1
|
Zhang X, Shao W, Gao Y, Wang X. Macrophage polarization-mediated PKM2/mTORC1/YME1L signaling pathway activation in fibrosis associated with Cardiorenal syndrome. Cell Signal 2025; 131:111664. [PMID: 39961408 DOI: 10.1016/j.cellsig.2025.111664] [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: 10/23/2024] [Revised: 12/16/2024] [Accepted: 02/14/2025] [Indexed: 04/04/2025]
Abstract
BACKGROUND Cardiorenal syndrome (CRS) is a complex condition characterized by the interplay between cardiac and renal dysfunction, often culminating in renal fibrosis. The role of macrophage polarization and its downstream effects in CRS-induced renal fibrosis remains an area of active investigation. METHODS Single-cell RNA sequencing (scRNA-seq) and immune infiltration analyses were employed to identify key immune cells and genes involved in renal fibrosis in CRS. Meta-analysis and pseudo-time analysis were conducted to validate the functional relevance of these genes. Functional studies utilizing CRISPR/Cas9 gene editing and lentiviral vectors assessed macrophage polarization and epithelial-to-mesenchymal transition (EMT). In vivo, a CRS mouse model was established, and fibrosis progression was tracked using histological and imaging methods. RESULTS The PKM2/mTORC1/YME1L signaling axis was identified as a critical pathway driving renal fibrosis, mediated by HIF-1α-induced M1 macrophage polarization. Inhibition of HIF-1α significantly alleviated renal fibrosis by restricting M1 polarization and suppressing the PKM2/mTORC1/YME1L axis. Co-culture models further demonstrated the involvement of EMT and metabolic reprogramming in affected cells. CONCLUSION Targeting the HIF-1α signaling pathway offers a promising therapeutic strategy for renal fibrosis by modulating macrophage polarization and the PKM2/mTORC1/YME1L axis.
Collapse
Affiliation(s)
- Xuefeng Zhang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan 030032, China.
| | - Wen Shao
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Yun Gao
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Xiaojun Wang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan 030032, China
| |
Collapse
|
2
|
Bao S, Bu H, Dong S, Wang Y, Liu R, Wang H, Ma W, Li Z, Shen N, Lin T, Chen J, Wan Q. Identification of NMT1/MA/VPS15 signal pathway as potential therapeutic target in rat cerebral ischemia injury. Exp Neurol 2025; 389:115252. [PMID: 40221008 DOI: 10.1016/j.expneurol.2025.115252] [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: 02/26/2025] [Revised: 04/06/2025] [Accepted: 04/08/2025] [Indexed: 04/14/2025]
Abstract
Fatty acids play a critical role in cerebral ischemia injury through the regulation of lipid metabolism and inflammatory signaling. Myristic acid (MA), a 14‑carbon saturated fatty acid, serves as a substrate of N-myristoyltransferase 1 (NMT1) and modulates protein function and subcellular localization via myristoylation. We show here that intraperitoneal injection of MA has no effect on the infarct volume after rat cerebral ischemia-reperfusion (I/R) injury. However, our results reveal that the level of MA within the penumbra of ischemic brain is increased and that ischemia-induced downregulation of NMT1 is responsible for the increase of MA. We further show that upregulation of MA by knockdown of NMT1 exacerbates cerebral ischemia injury, while downregulation of MA by BCtDCS (bilateral and cathodal transcranial direct current stimulation) protects against cerebral ischemia injury. Furthermore, we demonstrate that MA reduces the expression of VPS15 (phosphoinositide 3-kinase regulatory subunit 4) to exacerbate cerebral ischemia injury, and that NMT1 acts on MA to regulate VPS15 expression in the ischemic cerebral cortex. Together, this study provides the first evidence identifying NMT1/MA/VPS15 signal pathway as potential target for stroke therapy and suggests that BCtDCS may act on MA-dependent signal pathway to confer neuroprotection.
Collapse
Affiliation(s)
- Shuguang Bao
- Institute of Neuroregeneration & Neurorehabilitation, School of Basic Medicine, Qingdao University, 308 Ningxia Street, Qingdao, China
| | - He Bu
- Institute of Neuroregeneration & Neurorehabilitation, School of Basic Medicine, Qingdao University, 308 Ningxia Street, Qingdao, China
| | - Shan Dong
- Faculty of Life and Health Sciences, Shenzhen University of Advanced Technology, Shenzhen, China
| | - Yajie Wang
- Faculty of Life and Health Sciences, Shenzhen University of Advanced Technology, Shenzhen, China
| | - Rui Liu
- Institute of Neuroregeneration & Neurorehabilitation, School of Basic Medicine, Qingdao University, 308 Ningxia Street, Qingdao, China
| | - Hui Wang
- Institute of Neuroregeneration & Neurorehabilitation, School of Basic Medicine, Qingdao University, 308 Ningxia Street, Qingdao, China
| | - Wenlong Ma
- Institute of Neuroregeneration & Neurorehabilitation, School of Basic Medicine, Qingdao University, 308 Ningxia Street, Qingdao, China
| | - Zhuo Li
- Institute of Neuroregeneration & Neurorehabilitation, School of Basic Medicine, Qingdao University, 308 Ningxia Street, Qingdao, China
| | - Na Shen
- Institute of Neuroregeneration & Neurorehabilitation, School of Basic Medicine, Qingdao University, 308 Ningxia Street, Qingdao, China
| | - Tao Lin
- Institute of Neuroregeneration & Neurorehabilitation, School of Basic Medicine, Qingdao University, 308 Ningxia Street, Qingdao, China
| | - Juan Chen
- Department of Neurology, the Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science & Technology, 26 Shengli Street, Wuhan, China
| | - Qi Wan
- Faculty of Life and Health Sciences, Shenzhen University of Advanced Technology, Shenzhen, China.
| |
Collapse
|
3
|
Jafari N, Zolfi Gol A, Shahabi Rabori V, Saberiyan M. Exploring the role of exosomal and non-exosomal non-coding RNAs in Kawasaki disease: Implications for diagnosis and therapeutic strategies against coronary artery aneurysms. Biochem Biophys Rep 2025; 42:101970. [PMID: 40124995 PMCID: PMC11930191 DOI: 10.1016/j.bbrep.2025.101970] [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: 12/04/2024] [Revised: 02/19/2025] [Accepted: 03/03/2025] [Indexed: 03/25/2025] Open
Abstract
Kawasaki disease (KD) is an acute vasculitis primarily affecting children, with a potential risk of developing coronary artery aneurysms (CAAs) and cardiovascular complications. The emergence of non-coding RNAs (ncRNAs), including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), has provided insights into Kawasaki disease pathogenesis and opened new avenues for diagnosis and therapeutic intervention. Furthermore, polymorphism analysis of ncRNA genes offers significant insights into genetic predisposition to Kawasaki disease, facilitating tailored treatment approaches and risk assessment to improve patient outcomes. Exosomal ncRNAs, which are ncRNAs encapsulated within extracellular vesicles, have garnered significant attention as potential biomarkers for Kawasaki disease and CAA due to their stability and accessibility in biological fluids. This review comprehensively discusses the biogenesis, components, and potential of exosomal and non-exosomal ncRNAs in Kawasaki disease diagnosis and prognosis prediction. It also highlights the roles of non-exosomal ncRNAs, such as miRNAs, lncRNAs, and circRNAs, in Kawasaki disease pathogenesis and their implications as therapeutic targets. Additionally, the review explores the current diagnostic and therapeutic approaches for Kawasaki disease and emphasizes the need for further research to validate these ncRNA-based biomarkers in diverse populations and clinical settings.
Collapse
Affiliation(s)
- Negar Jafari
- Department of Cardiology, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Ali Zolfi Gol
- Department of Cardiology, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Venus Shahabi Rabori
- Department of Cardiology, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Mohammadreza Saberiyan
- Department of Medical Genetics, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| |
Collapse
|
4
|
Wang L, Zhang Y, Wang J, Jiang X, Wang G, Wang H, Shu Y, Huiyuan H. Exosomal lncRNA profiles in patients with HFrEF: Evidence for KLF3-AS1 as a novel diagnostic biomarker. Mol Cell Probes 2025; 82:102032. [PMID: 40404068 DOI: 10.1016/j.mcp.2025.102032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 02/25/2025] [Accepted: 05/17/2025] [Indexed: 05/24/2025]
Abstract
BACKGROUND Serum exosomal long noncoding RNAs (lncRNAs) have not been studied extensively as biomarkers in heart failure (HF) with reduced ejection fraction (HFrEF). We compared lncRNA expression in patients with HFrEF hospitalized for acute HF with that in healthy individuals to identify differentially expressed exosomal lncRNAs. Furthermore, we explored the clinical value of exosomal KLF3-AS1 in diagnosing HF and investigated its role in cardiac hypertrophy. METHOD Exosomes were isolated from patients with HFrEF and healthy individuals. We performed microarray analysis of differentially expressed lncRNAs and genes (DELs and DEGs, respectively) associated with HF. Protein-protein interaction (PPI), lncRNA-mRNA-KEGG pathway, and interaction networks between lncRNAs and RNA-binding proteins (RBPs) were developed. Expression patterns were verified using qRT-PCR. The diagnostic applicability of exosomal lncRNAs in HF was quantified by plotting receiver operating characteristic (ROC) curves. The size of the cardiomyocytes was evaluated using α-actinin immunostaining. RESULTS In total, 138 DELs and 1132 DEGs were identified. PPI network analysis identified INS, CTNNB1, and CAT as the most prominent hub genes, whereas MDM2, MYH6, ENAH, and KLF3-AS1 were significantly enriched in the RBP interaction network. In the validation phase, patients with HFrEF exhibited a significant increase in KLF3-AS1 expression compared with healthy individuals. Exosomal KLF3-AS1 had an area under the ROC curve of 0.861. Functionally, KLF3-AS1 overexpression reduced Ang II-induced cardiac hypertrophy in vitro. CONCLUSION Our results elucidated the exact patterns of circulating exosomal mRNAs and lncRNA expression in patients with HFrEF hospitalized for acute HF. Moreover, the high expression of exosomal KLF3-AS1 is a potential diagnostic biomarker for HFrEF.
Collapse
Affiliation(s)
- Lei Wang
- Department of Cardiology, Shanxi Cardiovascular Hospital, Taiyuan, 030001, Shanxi, China
| | - Yanan Zhang
- Department of Critical Medicine, Shanxi Bethune Hospital, Taiyuan, 030001, Shanxi, China
| | - Jiapu Wang
- Department of Cardiology, Shanxi Cardiovascular Hospital, Taiyuan, 030001, Shanxi, China
| | - Xiao Jiang
- Department of Cardiology, Shanxi Cardiovascular Hospital, Taiyuan, 030001, Shanxi, China
| | - Gang Wang
- Department of Cardiology, Shanxi Cardiovascular Hospital, Taiyuan, 030001, Shanxi, China
| | - Haixiong Wang
- Department of Cardiology, Shanxi Cardiovascular Hospital, Taiyuan, 030001, Shanxi, China
| | - Yan Shu
- Department of Cardiology, Shanxi Cardiovascular Hospital, Taiyuan, 030001, Shanxi, China.
| | - Han Huiyuan
- Department of Cardiology, Shanxi Cardiovascular Hospital, Taiyuan, 030001, Shanxi, China.
| |
Collapse
|
5
|
Gao M, Yin L, Zhang B, Dong Z, Jiang W, Bai Z, Zhao X, Xu L, Wang N, Peng J. Targeting Ischemic Myocardium: Nanoparticles Loaded with Long Noncoding RNA AK156373 siRNA Alleviate Myocardial Infarction. ACS NANO 2025; 19:18475-18491. [PMID: 40338223 DOI: 10.1021/acsnano.5c01641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2025]
Abstract
Despite advancements in the development of targeted approaches for the treatment of myocardial infarction (MI), there is a continuing need for improvements in treatment approaches due to the high mortality and prevalence of MI. The identification of specific therapeutic targets and the development of efficient delivery systems are essential. In this study, a nanoparticle delivery system targeting necrotic cardiomyocytes was engineered. This system effectively downregulated long noncoding RNA (lncRNA) AK156373 and reduced oxidative stress and inflammation during MI progression. Mechanistically, silencing lncRNA AK156373 enhanced the viability and mitochondrial function of hypoxic cardiomyocytes and lowered intracellular inflammatory cytokine levels and reactive oxygen species (ROS) production. In vivo, cardiac-specific lncRNA AK15673 knockout mice were generated (AK156373flox/flox, Myh6-Cre mice), and lncRNA AK156373 knockout obviously reduced the infarct size, collagen fiber deposition, and ischemia severity in MI mice, leading to improved cardiac function. Additionally, lncRNA AK156373 modulated miR-204-5p to regulate C-X-C motif chemokine receptor 2 (CXCR2) protein expression via the competing endogenous RNA (ceRNA) mechanism, exacerbating myocardial damage and accelerating MI progression. Subsequently, nanoparticles loaded with lncRNA AK156373 siRNA were synthesized. The nanoparticles significantly inhibited MI progression by modulating the miR-204-5p/CXCR2 axis to reduce oxidative stress and inflammation. Overall, these findings establish a key regulatory role for lncRNA AK156373 in MI progression and present a direct preclinical approach for MI therapy.
