389/fimmu.2024.1440226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 07/22/2024] [Indexed: 08/21/2024] Open
Abstract
BACKGROUND Breast cancer, one of the most prevalent malignancies among women worldwide, has rising incidence rates. Physical activity, particularly exercise, has emerged as a significant modifier of cancer prognosis, influencing both tumor biology and patient outcomes. METHODS In this study, we utilized a murine breast cancer model, dividing mice into a control group and an exercise group; the latter underwent 21 days of voluntary running. We conducted RNA sequencing, bioinformatics analysis, pan-cancer analysis, and cellular experiments to investigate the underlying mechanisms influenced by exercise. RESULTS Exercise led to a significant reduction in tumor size and weight. Post-exercise mRNA sequencing indicated a notable upregulation of THSD7B in the exercised mice, with significant alterations observed in pathways such as MicroRNAs in cancers and the Calcium signaling pathway. In a broader cancer context, THSD7B showed considerable expression variability, being significantly downregulated in several cancers, correlating with positive prognostic outcomes in PRAD, LAML, KIRC, and GBM and highlighting its potential role as a prognostic marker and therapeutic target. THSD7B expression was also negatively associated with processes of breast cancer cell proliferation, migration, and invasion. CONCLUSION This study underscores the dual role of exercise in modulating gene expression relevant to tumor growth and highlights the potential of THSD7B as a therapeutic target in cancer. Future research should further explore the specific mechanisms by which exercise and THSD7B influence cancer progression and develop immunotherapy-enhanced strategies to change patient outcomes in clinical settings.
Collapse
Affiliation(s)
- Zhiwen Luo
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jinguo Zhu
- Department of Orthopaedics, Nantong Tongzhou Hospital of Traditional Chinese Medicine, Tongzhou, Jiangsu, China
| | - Zhengyuan Fang
- The First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, Liaoning, China
| | - Rui Xu
- The First Clinical Medicine College, Nanjing Medical University, Nanjing, China
| | - Renwen Wan
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yanwei He
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yisheng Chen
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Shuo Chen
- Internal Medicine of Chinese Medicine, Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Qing Wang
- Department of Orthopaedics, Kunshan Hospital of Chinese Medicine, Kunshan, Jiangsu, China
| | - Qizhi Liu
- Internal Medicine of Chinese Medicine, Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Shiyi Chen
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
5
|
Shi M, Yang S, Zhao X, Sun D, Li Y, Yang J, Li M, Cai C, Guo X, Li B, Lu C, Cao G. Effect of LncRNA LOC106505926 on myogenesis and Lipogenesis of porcine primary cells. BMC Genomics 2024; 25:530. [PMID: 38816813 PMCID: PMC11137989 DOI: 10.1186/s12864-024-10422-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 05/16/2024] [Indexed: 06/01/2024] Open
Abstract
BACKGROUND Skeletal muscle development and fat deposition have important effects on meat quality. The study of regulating skeletal muscle development and fat deposition is of great significance in improving the quality of carcass and meat. In the present study, whole transcriptome sequencing (including RNA-Seq and miRNA-Seq) was performed on the longissimus dorsi muscle (LDM) of Jinfen White pigs at 1, 90, and 180 days of age. RESULTS The results showed that a total of 245 differentially expressed miRNAs were screened in any two comparisons, which may be involved in the regulation of myogenesis. Among them, compared with 1-day-old group, miR-22-5p was significantly up-regulated in 90-day-old group and 180-day-old group. Functional studies demonstrated that miR-22-5p inhibited the proliferation and differentiation of porcine skeletal muscle satellite cells (PSCs). Pearson correlation coefficient analysis showed that long non-coding RNA (lncRNA) LOC106505926 and CXXC5 gene had strong negative correlations with miR-22-5p. The LOC106505926 and CXXC5 were proven to promote the proliferation and differentiation of PSCs, as opposed to miR-22-5p. In terms of mechanism, LOC106505926 functions as a molecular sponge of miR-22-5p to modulate the expression of CXXC5, thereby inhibits the differentiation of PSCs. In addition, LOC106505926 regulates the differentiation of porcine preadipocytes through direct binding with FASN. CONCLUSIONS Collectively, our results highlight the multifaceted regulatory role of LOC106505926 in controlling skeletal muscle and adipose tissue development in pigs and provide new targets for improving the quality of livestock products by regulating skeletal muscle development and fat deposition.
Collapse
Affiliation(s)
- Mingyue Shi
- College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Shuai Yang
- Shanxi Animal Husbandry Technology Extension Service Center, Taiyuan, 030001, China
| | - Xiaolei Zhao
- College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Di Sun
- College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Yifei Li
- College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Jingxian Yang
- College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Meng Li
- College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Chunbo Cai
- College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Xiaohong Guo
- College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Bugao Li
- College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Chang Lu
- College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China.
| | - Guoqing Cao
- College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China.
| |
Collapse
|
6
|
Huang H, Chen P, Feng X, Qian Y, Peng Z, Zhang T, Wang Q. Translational studies of exosomes in sports medicine - a mini-review. Front Immunol 2024; 14:1339669. [PMID: 38259444 PMCID: PMC10800726 DOI: 10.3389/fimmu.2023.1339669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 12/18/2023] [Indexed: 01/24/2024] Open
Abstract
This review in sports medicine focuses on the critical role of exosomes in managing chronic conditions and enhancing athletic performance. Exosomes, small vesicles produced by various cells, are essential for cellular communication and transporting molecules like proteins and nucleic acids. Originating from the endoplasmic reticulum, they play a vital role in modulating inflammation and tissue repair. Their significance in sports medicine is increasingly recognized, particularly in healing athletic injuries, improving articular cartilage lesions, and osteoarthritic conditions by modulating cellular behavior and aiding tissue regeneration. Investigations also highlight their potential in boosting athletic performance, especially through myocytes-derived exosomes that may enhance adaptability to physical training. Emphasizing a multidisciplinary approach, this review underlines the need to thoroughly understand exosome biology, including their pathways and classifications, to fully exploit their therapeutic potential. It outlines future directions in sports medicine, focusing on personalized treatments, clinical evaluations, and embracing technological advancements. This research represents a frontier in using exosomes to improve athletes' health and performance capabilities.
Collapse
Affiliation(s)
- Haoqiang Huang
- Department of Orthopaedics, Kunshan Hospital of Traditional Chinese Medicine, Kunshan, Jiangsu, China
| | - Peng Chen
- Department of Sports Medicine, Peking University Shenzhen Hospital, Shenzhen, China
| | - Xinting Feng
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yinhua Qian
- Department of Orthopaedics, Kunshan Hospital of Traditional Chinese Medicine, Kunshan, Jiangsu, China
| | - Zhijian Peng
- Department of Orthopaedics, Kunshan Hospital of Traditional Chinese Medicine, Kunshan, Jiangsu, China
| | - Ting Zhang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Qing Wang
- Department of Orthopaedics, Kunshan Hospital of Traditional Chinese Medicine, Kunshan, Jiangsu, China
| |
Collapse
|
7
|
Yang Y, Wu J, Liu W, Zhao Y, Chen H. The Function and Regulation Mechanism of Non-Coding RNAs in Muscle Development. Int J Mol Sci 2023; 24:14534. [PMID: 37833983 PMCID: PMC10572267 DOI: 10.3390/ijms241914534] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 08/25/2023] [Accepted: 08/28/2023] [Indexed: 10/15/2023] Open
Abstract
Animal skeletal muscle growth is regulated by a complex molecular network including some non-coding RNAs (ncRNAs). In this paper, we review the non-coding RNAs related to the growth and development of common animal skeletal muscles, aiming to provide a reference for the in-depth study of the role of ncRNAs in the development of animal skeletal muscles, and to provide new ideas for the improvement of animal production performance.