Collapse
Affiliation(s)
- Meng Gao
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Lianhong Yin
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Bo Zhang
- Department of Cardiology, The First Affiliated Hospital, Dalian Medical University, Dalian 116011, China
| | - Zhichao Dong
- Department of Cardiology, The First Affiliated Hospital, Dalian Medical University, Dalian 116011, China
| | - Wenjiao Jiang
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Zhuoya Bai
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Xuerong Zhao
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Lina Xu
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Ning Wang
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Jinyong Peng
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
- Hubei Shizhen Laboratory, Wuhan 430065, China
| |
Collapse
|
6
|
Zhao H, Zhao D, Li S, Liu Y, Zhao R, Zhu X, Xiong P, Mo Y, Gu H, Liu J. PRAP1 regulates colorectal cancer cell proliferation and ferroptosis through the Nrf2 signaling pathway. Cell Signal 2025:111863. [PMID: 40373840 DOI: 10.1016/j.cellsig.2025.111863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 04/22/2025] [Accepted: 05/11/2025] [Indexed: 05/17/2025]
Abstract
BACKGROUND Colorectal cancer (CRC) is a common type of cancer that impacts the digestive tract, and current treatment options have limitations. Studies have confirmed that ferroptosis plays a key role in CRC progression. This research sought to clarify how Proline-rich acidic protein 1 (PRAP1) influences CRC advancement and ferroptosis, and to uncover the underlying mechanisms involved. METHODS Real-time quantitative PCR (RT-qPCR) and western blot were employed to ascertain the levels of PRAP1 in CRC cells (SW480, SW620, and LOVO) and tissues. Immunofluorescence was utilized to locate PRAP1. Biological characterization of CRC cells was determined through CCK-8 assay, EdU staining, Transwell assay, TUNEL staining and Scratch-wound assay. Iron and Fe2+ content was measured using prussian blue staining and iron assay kit. A nude mouse model of xenograft was established, and the impact of PRAP1 on tumor growth was investigated by pathological staining. Expression of ferroptosis-related proteins as well as nuclear factor-erythroid factor 2-related factor 2 (Nrf2) pathway proteins was detected by Western blot. RESULTS PRAP1 levels were elevated in CRC. Overexpression PRAP1 promoted cell proliferation, inhibited apoptosis and ferroptosis. Additionally, overexpression PRAP1 can activate the Nrf2 pathway. However, silencing PRAP1 had the opposite effect. In vivo tumor xenograft experiments showed that silencing PRAP1 resulted in decreased Ki67 positivity and increased TUNEL positivity in tumor tissues, and blocked Nrf2 pathway, thereby inhibited tumor growth. CONCLUSION PRAP1 promotes CRC cell proliferation and inhibits ferroptosis by Nrf2 pathway. This study provides a conceptual framework for the development of novel targeted drugs.
Collapse
Affiliation(s)
- Hongchao Zhao
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province 450052, China
| | - Deyao Zhao
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province 450052, China
| | - Siting Li
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province 450052, China
| | - Yang Liu
- Endocrinology Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province 450000, China
| | - Ruiwen Zhao
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province 450052, China
| | - Xiaorong Zhu
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province 450052, China
| | - Pingping Xiong
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, Henan Province 471003, China
| | - Yingyi Mo
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province 450052, China
| | - Hao Gu
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province 450052, China.
| | - Junqi Liu
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province 450052, China.
| |
Collapse
|
7
|
Li L, Zheng Z, Lan W, Tang N, Zhang D, Ling J, Wu Y, Yang P, Fu L, Liu J, Zhang J, Yu P, Huang T. Role of Exosomes in Cardiovascular Disease: A Key Regulator of Intercellular Communication in Cardiomyocytes. ACS OMEGA 2025; 10:18145-18169. [PMID: 40385188 PMCID: PMC12079207 DOI: 10.1021/acsomega.4c11423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 03/27/2025] [Accepted: 04/22/2025] [Indexed: 05/20/2025]
Abstract
In the cardiovascular system, different types of cardiovascular cells can secrete specific exosomes and participate in the maintenance of cardiovascular function and the occurrence and development of diseases. Exosomes carry biologically active substances such as proteins and nucleic acids from cells of origin and can be used as biomarkers for disease diagnosis and prognosis assessment. In addition, exosome-mediated intercellular communication plays a key role in the occurrence and development of cardiovascular diseases and has become a potential therapeutic target. This article emphasizes the importance of understanding the mechanism of exosomes in cardiovascular diseases and systematically details the current understanding of exosomes as regulators of intercellular communication in cardiomyocytes, providing a basis for future research and therapeutic intervention.
Collapse
Affiliation(s)
- Liuxin Li
- Department of Endocrinology and Metabolism, second Affiliated Hospital
of Nanchang University, Nanchang, People’s Republic of China, The second Clinical Medical College, Nanchang University, Nanchang 330006, Republic of China
| | - Zhidong Zheng
- Department of Endocrinology and Metabolism, second Affiliated Hospital
of Nanchang University, Nanchang, People’s Republic of China, The second Clinical Medical College, Nanchang University, Nanchang 330006, Republic of China
| | - Wenyu Lan
- The
Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China
| | - Nan Tang
- The
Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China
| | - Deju Zhang
- Food
and Nutritional Sciences, School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong 0000, Hong Kong
| | - Jitao Ling
- Department
of Endocrinology and Metabolism, The Second Affiliated Hospital, Jiangxi
Medical College, Nanchang University, Nanchang 330006, Jiangxi,China
| | - Yuting Wu
- Department
of Endocrinology and Metabolism, The Second Affiliated Hospital, Jiangxi
Medical College, Nanchang University, Nanchang 330006, Jiangxi,China
| | - Pingping Yang
- Department
of Endocrinology and Metabolism, The Second Affiliated Hospital, Jiangxi
Medical College, Nanchang University, Nanchang 330006, Jiangxi,China
| | - Linhua Fu
- Department
of Cardiovascular Medicine, The Second Affiliated Hospital, Jiangxi
Medical College, Nanchang University, Nanchang 330006, Jiangxi,China
| | - Jianping Liu
- Department
of Endocrinology and Metabolism, The Second Affiliated Hospital, Jiangxi
Medical College, Nanchang University, Nanchang 330006, Jiangxi,China
| | - Jing Zhang
- Department
of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical
College, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Peng Yu
- Department
of Metabolism and Endocrinology, The Second
Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China
| | - Tieqiu Huang
- Department
of Cardiovascular Medicine, The Second Affiliated Hospital, Jiangxi
Medical College, Nanchang University, Nanchang 330006, Jiangxi,China
| |
Collapse
|
8
|
Zou T, Tang X, Wang H, Shang X, Liang X, Ma X. Nanocrystalline cellulose-geniposide complex enhances gut-brain axis modulation for depression treatment. Commun Biol 2025; 8:667. [PMID: 40287572 PMCID: PMC12033350 DOI: 10.1038/s42003-025-07934-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: 05/30/2024] [Accepted: 03/13/2025] [Indexed: 04/29/2025] Open
Abstract
Depression, a major global health issue, is closely associated with imbalances in gut microbiota and altered intestinal functions. This study investigates the antidepressant potential of a composite of Geniposide (GP) and Nanocrystalline Cellulose (NCC), focusing on its effects on the gut-brain axis. Utilizing network pharmacology, GP was identified as a key compound targeting the BCL2 gene in depression management. Experimental approaches, including a chronic unpredictable mild stress (CUMS) model in mice, cellular assays, and fecal microbiota transplantation (FMT), were used to evaluate the composite's effectiveness. Results indicate that GP activates the adenosine monophosphate-activated protein kinase (AMPK) pathway by upregulating BCL2, enhancing intestinal barrier integrity, and balancing gut flora. These mechanisms contribute to its positive effects on hippocampal function and depressive-like behaviors in mice, suggesting that the GP-NCC composite could be a promising avenue for developing depression therapies that target gut health.
Collapse
Affiliation(s)
- Tianyu Zou
- Department of Encephalopathy, Shenzhen Luohu District Hospital of Traditional Chinese Medicine (Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine), Shenzhen, 518000, Guangdong, PR China.
| | - Xiang Tang
- Department of Encephalopathy, Shenzhen Luohu District Hospital of Traditional Chinese Medicine (Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine), Shenzhen, 518000, Guangdong, PR China
| | - Haiping Wang
- Department of Encephalopathy, Shenzhen Luohu District Hospital of Traditional Chinese Medicine (Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine), Shenzhen, 518000, Guangdong, PR China
| | - Xiaolong Shang
- Department of Encephalopathy, Shenzhen Luohu District Hospital of Traditional Chinese Medicine (Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine), Shenzhen, 518000, Guangdong, PR China
| | - Xiaoyu Liang
- Department of Encephalopathy, Shenzhen Luohu District Hospital of Traditional Chinese Medicine (Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine), Shenzhen, 518000, Guangdong, PR China
| | - Xuemiao Ma
- Department of Encephalopathy, Shenzhen Luohu District Hospital of Traditional Chinese Medicine (Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine), Shenzhen, 518000, Guangdong, PR China
| |
Collapse
|
9
|
Jiang J, Liu F, Cui D, Xu C, Chi J, Yan T, Guo F. Novel molecular mechanisms of immune evasion in hepatocellular carcinoma: NSUN2-mediated increase of SOAT2 RNA methylation. Cancer Commun (Lond) 2025. [PMID: 40227950 DOI: 10.1002/cac2.70023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 03/13/2025] [Accepted: 03/17/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a deadly malignancy known for its ability to evade immune surveillance. NOP2/Sun RNA methyltransferase family member 2 (NSUN2), an RNA methyltransferase involved in carcinogenesis, has been associated with immune evasion and energy metabolism reprogramming. This study aimed to examine the molecular mechanisms underlying the involvement of NSUN2 in immune evasion and metabolic reprogramming of HCC. METHODS Single-cell transcriptomic sequencing was applied to examine cellular composition changes, particularly immune cell dynamics, in HCC and adjacent normal tissues. Bulk RNA-seq and proteomics identified key genes and proteins. Methylation sequencing and methylated RNA immunoprecipitation (MeRIP) were carried out to characterize the role of NSUN2 in 5-methylcytosine (m5C) modification of sterol O-acyltransferase 2 (SOAT2). Clinical samples from 30 HCC patients were analyzed using reverse transcription-quantitative polymerase chain reaction and Western blotting. Gene expression was manipulated using CRISPR/Cas9 and lentiviral vectors. In vitro co-culture models and metabolomics were used to study HCC cell-T cell interactions, energy metabolism, and immune evasion. Tumor growth in an orthotopic mouse model was monitored by bioluminescence imaging, with subsequent measurements of tumor weight, volume, and immunohistochemical staining. RESULTS Single-cell transcriptomic analysis identified a marked increase in malignant cells in HCC tissues. Cell communication analysis indicated that tumor cells might promote cancer progression by evading immune clearance. Multi-omics analyses identified NSUN2 as a key regulator in HCC development. MeRIP confirmed that NSUN2 facilitated the m5C modification of SOAT2. Analysis of human HCC tissue samples demonstrated pronounced upregulation of NSUN2 and SOAT2, along with elevated m5C levels in HCC tissues. In vitro experiments uncovered that NSUN2 augmented the reprogramming of energy metabolism and repressed the activity and cytotoxicity of CD8+ T cells, contributing to immune evasion. In vivo studies further substantiated the role of NSUN2 in fostering immune evasion and tumor formation of HCC by modulating the m5C modification of SOAT2. CONCLUSIONS The findings highlight the critical role of NSUN2 in driving HCC progression through the regulation of m5C modification on SOAT2. These findings present potential molecular markers for HCC diagnosis and therapeutic targets for its treatment.
Collapse
Affiliation(s)
- Jinhua Jiang
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, P. R. China
- Key Laboratory of Systems Biomedicine, Ministry of Education, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, P. R. China
| | - Feng Liu
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, P. R. China
| | - Dan Cui
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, P. R. China
| | - Caixia Xu
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, P. R. China
| | - Jiachang Chi
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, P. R. China
| | - Tinghua Yan
- The First Clinical Medical College of Jinan University, Guangzhou, Guangdong, P. R. China
| | - Fang Guo
- Key Laboratory of Systems Biomedicine, Ministry of Education, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, P. R. China
| |
Collapse
|
10
|
Lu L, Wang L, Yang M, Wang H. Role of METTL16 in PPARγ methylation and osteogenic differentiation. Cell Death Dis 2025; 16:271. [PMID: 40210616 PMCID: PMC11986173 DOI: 10.1038/s41419-025-07527-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 02/11/2025] [Accepted: 03/12/2025] [Indexed: 04/12/2025]
Abstract
Osteoporosis, a prevalent bone disease, is characterized by the deterioration of bone tissue microstructure and imbalanced osteogenesis. The regulatory role of PPARγ m6A methylation mediated by METTL16 remains poorly elucidated. This study utilized advanced single-cell RNA sequencing (scRNA-seq) and Bulk RNA-seq techniques to explore how METTL16 influences the osteogenic differentiation of Bone Marrow-Derived Mesenchymal Stem Cells (BMSCs) and its implication in osteoporosis. The research revealed that METTL16 enhances the suppression of osteogenic differentiation in BMSCs, while PPARγ is associated with BMSC ferroptosis. Mechanistically, METTL16 facilitates the m6A modification of PPARγ transcription, thereby promoting ferroptosis in BMSCs and impeding their osteogenic differentiation. The in vivo animal experiments confirmed the pivotal role of the METTL16-PPARγ axis in osteoporosis development in mice. These findings suggest that the regulation of PPARγ m6A methylation by METTL16, leading to ferroptosis, is a critical mechanism impacting BMSC osteogenic differentiation and the pathogenesis of osteoporosis.
Collapse
Affiliation(s)
- Liangjie Lu
- Department of Orthopedics, Ningbo Medical Center Li Huili Hospital, Li Huili Hospital Affiliated to Ningbo University, Ningbo, China.
| | - Lijun Wang
- Department of Pediatrics, The First Hospital of Jilin University, Changchun, China
| | - Minjie Yang
- Department of Orthopaedics, Jiu jiang NO.1 People's Hospital, Jiu jiang, China
| | - Huihan Wang
- Department of Orthopaedics, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| |
Collapse
|
11
|
Wu H, Lan Q, He YX, Xue JY, Liu H, Zou Y, Liu P, Luo G, Chen MT, Liu MN. Programmed cardiomyocyte death in myocardial infarction. Apoptosis 2025; 30:597-615. [PMID: 39833636 DOI: 10.1007/s10495-025-02075-3] [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] [Accepted: 01/02/2025] [Indexed: 01/22/2025]
Abstract
Cardiovascular disease (CVD) is a leading cause of human mortality worldwide, with patients often at high risk of heart failure (HF) in myocardial infarction (MI), a common form of CVD that results in cardiomyocyte death and myocardial necrosis due to inadequate myocardial perfusion. As terminally differentiated cells, cardiomyocytes possess a severely limited capacity for regeneration, and an excess of dead cardiomyocytes will further stress surviving cells, potentially exacerbating to more extensive heart disease. The article focuses on the relationship between programmed cell death (PCD) of cardiomyocytes, including different forms of apoptosis, necrosis, and autophagy, and MI, as well as the potential application of these mechanisms in the treatment of MI. By gaining a deeper understanding of the mechanisms of cardiomyocyte death, it aims to provide new insights into the prevention and treatment of MI.