Collapse
Affiliation(s)
- Yaling Yang
- College of Animal Science, Xinjiang Agricultural University, Urumqi 830052, China; (Y.Y.); (W.L.)
| | - Jian Wu
- Key Laboratory of Beef Cattle Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Academy of Agricultural Sciences of Jilin Province, Changchun 136100, China;
| | - Wujun Liu
- College of Animal Science, Xinjiang Agricultural University, Urumqi 830052, China; (Y.Y.); (W.L.)
| | - Yumin Zhao
- Key Laboratory of Beef Cattle Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Academy of Agricultural Sciences of Jilin Province, Changchun 136100, China;
| | - Hong Chen
- College of Animal Science, Xinjiang Agricultural University, Urumqi 830052, China; (Y.Y.); (W.L.)
| |
Collapse
|
8
|
Li Y, Kong M, Wang J, Han P, Zhang N, Yang X, Wang J, Hu Y, Duo Y, Liu D. Exercise-induced circulating exosomes potentially prevent pelvic organ prolapse in clinical practice via inhibition of smooth muscle apoptosis. Heliyon 2022; 9:e12583. [PMID: 37077375 PMCID: PMC10106923 DOI: 10.1016/j.heliyon.2022.e12583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 11/14/2022] [Accepted: 12/15/2022] [Indexed: 12/26/2022] Open
Abstract
Background This study aimed to explore the potential mechanisms of exercise to prevent pelvic organ prolapse (POP) and search for diagnostic indictors for POP. Methods We used two clinical POP datasets with patients' information (GSE12852 and GSE53868), a dataset consisting of altered microRNA expression in circulating blood after exercise (GSE69717) for bioinformatic analysis and clinical diagnostic analysis, while a series of cellular experiments were conducted for preliminary mechanical validation. Results Our results show that AXUD1 is highly expressed in the smooth muscle of the ovary and is a key pathogenic gene in POP, while miR-133b is a key molecule in the regulation of POP by exercise-induced serum exosomes. The AUCs of AXUD1 for POP diagnosis were 0.842 and 0.840 in GSE12852 and GSE53868 respectively. At cut-off value = 9.627, the sensitivity and specificity of AXUD1 for predicating POP is 1.000 and 0.833 respectively for GSE53868, while at cut-off value = 3324.640, the sensitivity and specificity of AXUD1 for predicating POP is 0.941 and 0.812 separately for GSE12852. Analysis and experiments confirmed that miR-133b can directly regulate AXUD1. miR-133b mediated C2C12 myoblasts proliferation and inhibited hydrogen peroxide-induced apoptosis. Conclusions Our study proved that AXUD1 is a good clinical diagnostic indicator for POP and provided a theoretical basis for future prevention of POP through exercise and a potential target for intervention in muscle dysfunction.
Collapse
|
9
|
Xie C, Gul A, Yu H, Huang X, Deng L, Pan Y, Ni S, Nurahmat M, Abduwaki M, Luo Q, Dong J. Integrated systems pharmacology and transcriptomics to dissect the mechanisms of Loki Zupa decoction in the treatment of murine allergic asthma. JOURNAL OF ETHNOPHARMACOLOGY 2022; 294:115351. [PMID: 35533913 DOI: 10.1016/j.jep.2022.115351] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 04/17/2022] [Accepted: 05/03/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Loki zupa (LKZP) decoction, a traditional Uyghur medicine prescription, has been commonly used to treat numerous respiratory ailments in the Xinjiang region of western China, especially chronic airway inflammatory diseases such as allergic asthma. Due to its complex chemical composition, however, the mechanism of action of LKZP has yet to be fully elucidated. AIM OF THE STUDY Based on the balanced regulation theory of pro-inflammation and anti-inflammation, we tried to investigate the effectiveness of LKZP on asthma and its related protective mechanisms. MATERIALS AND METHODS In this study, an experimental model of asthma was established using ovalbumin (OVA) in BALB/c mice to assess the effects of LKZP. The potential mechanism of LKZP anti allergic asthma were researched by the combination of in silico systems pharmacology and in vivo transcriptomics. RESULTS Our data revealed that LKZP exerted a therapeutic effect against OVA-induced asthma by reducing airway hyperresponsiveness (AHR), peribronchial inflammation, and mucus hypersecretion. Meanwhile, LKZP downregulated the expression of OVA-induced IgE, interleukin (IL)-4, IL-5, IL-13, and tumor necrosis factor (TNF)-α and concurrently promoted the expression of interferon (IFN)-γ in serum and bronchoalveolar lavage fluid (BALF). Systems pharmacology analysis identified 10 core bioactive ingredients and 26 hub targets of LKZP against asthma. Transcriptomic analysis confirmed 246 differentially expressed genes (DEGs) after LKZP treatment. These were mainly expressed in cytokine-cytokine receptor interactions and immune and inflammatory response-related signaling pathways. Additionally, the real-time quantitative PCR (qPCR) results for the nine selected DEGs matched those of the RNA-seq analysis. Nuclear factor (NF)-κB and hypoxia-inducible factor (HIF)-1 signaling pathways were identified as candidate targets involved in the action of LKZP on allergic asthma, which was highly consistent with the findings in silico. By qPCR, Western blot, and immunohistochemical analysis, it was verified that LKZP treatment dramatically inhibited the activation of NF-κB p65 and HIF-1α stimulated by OVA in asthmatic mice. CONCLUSIONS Taken together, our experimental data revealed that LKZP could be a candidate for the treatment of allergic asthma via NF-κB and HIF-1 signaling pathways.
Collapse
Affiliation(s)
- Cong Xie
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China.
| | - Aman Gul
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China.
| | - Hang Yu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China.
| | - Xi Huang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China.
| | - Lingling Deng
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China.
| | - Yue Pan
- Institute of Integrative Medicine, Fudan University, Shanghai, China; School of Pharmacy, Fudan University, Shanghai, China.
| | - Shuangshuang Ni
- Sinopharm Xinjiang Pharmaceutical Co., Ltd., Urumqi, Xinjiang, China.
| | - Mammat Nurahmat
- College of Xinjiang Uyghur Medicine, Hotan, Xinjiang, China.
| | | | - Qingli Luo
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China.
| | - Jingcheng Dong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China.
| |
Collapse
|
10
|
Luo Z, Qi B, Sun Y, Chen Y, Lin J, Qin H, Wang N, Shi R, Shang X, Chen S, Chen J. Engineering Bioactive M2 Macrophage-Polarized, Anti-inflammatory, miRNA-Based Liposomes for Functional Muscle Repair: From Exosomal Mechanisms to Biomaterials. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2201957. [PMID: 35802903 DOI: 10.1002/smll.202201957] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 06/03/2022] [Indexed: 02/05/2023]
Abstract
Severe inflammation and myogenic differentiation disorder are the major obstacles to skeletal muscle healing after injury. MicroRNAs (miRNAs) play an important role as regulatory molecules during the process of muscle healing, but the detailed mechanism of miRNA-mediated intercellular communication between myoblasts and macrophages remains unclear. Here, it is reported that myoblasts secrete miRNAs-enriched exosomes in the inflammatory environment, through which miR-224 is transferred into macrophages to inhibit M2 polarization. Further data demonstrate that WNT-9a may be a direct target of miR-224 for macrophage polarization. In turn, the secretome of M1 macrophages impairs myogenic differentiation and promotes proliferation. Single-cell integration analysis suggests that the elevation of exosome-derived miR-224 is caused by the activation of the key factor E2F1 in myoblasts and demonstrates the RB/E2F1/miR-224/WNT-9a axis. In vivo results show that treatment with antagomir-224 or liposomes containing miR-224 inhibitors suppresses fibrosis and improves muscle recovery. These findings indicate the importance of the crosstalk between myoblasts and macrophages via miRNA-containing exosomes in the regulation of macrophage polarization and myogenic differentiation/proliferation during muscle healing. This study provides a strategy for treating muscle injury through designing an M2 polarization-enabling anti-inflammatory and miRNA-based bioactive material.