Collapse
Affiliation(s)
- Hao Wu
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Qi Lan
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Yi-Xiang He
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Jin-Yi Xue
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Hao Liu
- Department of Pediatrics, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Yuan Zou
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Ping Liu
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Gang Luo
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China.
| | - Ming-Tai Chen
- Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, 518033, Shenzhen, People's Republic of China.
| | - Meng-Nan Liu
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China.
| |
Collapse
|
12
|
Xie X, Huang M, Ma S, Xin Q, Wang Y, Hu L, Zhao H, Li P, Liu M, Yuan R, Miao Y, Zhu Y, Cong W. The role of long non-coding RNAs in cardiovascular diseases: A comprehensive review. Noncoding RNA Res 2025; 11:158-187. [PMID: 39896344 PMCID: PMC11783329 DOI: 10.1016/j.ncrna.2024.12.009] [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: 09/23/2024] [Revised: 12/25/2024] [Accepted: 12/26/2024] [Indexed: 02/04/2025] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of morbidity and mortality worldwide, posing significant challenges to healthcare systems. Despite advances in medical interventions, the molecular mechanisms underlying CVDs are not yet fully understood. For decades, protein-coding genes have been the focus of CVD research. However, recent advances in genomics have highlighted the importance of long non-coding RNAs (lncRNAs) in cardiovascular health and disease. Changes in lncRNA expression specific to tissues may result from various internal or external factors, leading to tissue damage, organ dysfunction, and disease. In this review, we provide a comprehensive discussion of the regulatory mechanisms underlying lncRNAs and their roles in the pathogenesis and progression of CVDs, such as coronary heart disease, atherosclerosis, heart failure, arrhythmias, cardiomyopathies, and diabetic cardiomyopathy, to explore their potential as therapeutic targets and diagnostic biomarkers.
Collapse
Affiliation(s)
- Xuena Xie
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau SAR, 999078, China
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Meiwen Huang
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau SAR, 999078, China
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Shudong Ma
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
- Faculty of Chinese Medicine, Macau University of Science and Technology, 999078, China
| | - Qiqi Xin
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Yuying Wang
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Lantian Hu
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Han Zhao
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Pengqi Li
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Mei Liu
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Rong Yuan
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Yu Miao
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Yizhun Zhu
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau SAR, 999078, China
| | - Weihong Cong
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau SAR, 999078, China
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| |
Collapse
|
13
|
Zhong Z, Li X, Gao L, Wu X, Ye Y, Zhang X, Zeng Q, Zhou C, Lu X, Wei Y, Ding Y, Chen S, Zhou G, Xu J, Liu S. Long Non-coding RNA Involved in the Pathophysiology of Atrial Fibrillation. Cardiovasc Drugs Ther 2025; 39:435-458. [PMID: 37702834 PMCID: PMC11954709 DOI: 10.1007/s10557-023-07491-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/12/2023] [Indexed: 09/14/2023]
Abstract
BACKGROUND Atrial fibrillation (AF) is a prevalent and chronic cardiovascular disorder associated with various pathophysiological alterations, including atrial electrical and structural remodeling, disrupted calcium handling, autonomic nervous system dysfunction, aberrant energy metabolism, and immune dysregulation. Emerging evidence suggests that long non-coding RNAs (lncRNAs) play a significant role in the pathogenesis of AF. OBJECTIVE This discussion aims to elucidate the involvement of AF-related lncRNAs, with a specific focus on their role as miRNA sponges that modulate crucial signaling pathways, contributing to the progression of AF. We also address current limitations in AF-related lncRNA research and explore potential future directions in this field. Additionally, we summarize feasible strategies and promising delivery systems for targeting lncRNAs in AF therapy. CONCLUSION In conclusion, targeting AF-related lncRNAs holds substantial promise for future investigations and represents a potential therapeutic avenue for managing AF.
Collapse
Affiliation(s)
- Zikan Zhong
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xintao Li
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Longzhe Gao
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoyu Wu
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yutong Ye
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoyu Zhang
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qingye Zeng
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Changzuan Zhou
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaofeng Lu
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yong Wei
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Ding
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Songwen Chen
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Genqing Zhou
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Juan Xu
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Shaowen Liu
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
14
|
Du J, Meng X, Yang M, Chen G, Li J, Zhu Z, Wu X, Hu W, Tian M, Li T, Ren S, Zhao P. NGR-Modified CAF-Derived exos Targeting Tumor Vasculature to Induce Ferroptosis and Overcome Chemoresistance in Osteosarcoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410918. [PMID: 39889249 PMCID: PMC11948032 DOI: 10.1002/advs.202410918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 12/13/2024] [Indexed: 02/02/2025]
Abstract
Osteosarcoma (OS) chemoresistance presents a significant clinical challenge. This study aims to investigate the potential of using tumor vascular-targeting peptide NGR-modified cancer-associated fibroblasts (CAFs)-derived exosomes (exos) to deliver circ_0004872-encoded small peptides promoting autophagy-dependent ferroptosis to reverse chemoresistance in OS. Through combined single-cell transcriptome analysis and high-throughput sequencing, it identified circ_0004872 associated with chemoresistance. Subsequent experiments demonstrated that the small peptide encoded by this Circular RNA (circRNA) can effectively reverse chemoresistance by enhancing OS cell sensitivity to chemotherapy via the mechanism of promoting autophagy-dependent ferroptosis. Moreover, in vitro and in vivo results confirmed the efficient delivery of NGR-modified CAFs-derived exo-packaged circ_0004872-109aa to tumor cells, thereby improving targeted therapy efficacy. This study not only offers a novel strategy to overcome chemoresistance in OS but also highlights the potential application value of utilizing exos for drug delivery.
Collapse
Affiliation(s)
- Jianxin Du
- Center of Translational MedicineZibo Central Hospital Affiliated to Binzhou Medical UniversityZibo255036China
| | - Xiangwei Meng
- Center of Translational MedicineZibo Central Hospital Affiliated to Binzhou Medical UniversityZibo255036China
| | - Minghao Yang
- Department of RadiologyYantai Affiliated Hospital of Binzhou Medical UniversityYantai264100China
| | - Guancheng Chen
- State Key Laboratory of Reproductive Medicine and Offspring HealthNanjing Medical UniversityNanjing211166China
| | - Jigang Li
- Department of OrthopedicsZibo Central Hospital Affiliated to Binzhou Medical UniversityZibo255036China
| | - Zengjun Zhu
- School of Medical LaboratoryShandong Second Medical UniversityWeifang261042China
| | - Xuanxuan Wu
- School of Medical LaboratoryShandong Second Medical UniversityWeifang261042China
| | - Wei Hu
- Center of Translational MedicineZibo Central Hospital Affiliated to Binzhou Medical UniversityZibo255036China
| | - Maojin Tian
- Center of Translational MedicineZibo Central Hospital Affiliated to Binzhou Medical UniversityZibo255036China
| | - Tao Li
- Department of OrthopedicsNanjing Jiangbei HospitalNanjing210044China
| | - Shuai Ren
- Center of Translational MedicineZibo Central Hospital Affiliated to Binzhou Medical UniversityZibo255036China
| | - Peiqing Zhao
- Center of Translational MedicineZibo Central Hospital Affiliated to Binzhou Medical UniversityZibo255036China
| |
Collapse
|
15
|
Zhang Y, Yang L, Mu H, Li N, Wang X, Lei H, Pang M. CVB3 regulates Treg cell pyroptosis through the lncRNA XIST/miR-195-5p/caspase-1 molecular axis. Immunobiology 2025; 230:152882. [PMID: 39987748 DOI: 10.1016/j.imbio.2025.152882] [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: 12/10/2024] [Revised: 01/22/2025] [Accepted: 02/12/2025] [Indexed: 02/25/2025]
Abstract
Viral myocarditis (VMC) is characterized by severe cardiac inflammation and is a major cause of congestive heart failure and sudden cardiac death in healthy young people. The lncRNA XIST plays an important regulatory role in myocardial injury, but its role in VMC caused by coxsackievirus B3 (CVB3) infection is unclear. In this study, we evaluated the effects of the lncRNA XIST on a CVB3-induced VMC mouse model and on pyroptosis in CVB3-exposed Treg cells. The results showed that in CVB3-infected VMC and Treg cells, the expression level of the lncRNA XIST was increased, whereas miR-195-5p expression was decreased. In CVB3-induced VMC mice, inflammation was elevated, whereas the Treg/Th17 ratio was reduced. Knocking down the lncRNA XIST suppressed pyroptosis in Treg cells caused by CVB3 infection and inhibited VMC progression in vivo. Studies on downstream mechanisms have shown that the lncRNA XIST targets miR-195-5p, induces caspase-1 expression through the inhibition of miR-195-5p, promotes the expression of the inflammatory factors IL-1β and IL-18 associated with pyroptosis, inhibits the secretion of the anti-inflammatory factors IL-10 and TGF-β1, and ultimately promotes pyroptosis in Treg cells. In conclusion, knocking down the lncRNA XIST inhibits CVB3-induced pyroptosis of Treg cells and VMC progression in mice induced by CVB3 infection. These findings provide a potential theoretical basis for the treatment of VMC.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Magnetic Resonance Imaging, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, Yunnan, China
| | - Lei Yang
- Department of Cardiology, The First People's Hospital of Yunnan Province. The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, Yunnan, China
| | - Huiting Mu
- Department of Cardiology, The First People's Hospital of Yunnan Province. The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, Yunnan, China
| | - Na Li
- Department of Cardiology, The First People's Hospital of Yunnan Province. The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, Yunnan, China
| | - Xuejia Wang
- Department of Cardiology, The First People's Hospital of Yunnan Province. The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, Yunnan, China
| | - Hualan Lei
- Department of Cardiology, The First People's Hospital of Yunnan Province. The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, Yunnan, China
| | - Mingjie Pang
- Department of Cardiology, The First People's Hospital of Yunnan Province. The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, Yunnan, China..
| |
Collapse
|
16
|
Yang N, Zhou X, Gong Y, Deng Z. The role of MUC16 in tumor biology and tumor immunology in ovarian cancer. BMC Cancer 2025; 25:294. [PMID: 39972413 PMCID: PMC11837316 DOI: 10.1186/s12885-025-13461-0] [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: 06/24/2024] [Accepted: 01/06/2025] [Indexed: 02/21/2025] Open
Abstract
In this study, the influence of glycoproteomic changes, specifically MUC16, on NK cell-mediated immunotherapy response in ovarian cancer is explored. Analysis of glycoprotein data from the CPTAC database identified significant upregulation of MUC16 in ovarian cancer tissues, associated with tumor invasiveness and immune evasion. Experimental findings showed that MUC16 knockdown increased NK cell cytotoxicity, decreased invasiveness, and boosted NK cell activation, while MUC16 overexpression resulted in the opposite effects. In vivo experiments demonstrated that MUC16 knockdown suppressed tumor growth, enhanced NK cell infiltration, and bolstered NK cell activation, underscoring the potential of MUC16 as a target for novel immunotherapy approaches in ovarian cancer treatment.
Collapse
Affiliation(s)
- Na Yang
- The First Affiliated Hospital, Gynecology & Obstetrics and Reproductive Medical Center, Hengyang Medical School, University of South China, No. 69, Chuanshan Avenue, Shigu District, Hengyang, 421001, Hunan Province, China
| | - Xi Zhou
- The First Affiliated Hospital, Gynecology & Obstetrics and Reproductive Medical Center, Hengyang Medical School, University of South China, No. 69, Chuanshan Avenue, Shigu District, Hengyang, 421001, Hunan Province, China
| | - Yangmei Gong
- The First Affiliated Hospital, Gynecology & Obstetrics and Reproductive Medical Center, Hengyang Medical School, University of South China, No. 69, Chuanshan Avenue, Shigu District, Hengyang, 421001, Hunan Province, China
| | - Zhizhi Deng
- The First Affiliated Hospital, Gynecology & Obstetrics and Reproductive Medical Center, Hengyang Medical School, University of South China, No. 69, Chuanshan Avenue, Shigu District, Hengyang, 421001, Hunan Province, China.
| |
Collapse
|
17
|
Zhao L, Gu M, Sun Z, Shi L, Yang Z, Zheng M, Wang Y, Sun L, Liu G, Miao F, Tang N. The role of exosomal lncRNAs in cardiovascular disease: Emerging insights based on molecular mechanisms and therapeutic target level. Noncoding RNA Res 2025; 10:198-205. [PMID: 40248838 PMCID: PMC12004008 DOI: 10.1016/j.ncrna.2024.10.001] [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: 06/17/2024] [Revised: 08/28/2024] [Accepted: 10/09/2024] [Indexed: 04/19/2025] Open
Abstract
Cardiovascular diseases (CVD) are widely recognized as a leading cause of death worldwide; however, early diagnosis and disease progression monitoring play a crucial role in their clinical management. The common diagnostic and prognostic biomarkers have represented a revolutionary tool for studying CVD; however, their applications are limited to invasive irreversible heart diseases, such as drug-induced myocardial injury. In light of this information, a growing number of studies are currently investigating the diagnostic and prognostic potential of novel CVD biomarkers. Examples of this are long non-coding RNA (lncRNA) and other RNAs that are specifically expressed at the early stages of heart disease. These RNAs have been reported to be involved in the development of CVD via activating or inhibiting inflammatory mediators and angiogenesis-related factors, as well as endothelial cell proliferation, migration and phenotypic transformation. This review collectively summarizes the recent studies' results concerning exosomal lncRNA biogenesis, characterization, and function, as well as its role as a novel biomarker in a variety of CVD.