Collapse
Affiliation(s)
- Zhiwen Luo
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Beijie Qi
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Yaying Sun
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Yisheng Chen
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Jinrong Lin
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Haocheng Qin
- Department of Rehabilitation, Huashan Hospital, Fudan University, Shanghai, 200433, P. R. China
| | - Ning Wang
- Department of Ophthalmology, Shanghai 9th People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China
| | - Runjie Shi
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai ninth people's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China
| | - Xiliang Shang
- Department of Ophthalmology, Shanghai 9th People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China
| | - Shiyi Chen
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Jiwu Chen
- Department of Sports Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200080, P. R. China
| |
Collapse
|
11
|
Pakravan K, Razmara E, Mahmud Hussen B, Sattarikia F, Sadeghizadeh M, Babashah S. SMAD4 contributes to chondrocyte and osteocyte development. J Cell Mol Med 2022; 26:1-15. [PMID: 34841647 PMCID: PMC8742202 DOI: 10.1111/jcmm.17080] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 10/25/2021] [Accepted: 11/11/2021] [Indexed: 12/12/2022] Open
Abstract
Different cellular and molecular mechanisms contribute to chondrocyte and osteocyte development. Although vital roles of the mothers against decapentaplegic homolog 4 (also called 'SMAD4') have been discussed in different cancers and stem cell-related studies, there are a few reviews summarizing the roles of this protein in the skeletal development and bone homeostasis. In order to fill this gap, we discuss the critical roles of SMAD4 in the skeletal development. To this end, we review the different signalling pathways and also how SMAD4 defines stem cell features. We also elaborate how the epigenetic factors-ie DNA methylation, histone modifications and noncoding RNAs-make a contribution to the chondrocyte and osteocyte development. To better grasp the important roles of SMAD4 in the cartilage and bone development, we also review the genotype-phenotype correlation in animal models. This review helps us to understand the importance of the SMAD4 in the chondrocyte and bone development and the potential applications for therapeutic goals.
Collapse
Affiliation(s)
- Katayoon Pakravan
- Department of Molecular GeneticsFaculty of Biological SciencesTarbiat Modares UniversityTehranIran
| | - Ehsan Razmara
- Department of Medical GeneticsFaculty of Medical SciencesTarbiat Modares UniversityTehranIran
| | - Bashdar Mahmud Hussen
- Department of PharmacognosyCollege of PharmacyHawler Medical UniversityKurdistan RegionIraq
| | - Fatemeh Sattarikia
- Department of Molecular GeneticsFaculty of Biological SciencesTarbiat Modares UniversityTehranIran
| | - Majid Sadeghizadeh
- Department of Molecular GeneticsFaculty of Biological SciencesTarbiat Modares UniversityTehranIran
| | - Sadegh Babashah
- Department of Molecular GeneticsFaculty of Biological SciencesTarbiat Modares UniversityTehranIran
| |
Collapse
|
12
|
CREB1 transcription-activated lncRNA PVT1 promotes cardiac fibrosis via miR-145/HCN1 axis. Int J Cardiol 2022; 353:88-95. [DOI: 10.1016/j.ijcard.2022.01.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 12/30/2021] [Accepted: 01/12/2022] [Indexed: 12/16/2022]
|
13
|
Zhang Z, Fan Y, Deng K, Liang Y, Zhang G, Gao X, El-Samahy MA, Zhang Y, Deng M, Wang F. Circular RNA circUSP13 sponges miR-29c to promote differentiation and inhibit apoptosis of goat myoblasts by targeting IGF1. FASEB J 2021; 36:e22097. [PMID: 34935184 DOI: 10.1096/fj.202101317r] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 11/10/2021] [Accepted: 11/29/2021] [Indexed: 12/24/2022]
Abstract
Circular RNAs (circRNAs) are an indispensable element of post-transcriptional gene regulation, influencing a variety of biological processes including myogenic differentiation; however, little is known about the function of circRNA in goat myogenic differentiation. Using RNA-sequencing data from our laboratory, we explored the influences of circUSP13, as a candidate circRNA, on myoblast differentiation since its expression is higher in myoblasts of lamb (first day of age) than that of the fetus (75th day of pregnancy). In in vitro experiments, circUSP13 significantly promoted differentiation and inhibited apoptosis in goat primary myoblasts. Mechanistically, circUSP13 localized with miR-29c in the cytoplasm of goat myoblasts to regulate IGF1 expression. We further demonstrated that circUSP13 sponges miR-29c, promoting IGF1 expression that upregulated the expression of MyoG and MyHC. Thus, our results identified circUSP13 as a molecular marker for breeding programs of mutton production, as well as the circUSP13-miR-29c-IGF1 axis as a potential therapeutic target for combating muscle wasting.
Collapse
Affiliation(s)
- Zhen Zhang
- Institute of Sheep and Goat Science, Nanjing Agricultural University, Nanjing, China
| | - Yixuan Fan
- Institute of Sheep and Goat Science, Nanjing Agricultural University, Nanjing, China
| | - Kaiping Deng
- Institute of Sheep and Goat Science, Nanjing Agricultural University, Nanjing, China
| | - Yaxu Liang
- Institute of Sheep and Goat Science, Nanjing Agricultural University, Nanjing, China
| | - Guomin Zhang
- Institute of Sheep and Goat Science, Nanjing Agricultural University, Nanjing, China
| | - Xiaoxiao Gao
- Institute of Sheep and Goat Science, Nanjing Agricultural University, Nanjing, China
| | - M A El-Samahy
- Institute of Sheep and Goat Science, Nanjing Agricultural University, Nanjing, China
| | - Yanli Zhang
- Institute of Sheep and Goat Science, Nanjing Agricultural University, Nanjing, China
| | - Mingtian Deng
- Institute of Sheep and Goat Science, Nanjing Agricultural University, Nanjing, China
| | - Feng Wang
- Institute of Sheep and Goat Science, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
14
|
Luo ZW, Sun YY, Lin JR, Qi BJ, Chen JW. Exosomes derived from inflammatory myoblasts promote M1 polarization and break the balance of myoblast proliferation/differentiation. World J Stem Cells 2021; 13:1762-1782. [PMID: 34909122 PMCID: PMC8641021 DOI: 10.4252/wjsc.v13.i11.1762] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 06/28/2021] [Accepted: 09/02/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Acute muscle injuries are one of the most common injuries in sports. Severely injured muscles are prone to re-injury due to fibrotic scar formation caused by prolonged inflammation. How to regulate inflammation and suppress fibrosis is the focus of promoting muscle healing. Recent studies have found that myoblasts and macrophages play important roles in the inflammatory phase following muscle injury; however, the crosstalk between these two types of cells in the inflammatory environment, particularly the exosome-related mechanisms, had not been well studied. AIM To evaluate the effects of exosomes from inflammatory C2C12 myoblasts (IF-C2C12-Exos) on macrophage polarization and myoblast proliferation/differentiation. METHODS A model of inflammation was established in vitro by lipopolysaccharide stimulation of myoblasts. C2C12-Exos were isolated and purified from the supernatant of myoblasts by gradient centrifugation. Multiple methods were used to identify the exosomes. Gradient concentrations of IF-C2C12-Exos were added to normal macrophages and myoblasts. PKH67 fluorescence tracing was used to identify the interaction between exosomes and cells. Microscopic morphology, Giemsa stain, and immunofluorescence were carried out for histological analysis. Additionally, ELISA assays, flow cytometry, and western blot were conducted to analyze molecular changes. Moreover, myogenic proliferation was assessed by the BrdU test, scratch assay, and CCK-8 assay. RESULTS We found that the PKH-67-marked C2C12-Exos can be endocytosed by both macrophages and myoblasts. IF-C2C12-Exos induced M1 macrophage polarization and suppressed the M2 phenotype in vitro. In addition, these exosomes also stimulated the inflammatory reactions of macrophages. Furthermore, we demonstrated that IF-C2C12-Exos disrupted the balance of myoblast proliferation/differentiation, leading to enhanced proliferation and suppressed fibrogenic/myogenic differentiation. CONCLUSION IF-C2C12-Exos can induce M1 polarization, resulting in a sustained and aggravated inflammatory environment that impairs myoblast differentiation, and leads to enhanced myogenic proliferation. These results demonstrate why prolonged inflammation occurs after acute muscle injury and provide a new target for the regulation of muscle regeneration.