Collapse
Affiliation(s)
- Liyuan Zhao
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, 230000, China
- InnoStar Bio-tech Nantong Co., Ltd, Nantong, Jiangsu, 226133, China
- Yangtze Delta Drug Advanced Research Institute, Yangtze Delta Pharmaceutical College, Nantong, Jiangsu, 226133, China
| | - Mengyun Gu
- China State Institute of Pharmaceutical Industry, Shanghai Innostar Bio-Technology Co., Ltd, Shanghai, 201203, China
| | - Zhimin Sun
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, 230000, China
- InnoStar Bio-tech Nantong Co., Ltd, Nantong, Jiangsu, 226133, China
- Yangtze Delta Drug Advanced Research Institute, Yangtze Delta Pharmaceutical College, Nantong, Jiangsu, 226133, China
| | - Lei Shi
- China State Institute of Pharmaceutical Industry, Shanghai Innostar Bio-Technology Co., Ltd, Shanghai, 201203, China
| | - Zixuan Yang
- China State Institute of Pharmaceutical Industry, Shanghai Innostar Bio-Technology Co., Ltd, Shanghai, 201203, China
| | - Minhui Zheng
- China State Institute of Pharmaceutical Industry, Shanghai Innostar Bio-Technology Co., Ltd, Shanghai, 201203, China
| | - Yan Wang
- China State Institute of Pharmaceutical Industry, Shanghai Innostar Bio-Technology Co., Ltd, Shanghai, 201203, China
| | - Luyao Sun
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, 230000, China
- InnoStar Bio-tech Nantong Co., Ltd, Nantong, Jiangsu, 226133, China
- Yangtze Delta Drug Advanced Research Institute, Yangtze Delta Pharmaceutical College, Nantong, Jiangsu, 226133, China
| | - Gang Liu
- InnoStar Bio-tech Nantong Co., Ltd, Nantong, Jiangsu, 226133, China
| | - Feng Miao
- InnoStar Bio-tech Nantong Co., Ltd, Nantong, Jiangsu, 226133, China
| | - Naping Tang
- China State Institute of Pharmaceutical Industry, Shanghai Innostar Bio-Technology Co., Ltd, Shanghai, 201203, China
- Yangtze Delta Drug Advanced Research Institute, Yangtze Delta Pharmaceutical College, Nantong, Jiangsu, 226133, China
| |
Collapse
|
18
|
Hu Y, Zhang W, Ali SR, Takeda K, Vahl TP, Zhu D, Hong Y, Cheng K. Extracellular vesicle therapeutics for cardiac repair. J Mol Cell Cardiol 2025; 199:12-32. [PMID: 39603560 PMCID: PMC11788051 DOI: 10.1016/j.yjmcc.2024.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 10/30/2024] [Accepted: 11/21/2024] [Indexed: 11/29/2024]
Abstract
Extracellular vesicles (EVs) are cell-secreted heterogeneous vesicles that play crucial roles in intercellular communication and disease pathogenesis. Due to their non-tumorigenicity, low immunogenicity, and therapeutic potential, EVs are increasingly used in cardiac repair as cell-free therapy. There exist multiple steps for the design of EV therapies, and each step offers many choices to tune EV properties. Factors such as EV source, cargo, loading methods, routes of administration, surface modification, and biomaterials are comprehensively considered to achieve specific goals. PubMed and Google Scholar were searched in this review, 89 articles related to EV-based cardiac therapy over the past five years (2019 Jan - 2023 Dec) were included, and their key steps in designing EV therapies were counted and analyzed. We aim to provide a comprehensive overview that can serve as a reference guide for researchers to design EV-based cardiac therapies.
Collapse
Affiliation(s)
- Yilan Hu
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Weihang Zhang
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Shah Rukh Ali
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Koji Takeda
- Division of Cardiac Surgery, Department of Surgery, Columbia University, New York, NY 10032, USA
| | - Torsten Peter Vahl
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Donghui Zhu
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Stony Brook University, Stony Brook, NY 11794, USA
| | - Yi Hong
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76019, USA
| | - Ke Cheng
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
19
|
Min F, Dong Z, Zhong S, Li Z, Wu H, Zhang S, Zhang L, Zeng T. Impact of LITAF on Mitophagy and Neuronal Damage in Epilepsy via MCL-1 Ubiquitination. CNS Neurosci Ther 2025; 31:e70191. [PMID: 39764629 PMCID: PMC11705406 DOI: 10.1111/cns.70191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 11/14/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
OBJECTIVE This study aims to investigate how the E3 ubiquitin ligase LITAF influences mitochondrial autophagy by modulating MCL-1 ubiquitination, and its role in the development of epilepsy. METHODS Employing single-cell RNA sequencing (scRNA-seq) to analyze brain tissue from epilepsy patients, along with high-throughput transcriptomics, we identified changes in gene expression. This was complemented by in vivo and in vitro experiments, including protein-protein interaction (PPI) network analysis, western blotting, and behavioral assessments in mouse models. RESULTS Neuronal cells in epilepsy patients exhibited significant gene expression alterations, with increased activity in apoptosis-related pathways and decreased activity in neurotransmitter-related pathways. LITAF was identified as a key upregulated factor, inhibiting mitochondrial autophagy by promoting MCL-1 ubiquitination, leading to increased neuronal damage. Knockdown experiments in mouse models further confirmed that LITAF facilitates MCL-1 ubiquitination, aggravating neuronal injury. CONCLUSION Our findings demonstrate that LITAF regulates MCL-1 ubiquitination, significantly impacting mitochondrial autophagy and contributing to neuronal damage in epilepsy. Targeting LITAF and its downstream mechanisms may offer a promising therapeutic strategy for managing epilepsy.
Collapse
Affiliation(s)
- Fuli Min
- Department of Neurology, School of Medicine, Guangzhou First People's HospitalSouth China University of TechnologyGuangzhouChina
| | - Zhaofei Dong
- Department of Neurology, the Eighth Affiliated HospitalSun Yat‐Sen UniversityShenzhenChina
| | - Shuisheng Zhong
- Department of NeurologyGuangdong Sanjiu Brain HospitalGuangzhouChina
| | - Ze Li
- Department of Neurology, School of Medicine, Guangzhou First People's HospitalSouth China University of TechnologyGuangzhouChina
| | - Hong Wu
- Department of Neurology, School of Medicine, Guangzhou First People's HospitalSouth China University of TechnologyGuangzhouChina
| | - Sai Zhang
- Department of Neurology, School of Medicine, Guangzhou First People's HospitalSouth China University of TechnologyGuangzhouChina
| | - Linming Zhang
- Department of NeurologyThe First Affliated Hospital of Kunming Medical UniversityKunmingChina
| | - Tao Zeng
- Department of Neurology, School of Medicine, Guangzhou First People's HospitalSouth China University of TechnologyGuangzhouChina
- Department of Neurology, Zhujiang HospitalSouthern Medical UniversityGuangzhouChina
| |
Collapse
|
20
|
Wang N, Ma F, Song H, He N, Zhang H, Li J, Liu Q, Xu C. Mesenchymal Stem Cell-Derived Extracellular Vesicles for Regenerative Applications and Radiotherapy. Cell Transplant 2025; 34:9636897241311019. [PMID: 39780320 PMCID: PMC11713979 DOI: 10.1177/09636897241311019] [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: 09/26/2024] [Revised: 12/12/2024] [Accepted: 12/15/2024] [Indexed: 01/11/2025] Open
Abstract
Tissue repair is an extremely crucial part of clinical treatment. During the course of disease treatment, surgery, chemotherapy, and radiotherapy cause tissue damage. On the other hand, Normal tissue from accidental or therapeutic exposure to high-dose radiation can cause severe tissue damage. There is an urgent need for developing medical countermeasures against radiation injury for tissue repair. Tissue repair involves the regeneration, proliferation, differentiation, and migration of tissue cells; imbalance of local tissue homeostasis, progressive chronic inflammation; decreased cell activity and stem cell function; and wound healing. Although many clinical treatments are currently available for tissue repair, they are expensive. The long recovery time and some unavoidable complications such as cell damage and the inflammatory reaction caused by radiotherapy have led to unsatisfactory results. Extracellular vesicles (EVs) derived from mesenchymal stem cells (MSCs) have similar tissue repair functions as MSCs. In tissue damage, EVs can be used as an alternative to stem cell therapy, thereby avoiding related complications such as immunological rejection. EVs play a major role in regulating tissue damage, anti-inflammation, pro-proliferation, and immune response, thus providing a diversified and efficient solution for the repair of disease- and radiotherapy-induced tissue damage. This article reviews the research progress of mesenchymal stem cell-derived EVs in promoting the repair of tissue including heart, lung, liver, intestine, skin, blood system, central nervous system, and tissue damage caused by radiotherapy, thereby aiming to offer new directions and ideas for the radiotherapy and regenerative applications.
Collapse
Affiliation(s)
- Ning Wang
- School of Basic Medicine Sciences, Shandong Second Medical University, Weifang, China
| | - Feifei Ma
- School of Basic Medicine Sciences, Shandong Second Medical University, Weifang, China
| | - Huijuan Song
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, State Key Laboratory of Advanced Medical Materials and Devices, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
| | - Ningning He
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, State Key Laboratory of Advanced Medical Materials and Devices, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
| | - Huanteng Zhang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, State Key Laboratory of Advanced Medical Materials and Devices, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
| | - Jianguo Li
- School of Basic Medicine Sciences, Shandong Second Medical University, Weifang, China
| | - Qiang Liu
- School of Basic Medicine Sciences, Shandong Second Medical University, Weifang, China
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, State Key Laboratory of Advanced Medical Materials and Devices, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
| | - Chang Xu
- School of Basic Medicine Sciences, Shandong Second Medical University, Weifang, China
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, State Key Laboratory of Advanced Medical Materials and Devices, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
| |
Collapse
|
21
|
Zou P, Tao Z, Yang Z, Xiong T, Deng Z, Chen Q. The contribution of the novel CLTC-VMP1 fusion gene to autophagy regulation and energy metabolism in cisplatin-resistant osteosarcoma. Am J Physiol Cell Physiol 2025; 328:C148-C167. [PMID: 39466176 DOI: 10.1152/ajpcell.00302.2024] [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: 05/06/2024] [Revised: 08/26/2024] [Accepted: 09/09/2024] [Indexed: 10/29/2024]
Abstract
Osteosarcoma (OS) is a highly malignant tumor, and chemotherapy resistance is a major challenge in the treatment of this disease. This study aims to explore the role of the CLTC-VMP1 gene fusion in the mechanism of chemotherapy resistance in OS and investigate its molecular mechanisms in mediating energy metabolism reprogramming by regulating autophagy and apoptosis balance. Using single-cell transcriptome analysis, the heterogeneity of OS cells and their correlation with resistance to platinum drugs were revealed. Cisplatin-resistant cell lines were established in human OS cell lines for subsequent experiments. Based on transcriptomic analysis, the importance of VMP1 in chemotherapy resistance was confirmed. Lentiviral vectors overexpressing or interfering with VMP1 were used, and it was observed that inhibiting VMP1 could reverse cisplatin resistance, promote cell apoptosis, and inhibit autophagy, and mitochondrial respiration and glycolysis. Furthermore, the presence of CLTC-VMP1 gene fusion was validated, and its ability to regulate autophagy and apoptosis balance, promote mitochondrial respiration, and glycolysis was demonstrated. Mouse model experiments further confirmed the promoting effect of CLTC-VMP1 on tumor growth and chemotherapy resistance. In summary, the CLTC-VMP1 gene fusion mediates energy metabolism reprogramming by regulating autophagy and apoptosis balance, which promotes chemotherapy resistance in OS.NEW & NOTEWORTHY This study identifies the CLTC-VMP1 gene fusion as a key driver of chemotherapy resistance in osteosarcoma by regulating autophagy and reprogramming cellular energy metabolism. Through single-cell transcriptomics, the research reveals the heterogeneity of tumor cells and the role of VMP1 in promoting resistance to cisplatin. The findings suggest that targeting the CLTC-VMP1 fusion gene may offer new therapeutic strategies to overcome chemotherapy resistance in osteosarcoma.
Collapse
Affiliation(s)
- Pingan Zou
- Bone and Soft Tissue Sarcoma Department, Jiangxi Cancer Hospital, Nanchang, People's Republic of China
| | - Zhiwei Tao
- Bone and Soft Tissue Sarcoma Department, Jiangxi Cancer Hospital, Nanchang, People's Republic of China
| | - Zhengxu Yang
- Bone and Soft Tissue Sarcoma Department, Jiangxi Cancer Hospital, Nanchang, People's Republic of China
| | - Tao Xiong
- Bone and Soft Tissue Sarcoma Department, Jiangxi Cancer Hospital, Nanchang, People's Republic of China
| | - Zhi Deng
- Bone and Soft Tissue Sarcoma Department, Jiangxi Cancer Hospital, Nanchang, People's Republic of China
| | - Qincan Chen
- Bone and Soft Tissue Sarcoma Department, Jiangxi Cancer Hospital, Nanchang, People's Republic of China
| |
Collapse
|
22
|
Shang N, Zhu L, Li Y, Song C, Liu X. Targeting CDK1 and copper homeostasis in breast cancer via a nanopolymer drug delivery system. Cell Biol Toxicol 2024; 41:16. [PMID: 39724454 PMCID: PMC11671568 DOI: 10.1007/s10565-024-09958-2] [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: 06/05/2024] [Accepted: 11/25/2024] [Indexed: 12/28/2024]
Abstract
The prevalence of breast cancer (BRCA) is notable in the female population, being a commonly diagnosed malignancy, where the management of copper levels is crucial for treatment success. This research aims to explore the influence of copper homeostasis on BRCA therapy, with a specific focus on the role of Cyclin-Dependent Kinase 1 (CDK1) and its relationship to copper regulation. A novel thermosensitive hydrogel incorporating nanoparticles (NPs) was engineered to synergize with the chemotherapy drug vincristine (VCR) in inhibiting tumor growth and metastasis. Through a comprehensive approach involving bioinformatics analyses, in vitro experiments, and in vivo models, the study identified CDK1 as a significant factor in BRCA progression under copper homeostasis. MBVP-Gel, a novel thermosensitive hydrogel incorporating NPs, was developed to enhance the delivery of chemotherapy drugs and regulate copper homeostasis in breast cancer treatment. The MBVP-Gel, formulated with copper chelation and VCR NPs, effectively suppressed CDK1 expression, thereby restraining BRCA cell growth and metastasis while enhancing the therapeutic impact of VCR. This investigation offers fresh insights and experimental validation on the interaction between copper homeostasis and BRCA, providing a valuable foundation for refining future treatment strategies. These findings underscore the potential advantages of targeting copper homeostasis and CDK1 in enhancing BRCA therapy, setting the stage for individualized interventions and improved patient consequences.