Collapse
Affiliation(s)
- Zhi-Wen Luo
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Ya-Ying Sun
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Jin-Rong Lin
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Bei-Jie Qi
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Ji-Wu Chen
- Department of Sports Medicine, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
15
|
Lin J, Yang X, Liu S, Luo Z, Chen Q, Sun Y, Ding Z, Chen J. Long non-coding RNA MFAT1 promotes skeletal muscle fibrosis by modulating the miR-135a-5p-Tgfbr2/Smad4 axis as a ceRNA. J Cell Mol Med 2021; 25:4420-4433. [PMID: 33837645 PMCID: PMC8093971 DOI: 10.1111/jcmm.16508] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 03/16/2021] [Accepted: 03/18/2021] [Indexed: 02/05/2023] Open
Abstract
Fibrosis after skeletal muscle injury is common in sports and can cause irreversible damage to the biomechanical properties of skeletal muscle. Long non-coding RNAs (lncRNAs) have been validated to act as important modulators in the fibrosis of various organs. Here, we reported a novel lncRNA (the skeletal muscle fibrosis-associated transcript 1, lnc-MFAT1), which was highly expressed in skeletal muscle fibrosis. We demonstrate that lnc-MFAT1 knockdown can reduce TGFβ-induced fibrosis in vitro and attenuate skeletal muscle fibrosis after acute contusion in mice. Further study showed that lnc-MFAT1 acted as a competitive endogenous RNA of miR-135a-5p. Besides, the miR-135a-5p inhibition obviously promoted TGFβ-induced fibrosis in vitro via enhancing its target genes Tgfbr2/Smad4. Moreover, we discovered that lnc-MFAT1 regulates Tgfbr2/Smad4 expression by sponging miR-135a-5p to exert competing endogenous RNA function, resulting in TGFβ pathway activation. In conclusion, our study identified a crucial role of lnc-MFAT1-miR-135a-Tgfbr2/Smad4 axis in skeletal muscle fibrosis, providing a promising treatment option against skeletal muscle fibrosis.
Collapse
Affiliation(s)
- Jinrong Lin
- Department of Sports MedicineHuashan HospitalFudan UniversityShanghaiChina
| | - Xiaobao Yang
- Department of Medical Laboratory ScienceRuijin HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Shaohua Liu
- Department of Sports MedicineHuashan HospitalFudan UniversityShanghaiChina
| | - Zhiwen Luo
- Department of Sports MedicineHuashan HospitalFudan UniversityShanghaiChina
| | | | - Yaying Sun
- Department of Sports MedicineHuashan HospitalFudan UniversityShanghaiChina
| | - Zheci Ding
- Department of Sports MedicineHuashan HospitalFudan UniversityShanghaiChina
| | - Jiwu Chen
- Department of Sports MedicineHuashan HospitalFudan UniversityShanghaiChina
| |
Collapse
|