Collapse
Affiliation(s)
- Nan Shang
- Department of Urinary Surgery, the Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, People's Republic of China
| | - Lisi Zhu
- Department of General Surgery, the Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, People's Republic of China
| | - Yan Li
- Department of General Surgery, the Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, People's Republic of China
| | - Chengyang Song
- Department of Thoracic and Cardiac Surgery, the Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, People's Republic of China.
| | - Xiaodan Liu
- Department of General Surgery, the Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, People's Republic of China.
| |
Collapse
|
23
|
Zheng Y, Huang X. Identification of pyroptosis-associated miRNAs in the immunoscape and prognosis of hepatocellular carcinoma. BMC Cancer 2024; 24:1513. [PMID: 39695414 PMCID: PMC11657507 DOI: 10.1186/s12885-024-13276-5] [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: 04/25/2024] [Accepted: 11/29/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Hepatocellular carcinoma is one of the most prevalent types of liver malignancy and poses a severe threat to global health. Despite recent improvements in therapeutic approaches, treatment options for patients with advanced or recurrent HCC are still limited. MATERIALS AND METHODS Our study analyzed miRNA differential expression using data from hepatocellular carcinoma patients in the Cancer Genome Atlas. Pyroptosis-related genes were identified from gene cards. Differential expression of miRNAs was analyzed using DESeq2 and visualized using ggplot2 and pheatmap. A prognostic risk model for pyroptosis-associated miRNAs was constructed using LASSO regression and validated by principal component analysis, Kaplan-Meier survival and ROC curve analysis. We also performed gene and pathway enrichment analysis. Immune cell infiltration and function in HCC were assessed using single-sample genomic enrichment analysis, and correlations with immune cells and function were explored. Also, CCK-8 assay as well as migration and invasion assays were performed after knockdown of miR-6844. RESULTS We have established and validated a prognostic risk model based on ten DEmiRNAs, which is important for the survival of HCC patients. Significant changes in immune cell infiltration and immune function were also found in high-risk patients. It also demonstrated that knockdown of miR-6844 inhibited HCC cell proliferation, migration and invasion, highlighting its role in HCC progression. CONCLUSION Our study reveals the implications of pyroptosis-associated differential miRNAs on the prognosis of patients with hepatocellular carcinoma and provides a foundation for novel HCC therapies, especially immunotherapy.
Collapse
Affiliation(s)
- Yuting Zheng
- Departments of Anaesthesiology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Xing Huang
- Departments of Anaesthesiology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.
| |
Collapse
|
24
|
Kundu D, Shin SY, Chilian WM, Dong F. The Potential of Mesenchymal Stem Cell-Derived Exosomes in Cardiac Repair. Int J Mol Sci 2024; 25:13494. [PMID: 39769256 PMCID: PMC11727646 DOI: 10.3390/ijms252413494] [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: 11/27/2024] [Revised: 12/12/2024] [Accepted: 12/15/2024] [Indexed: 01/12/2025] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death worldwide, and effectively repairing the heart following myocardial injuries remains a significant challenge. Research has increasingly shown that exosomes derived from mesenchymal stem cells (MSC-Exo) can ameliorate myocardial injuries and improve outcomes after such injuries. The therapeutic benefits of MSC-Exo are largely due to their capacity to deliver specific cargo, including microRNAs and proteins. MSC-Exo can modulate various signaling pathways and provide several beneficial effects, including cytoprotection, inflammation modulation, and angiogenesis promotion to help repair the damaged myocardium. In this review, we summarize the cardioprotective effects of MSC-Exo in myocardial injury, the underlying molecular mechanism involved in the process, and various approaches studied to enhance their efficacy based on recent findings.
Collapse
Affiliation(s)
| | | | | | - Feng Dong
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA; (D.K.); (S.Y.S.); (W.M.C.)
| |
Collapse
|
25
|
Sun Q, Li Y, Yang W, Feng W, Zhou J, Cao Y, Zhang B, Zhu Z, Han C. CircMYH9/miR-133a-3p/CXCR4 axis: a novel regulatory network in sperm fertilization and embryo development. MOLECULAR BIOMEDICINE 2024; 5:69. [PMID: 39671083 PMCID: PMC11645365 DOI: 10.1186/s43556-024-00236-5] [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: 05/16/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 12/14/2024] Open
Abstract
This study aimed to investigate the influence of sperm miRNAs on fertilization rates (FR) in in vitro fertilization (IVF) and to explore potential regulatory mechanisms in sperm-mediated fertilization and embryo development. Through high-throughput sequencing, we identified differentially expressed miRNAs in sperm, with miR-133a-3p significantly upregulated in samples associated with low FR and available embryo rate (AER). Key regulatory circRNAs and mRNAs were further identified via the Starbase database, intersected with differentially expressed RNA, and analyzed through GO, KEGG, and PPI analyses. The circMYH9/miR-133a-3p/CXCR4 axis emerged as a critical regulatory network. In vitro assays using the GC-2 spd mouse spermatogenic cell line revealed that miR-133a-3p inhibited cell growth and proliferation while promoting apoptosis. circMYH9, acting as a competing endogenous RNA (ceRNA) for miR-133a-3p, modulated CXCR4 expression, enhancing GC-2 spd cell growth and inhibiting apoptosis through the miR-133a-3p/CXCR4 axis. In vivo experiments using a mouse model confirmed that circMYH9 overexpression increased IVF success rates and promoted embryo development via this axis. Mechanistically, miR-133a-3p suppresses sperm fertilization and embryo development by targeting the circMYH9/miR-133a-3p/CXCR4 axis. These findings suggest that this regulatory network could serve as a novel biomarker for assessing fertilization potential and embryo quality in clinical settings and as a potential therapeutic target to improve IVF outcomes and address infertility. This study provides valuable insights into the molecular mechanisms governing sperm function and early embryonic development.
Collapse
Affiliation(s)
- Qian Sun
- Suzhou Medical College, Soochow University, Suzhou, 215123, China
- Department of Gynecology, The First Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang, 222061, China
| | - Yanyu Li
- Suzhou Medical College, Soochow University, Suzhou, 215123, China
- Clinical Medicine Postgraduate Workstation, Soochow University, Xuzhou, 221009, China
- Department of Gynecology, Xuzhou Central Hospital, No. 199, South Jiefang Road, Quanshan District, Xuzhou, 221009, China
| | - Wen Yang
- Department of Gynecology, The First Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang, 222061, China
| | - Wen Feng
- Department of Gynecology, The First Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang, 222061, China
| | - Jiayun Zhou
- Department of Gynecology, Xuzhou Central Hospital, No. 199, South Jiefang Road, Quanshan District, Xuzhou, 221009, China
| | - Yijuan Cao
- Department of Reproductive Medicine, Xuzhou Central Hospital, Xuzhou, 221009, China
| | - Bei Zhang
- Suzhou Medical College, Soochow University, Suzhou, 215123, China.
- Clinical Medicine Postgraduate Workstation, Soochow University, Xuzhou, 221009, China.
- Department of Gynecology, Xuzhou Central Hospital, No. 199, South Jiefang Road, Quanshan District, Xuzhou, 221009, China.
| | - Zuobin Zhu
- Xuzhou Engineering Research Center of Medical Genetics and Transformation, Key Laboratory of Genetic Foundation and Clinical Application, Department of Genetics, Xuzhou Medical University, Xuzhou, 221004, China.
| | - Conghui Han
- Clinical Medicine Postgraduate Workstation, Soochow University, Xuzhou, 221009, China.
- Department of Urology, Xuzhou Central Hospital, Xuzhou, 221009, China.
- Department of Urology, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, 221009, China.
- School of Life Sciences, Jiangsu Normal University, Xuzhou, 221116, China.
- Department of Urology, Heilongjiang Provincial Hospital, Harbin, 150006, China.
| |
Collapse
|
26
|
Chen M, Wang R, Liao L, Li Y, Sun X, Wu H, Lan Q, Deng Z, Liu P, Xu T, Zhou H, Liu M. DanShen Decoction targets miR-93-5p to provide protection against MI/RI by regulating the TXNIP/NLRP3/Caspase-1 signaling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156225. [PMID: 39547100 DOI: 10.1016/j.phymed.2024.156225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 09/11/2024] [Accepted: 11/05/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUND Bone marrow mesenchymal stem cells (BMSCs) derived exosomes have demonstrated potential therapeutic efficacy on myocardial ischemia/reperfusion injury (MI/RI). This study has explored the underlying mechanisms of Danshen decoction (DSD) pretreated BMSCs-exosomes to treat MI/RI in vivo and in vitro. METHODS Extracellular vesicles extracted from BMSCs were identified, miRNA sequencing was performed to screen the effects of DSD, and verified to target TXNIP in vivo. After MI/RI modeling, rats were treated with BMSCs-exosomes pretreated with DSD or miRNA inhibitor. BMSCs-exosomes, DSD-pretreated BMSCs-exosomes, and miRNA inhibitor/anti-miRNA-pretreated BMSCs-exosomes were used to treat H9c2 cells or MI/RI rats. CCK-8, Tunnel staining, and flow cytometry were performed to measure cell viability. LDH, CK, CK-MB were detected to evaluate cell injury. MDA, SOD, and ROS were used to confirm oxidative stress. Furthermore, IL-1β, IL-18, cleaved-caspase-1, pro-caspase-1, NLRP3, TXNIP, and GSDMD were quantified for the TXNIP/NLRP3/Caspase-1 signaling activation. In addition, echocardiography was used to observe the heart function, and H&E stain was performed to detect pathological injury. RESULTS Following DSD pretreatment, there was a marked elevation in the expression levels of miR-93-5p, miR-16-5p, and miR-15b-5p, with miR-93-5p exhibiting the highest baseMean value. The administration of a miR-93-5p inhibitor yielded effects counteractive to those observed with DSD treatment, leading to reduced cell proliferation, heightened oxidative stress (as indicated by increased levels of SOD and ROS, alongside a decrease in MDA), and enhanced cell apoptosis. Furthermore, DSD effectively mitigated the miR-93-5p-induced upregulation of key inflammatory and apoptotic markers, including IL-1β, IL-18, caspase-1, NLRP3, TXNIP, and GSDMD. Notably, exosomes derived from DSD-pretreated BMSCs demonstrated a capacity to alleviate cardiac damage. CONCLUSION DSD may target miR-93-5p within BMSC-derived exosomes to confer protection against cardiac damage by inhibiting the activation of the TXNIP/NLRP3/Caspase-1 signaling pathway, thereby mitigating cardiomyocyte pyroptosis. This study provides a theoretical foundation for the application of DSD in the treatment of MI/RI.
Collapse
Affiliation(s)
- Mingtai Chen
- Department of Cardiovascular Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen 518000, PR China; Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau SAR 999078, PR China
| | - Raoqiong Wang
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau SAR 999078, PR China; Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou 646000, PR China
| | - Lishang Liao
- Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou 646000, PR China; College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310000, PR China
| | - Yuanyuan Li
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau SAR 999078, PR China
| | - Xingyu Sun
- Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou 646000, PR China
| | - Hao Wu
- Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou 646000, PR China
| | - Qi Lan
- Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou 646000, PR China
| | - Ziwen Deng
- Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou 646000, PR China
| | - Ping Liu
- Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou 646000, PR China
| | - Tengfei Xu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310000, PR China.
| | - Hua Zhou
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510000, China.
| | - Mengnan Liu
- Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou 646000, PR China; College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310000, PR China.
| |
Collapse
|
27
|
Zhang Z, Zou Y, Song C, Cao K, Cai K, Chen S, Wu Y, Geng D, Sun G, Zhang N, Zhang X, Zhang Y, Sun Y, Zhang Y. Advances in the study of exosomes in cardiovascular diseases. J Adv Res 2024; 66:133-153. [PMID: 38123019 PMCID: PMC11674797 DOI: 10.1016/j.jare.2023.12.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 12/15/2023] [Accepted: 12/16/2023] [Indexed: 12/23/2023] Open
Abstract
BACKGROUND Cardiovascular disease (CVD) has been the leading cause of death worldwide for many years. In recent years, exosomes have gained extensive attention in the cardiovascular system due to their excellent biocompatibility. Studies have extensively researched miRNAs in exosomes and found that they play critical roles in various physiological and pathological processes in the cardiovascular system. These processes include promoting or inhibiting inflammatory responses, promoting angiogenesis, participating in cell proliferation and migration, and promoting pathological progression such as fibrosis. AIM OF REVIEW This systematic review examines the role of exosomes in various cardiovascular diseases such as atherosclerosis, myocardial infarction, ischemia-reperfusion injury, heart failure and cardiomyopathy. It also presents the latest treatment and prevention methods utilizing exosomes. The study aims to provide new insights and approaches for preventing and treating cardiovascular diseases by exploring the relationship between exosomes and these conditions. Furthermore, the review emphasizes the potential clinical use of exosomes as biomarkers for diagnosing cardiovascular diseases. KEY SCIENTIFIC CONCEPTS OF REVIEW Exosomes are nanoscale vesicles surrounded by lipid bilayers that are secreted by most cells in the body. They are heterogeneous, varying in size and composition, with a diameter typically ranging from 40 to 160 nm. Exosomes serve as a means of information communication between cells, carrying various biologically active substances, including lipids, proteins, and small RNAs such as miRNAs and lncRNAs. As a result, they participate in both physiological and pathological processes within the body.
Collapse
Affiliation(s)
- Zhaobo Zhang
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Yuanming Zou
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Chunyu Song
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Kexin Cao
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Kexin Cai
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Shuxian Chen
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Yanjiao Wu
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Danxi Geng
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Guozhe Sun
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China.
| | - Naijin Zhang
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China; Institute of Health Sciences, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, 110122, Liaoning Province, People's Republic of China; Key Laboratory of Reproductive and Genetic Medicine, China Medical University, National Health Commission, 77 Puhe Road, Shenbei New District, Shenyang, 110122, Liaoning Province, People's Republic of China.
| | - Xingang Zhang
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China.
| | - Yixiao Zhang
- Department of Urology Surgery, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning Province, People's Republic of China.
| | - Yingxian Sun
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China; Institute of Health Sciences, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, 110122, Liaoning Province, People's Republic of China.
| | - Ying Zhang
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China; Institute of Health Sciences, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, 110122, Liaoning Province, People's Republic of China.
| |
Collapse
|
28
|
Cao Y, Wang D, Zhou D. MSC Promotes the Secretion of Exosomal lncRNA KLF3-AS1 to Regulate Sphk1 Through YY1-Musashi-1 Axis and Improve Cerebral Ischemia-Reperfusion Injury. Mol Neurobiol 2024; 61:10462-10480. [PMID: 38735900 DOI: 10.1007/s12035-024-04150-3] [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: 03/12/2023] [Accepted: 03/11/2024] [Indexed: 05/14/2024]
Abstract
Stroke remains the 3rd leading cause of long-term disability globally. Over the past decade, mesenchymal stem cell (MSC) transplantation has been proven as an effective therapy for ischemic stroke. However, the mechanism of MSC-derived exosomal lncRNAs during cerebral ischemia/reperfusion (I/R) remains ambiguous. The oxygen-glucose deprivation/reoxygenation (OGD/R) and middle cerebral artery occlusion (MCAO) rat model were generated. MSCs were isolated and characterized by flow cytometry and histochemical staining, and MSC exosomes were purified and characterized by transmission electron microscopy, flow cytometry and Western blot. Western blot, RT-qPCR and ELISA assay were employed to examine the expression or secretion of key molecules. CCK-8 and TUNEL assays were used to assess cell viability and apoptosis. RNA immunoprecipitation and RNA pull-down were used to investigate the direct association between krüppel-like factor 3 antisense RNA 1 (KLF3-AS1) and musashi-1(MSI1). Yin Yang 1 (YY1)-mediated transcriptional regulation was assessed by chromatin immunoprecipitation and luciferase assays. The histological changes and immunoreactivity of key molecules in brain tissues were examined by H&E and immunohistochemistry. MSCs were successfully isolated and exhibited directionally differential potentials. MSC exosomal KLF3-AS1 alleviated OGD/R-induced inflammation in SK-N-SH and SH-SY5Y cells via modulating Sphk1. Mechanistical studies showed that MSI1 positively regulated KLF3-AS1 expression through its direct binding to KLF3-AS1. YY1 was identified as a transcription activator of MSI1 in MSCs. Functionally, YY1/MSI1 axis regulated the release of MSC exosomal KLF3-AS1 to modulate sphingosine kinase 1 (Sphk1)/NF-κB pathway, thereby ameliorating OGD/R- or cerebral I/R-induced injury. MSCs promote the release of exosomal KLF3-AS1 to regulate Sphk1 through YY1/MSI axis and improve cerebral I/R injury.
Collapse
Affiliation(s)
- Yu Cao
- Department of Comprehensive Surgery, Hunan Provincial People's Hospital (The First-Affiliated Hospital of Hunan Normal University), Changsha, 410000, Hunan Province, People's Republic of China
| | - Daodao Wang
- Department of Neurosurgery, Hunan Provincial People's Hospital (The First-Affiliated Hospital of Hunan Normal University), Changsha, 410000, Hunan Province, People's Republic of China
| | - Dingzhou Zhou
- Department of Neurosurgery, Hunan Provincial People's Hospital (The First-Affiliated Hospital of Hunan Normal University), Changsha, 410000, Hunan Province, People's Republic of China.
| |
Collapse
|
29
|
Zhou B, Qin Q, Fang Y, Liu X, Zhang M, Wang S, Zhong L, Guo R. Exosomes from human bone marrow MSCs alleviate PD-1/PD-L1 inhibitor-induced myocardial injury in melanoma mice by regulating macrophage polarization and pyroptosis. Life Sci 2024; 358:123108. [PMID: 39374773 DOI: 10.1016/j.lfs.2024.123108] [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: 05/29/2024] [Revised: 09/30/2024] [Accepted: 10/01/2024] [Indexed: 10/09/2024]
Abstract
Myocarditis, which can be triggered by immune checkpoint inhibitor (ICI) treatment, represents a critical and severe adverse effect observed in cancer therapy. Thus, elucidating the underlying mechanism and developing effective strategies to mitigate its harmful impact is of utmost importance. The objective of this study is to investigate the potential role and regulatory mechanism of exosomes derived from human bone marrow mesenchymal stem cells (hBMSC-Exos) in providing protection against myocardial injury induced by ICIs. We observed that the administration of programmed death 1/programmed death-ligand 1 (PD-1/PD-L1) inhibitor BMS-1 in tumor-bearing mice led to evident cardiac dysfunction and myocardial injury, which were closely associated with M1 macrophage polarization and cardiac pyroptosis. Remarkably, these adverse effects were significantly alleviated through tail-vein injection of hBMSC-Exos. Moreover, either BMS-1 or hBMSC-Exos alone demonstrated the ability to reduce tumor size, while the combination of hBMSC-Exos with BMS-1 treatment not only effectively improved the probability of tumor inhibition but also alleviated cardiac anomalies induced by BMS-1.
Collapse
Affiliation(s)
- Bingqian Zhou
- College of Life Sciences, Institute of Life Science and Green Development, Hebei University, Baoding 071002, China
| | - Qin Qin
- College of Life Sciences, Institute of Life Science and Green Development, Hebei University, Baoding 071002, China
| | - Yue Fang
- College of Life Sciences, Institute of Life Science and Green Development, Hebei University, Baoding 071002, China
| | - Xiaoyu Liu
- College of Life Sciences, Institute of Life Science and Green Development, Hebei University, Baoding 071002, China
| | - Mengyu Zhang
- College of Life Sciences, Institute of Life Science and Green Development, Hebei University, Baoding 071002, China
| | - Shuo Wang
- College of Life Sciences, Institute of Life Science and Green Development, Hebei University, Baoding 071002, China
| | - Li Zhong
- College of Life Sciences, Institute of Life Science and Green Development, Hebei University, Baoding 071002, China; College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Rui Guo
- College of Life Sciences, Institute of Life Science and Green Development, Hebei University, Baoding 071002, China; The Key Laboratory of Zoological Systematics and Application, College of Life Sciences, Hebei University, Baoding 071002, China.
| |
Collapse
|
30
|
Zolfaghari Dehkharghani M, Mousavi S, Kianifard N, Fazlzadeh A, Parsa H, Tavakoli Pirzaman A, Fazlollahpour-Naghibi A. Importance of long non-coding RNAs in the pathogenesis, diagnosis, and treatment of myocardial infarction. IJC HEART & VASCULATURE 2024; 55:101529. [PMID: 39498345 PMCID: PMC11532444 DOI: 10.1016/j.ijcha.2024.101529] [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: 08/15/2024] [Revised: 10/02/2024] [Accepted: 10/07/2024] [Indexed: 11/07/2024]
Abstract
Myocardial infarction (MI), a major global cause of mortality and morbidity, continues to pose a significant burden on public health. Despite advances in understanding its pathogenesis, there remains a need to elucidate the intricate molecular mechanisms underlying MI progression. Long non-coding RNAs (lncRNAs) have emerged as key regulators in diverse biological processes, yet their specific roles in MI pathophysiology remain elusive. Conducting a thorough review of literature using PubMed and Google Scholar databases, we investigated the involvement of lncRNAs in MI, focusing on their regulatory functions and downstream signaling pathways. Our analysis revealed extensive dysregulation of lncRNAs in MI, impacting various biological processes through diverse mechanisms. Notably, lncRNAs act as crucial modulators of gene expression and signaling cascades, functioning as decoys, regulators, and scaffolds. Furthermore, studies identified the multifaceted roles of lncRNAs in modulating inflammation, apoptosis, autophagy, necrosis, fibrosis, remodeling, and ischemia-reperfusion injury during MI progression. Recent research highlights the pivotal contribution of lncRNAs to MI pathogenesis, offering novel insights into potential therapeutic interventions. Moreover, the identification of circulating lncRNA signatures holds promise for the development of non-invasive diagnostic biomarkers. In summary, findings underscore the significance of lncRNAs in MI pathophysiology, emphasizing their potential as therapeutic targets and diagnostic tools for improved patient management and outcomes.
Collapse
Affiliation(s)
| | - Safa Mousavi
- School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nazanin Kianifard
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amin Fazlzadeh
- School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamid Parsa
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | | |
Collapse
|
31
|
He C, Zeng Z, Yang Y, Ye S, Wu Q, Liu X, Liu C, Zeng W, Liu S. Silencing of CircTRIM25/miR-138-5p/CREB1 axis promotes chondrogenesis in osteoarthritis. Autoimmunity 2024; 57:2361749. [PMID: 39007896 DOI: 10.1080/08916934.2024.2361749] [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: 12/14/2023] [Revised: 05/23/2024] [Accepted: 05/26/2024] [Indexed: 07/16/2024]
Abstract
BACKGROUND Dysregulated circular RNAs (circRNAs) are involved in osteoarthritis (OA) progression. OBJECTIVE We aimed to explore the effect of hsa_circ_0044719 (circTRIM25) on the ferroptosis of chondrocytes. METHODS Chondrocytes were treated with interleukin (IL)-1β to generate cell model. Cellular behaviours were measured using cell counting kit-8, enzyme-linked immunosorbent assay, relevant kits, propidium iodide staining, and immunofluorescence assay. Quantitative real-time polymerase chain reaction was performed to examine the expression of circTRIM25, miR-138-5p, and cAMP responsive element binding protein 1 (CREB1), and their interactions were assessed using luciferase reporter analysis and RNA pull-down assay. RESULTS CircTRIM25 was upregulated in OA tissues and IL-1β-stimulated chondrocytes. Knockdown of circTRIM25 facilitated the viability and suppressed ferroptosis and inflammation of IL-1β-induced cells. CircTRIM25 served as a sponge of miR-138-5p, which directly targets CREB1. Downregulation of miR-138-5p abrogated the effect induced by knockdown of circTRIM25. Furthermore, enforced CREB1 reversed the miR-138-5p induced effect. Moreover, knockdown of circTRIM25 attenuated cartilage injury in vivo. CONCLUSION Silencing of circTRIM25 inhibited ferroptosis of chondrocytes via the miR-138-5p/CREB axis and thus attenuated OA progression.
Collapse
Affiliation(s)
- Chunlei He
- Department of Orthopedics, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | | | - Yadong Yang
- Department of Orthopedics, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Shanshan Ye
- Department of Orthopedics, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Qiang Wu
- Gannan Medical University, Ganzhou, China
| | - Xunzhi Liu
- Department of Orthopedics, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Chenghong Liu
- Department of Orthopedics, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Wanhui Zeng
- Department of Orthopedics, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Sheng Liu
- Department of Orthopedics, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| |
Collapse
|
32
|
Chen C, Xia Z, Zhang M, Cao Y, Chen Q, Cao Q, Li X, Jiang F. Molecular mechanism of HDAC6-mediated pyroptosis in neurological function recovery after cardiopulmonary resuscitation in rats. Brain Res 2024; 1843:149121. [PMID: 38997102 DOI: 10.1016/j.brainres.2024.149121] [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: 04/16/2024] [Revised: 07/05/2024] [Accepted: 07/08/2024] [Indexed: 07/14/2024]
Abstract
Brain injury after cardiac arrest (CA) and cardiopulmonary resuscitation (CPR) is the leading cause of neurological dysfunction and death. This study aimed to explore the mechanism of histone deacetylase 6 (HDAC6) in neurofunctional recovery following CA/CPR in rats. A rat model was established by CA/CPR treatment. Adenovirus-packaged sh-HDAC6 was injected into the tail vein. To evaluate the neurofunction of rats, survival time, neurofunctional scores, serum NSE/S100B, and brain water content were measured and Morris water maze test was performed. HDAC6, microRNA (miR)-138-5p, Nod-like receptor protein 3 (NLRP3), and pyroptotic factor levels were determined by real-time quantitative polymerase chain reaction or Western blot assay. HDAC6 and H3K9ac enrichment on miR-138-5p promoter were examined by chromatin immunoprecipitation. miR-138-5p-NLRP3 binding was analyzed by dual-luciferase reporter assay. NLRP3 inflammasome was activated with nigericin sodium salt. After CPR treatment, HDAC6 was highly expressed, while miR-138-5p was downregulated. HDAC6 downregulation improved neurofunction and reduced pyroptosis. HDAC6 enrichment on the miR-138-5p promoter deacetylated H3K9ac, inhibiting miR-138-5p, and promoting NLRP3-mediated pyroptosis. Downregulating miR-138-5p partially reversed the protective effect of HDAC6 inhibition after CPR. In Conclusion, HDAC6 enrichment on miR-138-5p promoter deacetylated H3K9ac, inhibiting miR-138-5p expression and promoting NLRP3-mediated pyroptosis, worsening neurological dysfunction in rats after CPR.
Collapse
Affiliation(s)
- Chunyan Chen
- Department of Infectious Diseases, Minhang Hospital, Fudan University, Shanghai 201199, China
| | - Zhuye Xia
- Department of Critical Care Medicine, Minhang Hospital, Fudan University, Shanghai 201199, China
| | - Min Zhang
- Department of Pathology, Gansu Provincial Hospital, Lanzhou 730000, China
| | - Yunshan Cao
- Department of Cardiology, Gansu Provincial Hospital, Lanzhou 730000, China
| | - Qingling Chen
- Department of Emergency Medicine, Minhang Hospital, Fudan University, Shanghai 201199, China
| | - Qinglian Cao
- Department of Emergency Medicine, Minhang Hospital, Fudan University, Shanghai 201199, China
| | - Xiang Li
- Department of Critical Care Medicine, Minhang Hospital, Fudan University, Shanghai 201199, China
| | - Fan Jiang
- Department of Emergency Medicine, Minhang Hospital, Fudan University, Shanghai 201199, China.
| |
Collapse
|
33
|
Chen H, Sun H, Yang Y, Wang P, Chen X, Yin J, Li A, Zhang L, Cai J, Huang J, Zhang S, Zhang Z, Feng X, Yin J, Wang Y, Xiong W, Wan B. Engineered melatonin-pretreated plasma exosomes repair traumatic spinal cord injury by regulating miR-138-5p/SOX4 axis mediated microglia polarization. J Orthop Translat 2024; 49:230-245. [PMID: 39512441 PMCID: PMC11541837 DOI: 10.1016/j.jot.2024.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 09/08/2024] [Accepted: 09/25/2024] [Indexed: 11/15/2024] Open
Abstract
Background Neuroinflammation plays a crucial role in the repair of spinal cord injury (SCI), with microglia, pivotal in neuroinflammation, driving either degeneration or recovery in this pathological process. Recently, plasma-derived exosomes (denoted Exos) have presented a high capacity for promoting functional recovery of SCI through the anti-inflammatory effects, and pretreated exosomes are associated with better outcomes. Thus, we aimed to explore whether melatonin-pretreated plasma-derived exosomes (denoted MExo) could exert superior effects on SCI, and attempted to elucidate the potential mechanisms. Methods Electron microscopy, nanoparticle tracking analysis, and western blot were applied to delineate the distinctions between Exos and MExos. To assess their therapeutic potentials, we established a contusion SCI rat model, complemented by a battery of in vitro experiments comparing both groups. Subsequently, a miRNA microarray analysis was conducted, followed by a series of rescue experiments to elucidate the specific role of miRNAs in MExos. To further delve into the molecular mechanisms involved, we employed western blot analysis and the luciferase reporter gene assay. Results Melatonin promoted the release of exosome from plasma, concurrently amplifying their anti-inflammatory properties. Furthermore, it was discerned that MExos facilitated a transition in microglia polarization from M1 to M2 phenotype, a phenomenon more pronounced than that observed with Exos. In an endeavor to elucidate this variance, we scrutinized miRNAs exhibiting elevated expression levels in MExos, pinpointing miR-138-5p as a pivotal element in this dynamic. Following this, an in-depth investigation into the role of miR-138-5p was undertaken, which uncovered its efficacy in driving phenotypic alterations within microglia. The analysis of downstream genes targeted by miR-138-5p revealed that it exerted a negative regulatory influence on SOX4, which was found to obstruct the generation of M2-type microglia and the secretion of anti-inflammatory cytokines, thereby partially elucidating the mechanism behind miR-138-5p's regulation of microglia polarization. Conclusions We innovatively observed that melatonin enhanced the anti-inflammatory function of Exos, which further decreased the expression of SOX4 by delivering miR-138-5p. This inhibition promoted the conversion of M1 microglia to M2 microglia, thus offering a viable option for the treatment of SCI. The translational potential of this article This study highlights that melatonin enhances the anti-inflammatory function of Exos through delivery of miR-138-5p. Activation of miR-138-5p/SOX4 axis by engineered melatonin-pretreated plasma exosomes may be a potential target for SCI treatment.
Collapse
Affiliation(s)
- Hao Chen
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University/Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Huihui Sun
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University/Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Yaqing Yang
- Department of Basic Medical Science, Jiangsu Vocational College of Medicine, Yancheng, China
| | - Pingchuan Wang
- Department of Orthopedics, Affiliated Hospital of Yangzhou University, Yangzhou, China
| | - Xizhao Chen
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University/Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Junxiang Yin
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University/Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Aoying Li
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University/Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Liang Zhang
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University/Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Jun Cai
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University/Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Jijun Huang
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University/Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Shengfei Zhang
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University/Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Zhiqiang Zhang
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University/Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Xinmin Feng
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University/Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Jian Yin
- Department of Orthopedics, the Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, China
| | - Yongxiang Wang
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University/Clinical Medical College, Yangzhou University, Yangzhou, China
- Department of Orthopedics, the Yangzhou Clinical Medical College of Xuzhou Medical University, Yangzhou, China
- Department of Orthopedics, Northern Jiangsu People's Hospital, Affiliated Hospital of Nanjing University Medical School, Yangzhou, China
| | - Wu Xiong
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Bowen Wan
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University/Clinical Medical College, Yangzhou University, Yangzhou, China
| |
Collapse
|
34
|
Liu C, Zhang D, Long K, Qi W, Pang L, Li J, Cheng KKY, Cai Y. From exosomes to mitochondria and myocardial infarction: Molecular insight and therapeutic challenge. Pharmacol Res 2024; 209:107468. [PMID: 39426469 DOI: 10.1016/j.phrs.2024.107468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/21/2024] [Accepted: 10/14/2024] [Indexed: 10/21/2024]
Abstract
Myocardial infarction (MI) remains a leading cause of mortality worldwide. Despite patients with MI benefit from timely reperfusion therapies, the rates of mortality and morbidity remain substantial, suggesting an enduring need for the development of new approaches. Molecular mechanisms underlying myocardial ischemic injury are associated with both cardiomyocytes and non-cardiomyocytes. Exosomes are nano-sized extracellular vesicles released by almost all eukaryotic cells. They facilitate the communication between various cells by transferring information via their cargo and altering different biological activities in recipient cells. Studies have created great prospects for therapeutic applications of exosomes in MI, as demonstrated through their beneficial effect on heart function and reducing ventricular remodeling in association with fibrosis, angiogenesis, apoptosis, and inflammation. Of note, myocardial ischemic injury is primarily due to restricted blood flow, reducing oxygen availability, and causing inefficient utilization of energy substrates. However, the impact of exosomes on cardiac energy metabolism has not been adequately investigated. Although exosomes have been engineered for targeted delivery to enhance clinical efficacy, challenges must be overcome to utilize them reliably in the clinic. In this review, we summarize the research progress of exosomes for MI with a focus on the known and unknown regarding the role of exosomes in energy metabolism in cardiomyocytes and non-cardiomyocytes; as well as potential research avenues of exosome-mitochondrial energy regulation as well as therapeutic challenges. We aim to help identify more efficient molecular targets that may promote the clinical application of exosomes.
Collapse
Affiliation(s)
- Chang Liu
- Department of Anesthesiology, The First Hospital of Jilin University, Jilin, China; Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Dengwen Zhang
- Department of Anesthesiology, Heyuan People's Hospital, Guangdong, China; Department of Anesthesiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangdong, China
| | - Kekao Long
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Wensheng Qi
- Department of Anesthesiology, The First Hospital of Jilin University, Jilin, China; Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Lei Pang
- Department of Anesthesiology, The First Hospital of Jilin University, Jilin, China
| | - Jia Li
- Department of Neurology, Wuhan No.1 Hospital, Hubei, China
| | - Kenneth King-Yip Cheng
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China.
| | - Yin Cai
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China; Research Center for Chinese Medicine Innovation, The Hong Kong Polytechnic University, Hong Kong SAR, China; Research Institute for Future Food, The Hong Kong Polytechnic University, Hong Kong SAR, China.
| |
Collapse
|
35
|
Ji Z, Wang C. Mesenchymal stem cell-derived exosomal mir-21-5p inhibits YAP1 expression and improves outcomes in myocardial infarction. BMC Cardiovasc Disord 2024; 24:547. [PMID: 39385107 PMCID: PMC11465913 DOI: 10.1186/s12872-024-04197-z] [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: 03/29/2024] [Accepted: 09/16/2024] [Indexed: 10/11/2024] Open
Abstract
BACKGROUND Myocardial infarction (MI) remains a significant global health concern, characterized by cardiomyocyte apoptosis and adverse ventricular remodeling. Nevertheless, the interplay between exosomal miR-21-5p and Yes-associated protein 1 (YAP1) in the context of MI remains unexplored. METHODS Rat mesenchymal stem cells (MSCs) and H9c2 cardiomyocytes were cultured, characterized, and instrumental in our experiments. Exosomes were meticulously isolated, and their identity confirmed. The internalization of these exosomes by H9c2 cells was assessed, while RNA and protein expression were quantified using Quantitative Real-Time PCR and Western blot, respectively. MTT assay was implemented for cell viability, and apoptosis was evaluated via flow cytometric analysis. To elucidate gene interactions, we conducted microarray profiling of miRNA expression, dual luciferase reporter assays, and RNA Immunoprecipitation. RESULTS MSC-derived exosomes exhibited a remarkable capacity to attenuate hypoxia-induced inflammation and apoptosis in H9c2 cells. Notably, these exosomes significantly upregulated miR-21-5p levels within H9c2 cells, and the abrogation of miR-21-5p function abated their protective effects. Through computational analysis, we unveiled a miR-21-5p binding site in the 3'UTR of YAP1, which directly inhibited YAP1 expression. Importantly, the inhibition of YAP1 effectively reinstated the protective effects of exosomes in cells with impaired exosomal miR-21-5p. CONCLUSION This study underscores the pivotal role played by MSC-derived exosomes in safeguarding against MI, primarily by mediating the transfer of miR-21-5p, which targets YAP1 signaling pathways. CLINICAL TRIAL NUMBER N/A.
Collapse
Affiliation(s)
- Zhou Ji
- Department of Cardiovascular Medicine, The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Chan Wang
- Jinzhou Hospital of Traditional Chinese Medicine, No. 91, Shifu Road, Taihe District, Jinzhou, Liaoning, 121000, China.
| |
Collapse
|
36
|
Abid AI, Conzatti G, Toti F, Anton N, Vandamme T. Mesenchymal stem cell-derived exosomes as cell free nanotherapeutics and nanocarriers. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2024; 61:102769. [PMID: 38914247 DOI: 10.1016/j.nano.2024.102769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 05/18/2024] [Accepted: 06/20/2024] [Indexed: 06/26/2024]
Abstract
Many strategies for regenerating the damaged tissues or degenerating cells are employed in regenerative medicine. Stem cell technology is a modern strategy of the recent approaches, particularly the use of mesenchymal stem cells (MCSs). The ability of MSCs to differentiate as well as their characteristic behaviour as paracrine effector has established them as key elements in tissue repair. Recently, extracellular vesicles (EVs) shed by MSCs have emerged as a promising cell free therapy. This comprehensive review encompasses MSCs-derived exosomes and their therapeutic potential as nanotherapeutics. We also discuss their potency as drug delivery nano-carriers in comparison with liposomes. A better knowledge of EVs behaviour in vivo and of their mechanism of action are key to determine parameters of an optimal formulation in pilot studies and to establish industrial processes.
Collapse
Affiliation(s)
- Ali Imran Abid
- UMR 1260, Regenerative Nanomedicine (RNM), INSERM (French National Institute of Health and Medical Research), University of Strasbourg, F-67000 Strasbourg, France
| | - Guillaume Conzatti
- UMR 1260, Regenerative Nanomedicine (RNM), INSERM (French National Institute of Health and Medical Research), University of Strasbourg, F-67000 Strasbourg, France; Faculty of Pharmacy, University of Strasbourg, 67400 Illkirch-Graffenstaden, France.
| | - Florence Toti
- UMR 1260, Regenerative Nanomedicine (RNM), INSERM (French National Institute of Health and Medical Research), University of Strasbourg, F-67000 Strasbourg, France; Faculty of Pharmacy, University of Strasbourg, 67400 Illkirch-Graffenstaden, France
| | - Nicolas Anton
- UMR 1260, Regenerative Nanomedicine (RNM), INSERM (French National Institute of Health and Medical Research), University of Strasbourg, F-67000 Strasbourg, France; Faculty of Pharmacy, University of Strasbourg, 67400 Illkirch-Graffenstaden, France
| | - Thierry Vandamme
- UMR 1260, Regenerative Nanomedicine (RNM), INSERM (French National Institute of Health and Medical Research), University of Strasbourg, F-67000 Strasbourg, France; Faculty of Pharmacy, University of Strasbourg, 67400 Illkirch-Graffenstaden, France.
| |
Collapse
|
37
|
Chen C, Wang J, Zhang S, Zhu X, Hu J, Liu C, Liu L. Epigenetic regulation of diverse regulated cell death modalities in cardiovascular disease: Insights into necroptosis, pyroptosis, ferroptosis, and cuproptosis. Redox Biol 2024; 76:103321. [PMID: 39186883 PMCID: PMC11388786 DOI: 10.1016/j.redox.2024.103321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 08/17/2024] [Accepted: 08/18/2024] [Indexed: 08/28/2024] Open
Abstract
Cell death constitutes a critical component of the pathophysiology of cardiovascular diseases. A growing array of non-apoptotic forms of regulated cell death (RCD)-such as necroptosis, ferroptosis, pyroptosis, and cuproptosis-has been identified and is intimately linked to various cardiovascular conditions. These forms of RCD are governed by genetically programmed mechanisms within the cell, with epigenetic modifications being a common and crucial regulatory method. Such modifications include DNA methylation, RNA methylation, histone methylation, histone acetylation, and non-coding RNAs. This review recaps the roles of DNA methylation, RNA methylation, histone modifications, and non-coding RNAs in cardiovascular diseases, as well as the mechanisms by which epigenetic modifications regulate key proteins involved in cell death. Furthermore, we systematically catalog the existing epigenetic pharmacological agents targeting novel forms of RCD and their mechanisms of action in cardiovascular diseases. This article aims to underscore the pivotal role of epigenetic modifications in precisely regulating specific pathways of novel RCD in cardiovascular diseases, thus offering potential new therapeutic avenues that may prove more effective and safer than traditional treatments.
Collapse
Affiliation(s)
- Cong Chen
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing, 100053, China
| | - Jie Wang
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing, 100053, China.
| | - Shan Zhang
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Xueying Zhu
- Department of Anatomy, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Jun Hu
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing, 100053, China
| | - Chao Liu
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing, 100053, China
| | - Lanchun Liu
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing, 100053, China
| |
Collapse
|
38
|
Hou Q, Ouyang S, Xie Z, He Y, Deng Y, Guo J, Yu P, Tan X, Ma W, Li P, Yu J, Mo Q, Zhang Z, Chen D, Lin X, Liu Z, Chen X, Peng T, Li L, Xie W. Exosome is a Fancy Mobile Sower of Ferroptosis. J Cardiovasc Transl Res 2024; 17:1067-1082. [PMID: 38776048 DOI: 10.1007/s12265-024-10508-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 04/01/2024] [Indexed: 10/29/2024]
Abstract
Exosomes, nano-sized small extracellular vesicles, have been shown to serve as mediators between intercellular communications by transferring bioactive molecules, such as non-coding RNA, proteins, and lipids from secretory to recipient cells, modulating a variety of physiological and pathophysiological processes. Recent studies have gradually demonstrated that altered exosome charges may represent a key mechanism driving the pathological process of ferroptosis. This review summarizes the potential mechanisms and signal pathways relevant to ferroptosis and then discusses the roles of exosome in ferroptosis. As well as transporting iron, exosomes may also indirectly convey factors related to ferroptosis. Furthermore, ferroptosis may be transmitted to adjacent cells through exosomes, resulting in cascading effects. It is expected that further research on exosomes will be conducted to explore their potential in ferroptosis and will lead to the creation of new therapeutic avenues for clinical diseases.
Collapse
Affiliation(s)
- Qin Hou
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Medical Research Center, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Siyu Ouyang
- Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Zhongcheng Xie
- Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Yinling He
- Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Yunong Deng
- Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Jiamin Guo
- Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Panpan Yu
- Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Xiaoqian Tan
- Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Wentao Ma
- Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Pin Li
- Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Jiang Yu
- Class of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Qinger Mo
- Class of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Zhixia Zhang
- Class of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Dandan Chen
- Class of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Xiaoyan Lin
- Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Zhiyang Liu
- Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Xi Chen
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Tianhong Peng
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| | - Liang Li
- Department of Physiology, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| | - Wei Xie
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
39
|
Zhang CH, Lu DC, Liu Y, Wang L, Sethi G, Ma Z. The role of extracellular vesicles in pyroptosis-mediated infectious and non-infectious diseases. Int Immunopharmacol 2024; 138:112633. [PMID: 38986299 DOI: 10.1016/j.intimp.2024.112633] [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: 04/06/2024] [Revised: 06/22/2024] [Accepted: 07/02/2024] [Indexed: 07/12/2024]
Abstract
Pyroptosis, a lytic and pro-inflammatory cell death, is important in various pathophysiological processes. Host- and bacteria-derived extracellular vesicles (EVs), as natural nanocarriers messengers, are versatile mediators of intercellular communication between different types of cells. Recently, emerging research has suggested that EVs exhibit multifaceted roles in disease progression by manipulating pyroptosis. This review focuses on new findings concerning how EVs shape disease progression in infectious and non-infectious diseases by regulating pyroptosis. Understanding the characteristics and activity of EVs-mediated pyroptotic death may conducive to the discovery of novel mechanisms and more efficient therapeutic targets in infectious and non-infectious diseases.
Collapse
Affiliation(s)
- Cai-Hua Zhang
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, China; Department of Oncology, People's Hospital Affiliated to Chongqing Three Gorges Medical College, Chongqing 404100, China
| | - Ding-Ci Lu
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, China
| | - Ying Liu
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, China
| | - Lingzhi Wang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600 Singapore; Cancer Science Institute of Singapore, National University of Singapore, 117599 Singapore; NUS Centre for Cancer Research (N2CR), National University of Singapore, 117599 Singapore.
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600 Singapore; NUS Centre for Cancer Research (N2CR), National University of Singapore, 117599 Singapore.
| | - Zhaowu Ma
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, China.
| |
Collapse
|
40
|
Qiu S, Cao L, Xiang D, Wang S, Wang D, Qian Y, Li X, Zhou X. Enhanced osteogenic differentiation in 3D hydrogel scaffold via macrophage mitochondrial transfer. J Nanobiotechnology 2024; 22:540. [PMID: 39237942 PMCID: PMC11375923 DOI: 10.1186/s12951-024-02757-1] [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: 03/22/2024] [Accepted: 08/05/2024] [Indexed: 09/07/2024] Open
Abstract
To assess the efficacy of a novel 3D biomimetic hydrogel scaffold with immunomodulatory properties in promoting fracture healing. Immunomodulatory scaffolds were used in cell experiments, osteotomy mice treatment, and single-cell transcriptomic sequencing. In vitro, fluorescence tracing examined macrophage mitochondrial transfer and osteogenic differentiation of bone marrow-derived mesenchymal stem cells (BMSCs). Scaffold efficacy was assessed through alkaline phosphatase (ALP), Alizarin Red S (ARS) staining, and in vivo experiments. The scaffold demonstrated excellent biocompatibility and antioxidant-immune regulation. Single-cell sequencing revealed a shift in macrophage distribution towards the M2 phenotype. In vitro experiments showed that macrophage mitochondria promoted BMSCs' osteogenic differentiation. In vivo experiments confirmed accelerated fracture healing. The GAD/Ag-pIO scaffold enhances osteogenic differentiation and fracture healing through immunomodulation and promotion of macrophage mitochondrial transfer.
Collapse
Affiliation(s)
- Shui Qiu
- Department of Orthopedics, First Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, Liaoning Province, China
| | - Lili Cao
- Department of Medical Oncology, First Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, China
| | - Dingding Xiang
- School of Mechanical Engineering and Automation, Foshan Graduate School of Innovation, Northeastern University, Shenyang, 110819, China
| | - Shu Wang
- School of Mechanical Engineering and Automation, Foshan Graduate School of Innovation, Northeastern University, Shenyang, 110819, China
| | - Di Wang
- School of Mechanical Engineering and Automation, Foshan Graduate School of Innovation, Northeastern University, Shenyang, 110819, China
| | - Yiyi Qian
- School of Mechanical Engineering and Automation, Foshan Graduate School of Innovation, Northeastern University, Shenyang, 110819, China
| | - Xiaohua Li
- Department of Orthopedics, Zhongmeng Hospital, Arong Banner, Hulunbuir City, Inner, Mongolia
| | - Xiaoshu Zhou
- Department of Orthopedics, First Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, Liaoning Province, China.
| |
Collapse
|
41
|
Ma T, Wang M, Wang S, Hu H, Zhang X, Wang H, Wang G, Jin Y. BMSC derived EVs inhibit colorectal Cancer progression by transporting MAGI2-AS3 or something similar. Cell Signal 2024; 121:111235. [PMID: 38806109 DOI: 10.1016/j.cellsig.2024.111235] [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: 01/14/2024] [Revised: 05/16/2024] [Accepted: 05/24/2024] [Indexed: 05/30/2024]
Abstract
In this study, we investigated the molecular mechanisms underlying the impact of extracellular vesicles (EVs) derived from bone marrow stromal cells (BMSCs) on colorectal cancer (CRC) development. The focus was on the role of MAGI2-AS3, delivered by BMSC-EVs, in regulating USP6NL DNA methylation-mediated MYC protein translation modification to promote CDK2 downregulation. Utilizing bioinformatics analysis, we identified significant enrichment of MAGI2-AS3 related to copper-induced cell death in CRC. In vitro experiments demonstrated the downregulation of MAGI2-AS3 in CRC cells, and BMSC-EVs were found to deliver MAGI2-AS3 to inhibit CRC cell proliferation, migration, and invasion. Further exploration revealed that MAGI2-AS3 suppressed MYC protein translation modification by regulating USP6NL DNA methylation, leading to CDK2 downregulation and prevention of colorectal cancer. Overexpression of MYC reversed the functional effects of BMSC-EVs-MAGI2-AS3. In vivo experiments validated the inhibitory impact of BMSC-EVs-MAGI2-AS3 on CRC tumorigenicity by promoting CDK2 downregulation through USP6NL DNA methylation-mediated MYC protein translation modification. Overall, BMSC-EVs-MAGI2-AS3 may serve as a potential intervention to prevent CRC occurrence by modulating key molecular pathways.
Collapse
Affiliation(s)
- Tianyi Ma
- Department of Colorectal Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin 150000, China
| | - Meng Wang
- Department of Colorectal Surgery, Zhejiang Cancer Hospital (Affiliated Cancer Hospital of the Chinese Academy of Sciences), Hangzhou 310000, China
| | - Song Wang
- Department of Colorectal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Hanqing Hu
- Department of Colorectal Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin 150000, China
| | - Xin Zhang
- Department of Colorectal Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin 150000, China
| | - Hufei Wang
- Department of Colorectal Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin 150000, China
| | - Guiyu Wang
- Department of Colorectal Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin 150000, China.
| | - Yinghu Jin
- Department of Colorectal Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin 150000, China.
| |
Collapse
|
42
|
Yang L, Liu N, Yang Y. Astragaloside IV-induced BMSC exosomes promote neovascularization and protect cardiac function in myocardial infarction mice via the miR-411/HIF-1α axis. J Liposome Res 2024; 34:452-463. [PMID: 38088046 DOI: 10.1080/08982104.2023.2293844] [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: 08/29/2023] [Accepted: 12/07/2023] [Indexed: 12/26/2023]
Abstract
This study focused on investigating the mechanism of the astragaloside IV-induced bone marrow mesenchymal stem cell exosome (AS-IV-MSC-exo)/microRNA(miR)-411/HIF-1α axis in affecting vascular neovascularization and protecting cardiac function in myocardial infarction (MI) mice. Exosomes (MSC-exo and AS-IV-MSC-exo) were separated by differential centrifugation and then characterized. MI mouse models were established by left anterior descending coronary artery ligation. Echocardiography was used to evaluate cardiac function. HE staining and Masson staining were performed to observe myocardial histopathology. Capillary density in the myocardium via immunohistochemistry and quantified the expression of vascular endothelial growth factor (VEGF) via RT-qPCR. The expression of miR-411 and HIF-1α was tested by RT-qPCR and western blot and the targeting relationship of miR-411 and HIF-1α was verified by bioinformatics website and dual luciferase reporter gene assay. Exosomes with lipid bi-layer membrane structure, expressing exosomal surface marker proteins, and being taken up by cardiomyocytes could be successfully isolated utilizing ultracentrifugation. Intramyocardial injection of MSC-exo could restore cardiac function, decrease myocardial pathological changes and collagen deposition, and promote neovascularization in MI mice; the effect of AS-IV-MSC-exo was more significant. The ability of AS-IV-MSC-exo to restore cardiac function, lower myocardial pathological changes and collagen deposition, and promote neovascularization in MI mice was diminished when miR-411 expression in AS-IV-MSC-exo was reduced. Mechanistically, miR-411 was found to target and inhibit HIF-1α expression. Overexpression of HIF-1α impaired the impact of AS-IV-MSC-exo on improving cardiac function and promoting neovascularization in MI mice. AS-IV-MSC-exo improves cardiac function and promoted neovascularization via the miR-411/HIF-1α axis, thereby ameliorating MI.
Collapse
Affiliation(s)
- Lei Yang
- School of Medicine, Zhumadian Key Laboratory of Chronic Disease Research and Translational Medicine, Huanghuai University, Zhumadian, People's Republic of China
- Department of Scientific Research Section, Zhumadian Central Hospital, Affiliated Hospital of Huanghuai University, Zhumadian, People's Republic of China
| | - Nuan Liu
- School of Medicine, Zhumadian Key Laboratory of Chronic Disease Research and Translational Medicine, Huanghuai University, Zhumadian, People's Republic of China
- Department of Scientific Research Section, Zhumadian Central Hospital, Affiliated Hospital of Huanghuai University, Zhumadian, People's Republic of China
- Institute of Cardiovascular and Cerebrovascular Diseases, Huanghuai University, Zhumadian, People's Republic of China
| | - Yang Yang
- Department of Scientific Research Section, Zhumadian Central Hospital, Affiliated Hospital of Huanghuai University, Zhumadian, People's Republic of China
| |
Collapse
|
43
|
Guo Q, Wang J, Ni C, Pan J, Zou J, Shi Y, Sun J, Zhang X, Wang D, Luan F. Research progress on the natural products in the intervention of myocardial infarction. Front Pharmacol 2024; 15:1445349. [PMID: 39239656 PMCID: PMC11374734 DOI: 10.3389/fphar.2024.1445349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 08/05/2024] [Indexed: 09/07/2024] Open
Abstract
Coronary heart disease is a prevalent cardiovascular ailment globally, with myocardial infarction (MI) being one of its most severe manifestations. The morbidity and mortality of MI are escalating, showing an increasing trend among younger, highly educated individuals, thereby posing a serious threat to public health. Currently, thrombolysis, percutaneous coronary intervention, and coronary artery bypass grafting are the primary clinical treatments for MI. Although these methods significantly reduce patient mortality, complications often result in poor prognoses. Due to limitations in chemical synthetic drug research, the focus has shifted towards developing herbs based on natural substances. Natural medicines represent a novel approach for safer and more effective MI management and treatment. They can control multiple pathogenic variables by targeting various pathways and systems. This paper investigates the molecular mechanisms of MI and evaluates the application of natural products and medicinal plants in MI treatment over the past 5 years, demonstrating their specific good therapeutic potential and superior tolerance. These natural therapies have been shown to mitigate myocardial cell damage caused by MI through mechanisms such as oxidative stress, inflammation, apoptosis, angiogenesis, myocardial fibrosis, autophagy, endoplasmic reticulum stress, mitophagy, and pyroptosis. This review offers the latest insights into the application of natural products and medicinal plants in MI treatment, elucidating their mechanisms of action and serving as an important reference for MI prevention.
Collapse
Affiliation(s)
- Qiuting Guo
- College of Pharmacy, Xianyang Polytechnic Institute, Xianyang, China
| | - Jinhui Wang
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, Shaanxi, China
| | - Caixia Ni
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, Sichuan, China
| | - Jiaojiao Pan
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, Shaanxi, China
| | - Junbo Zou
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, Shaanxi, China
| | - Yajun Shi
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, Shaanxi, China
| | - Jing Sun
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, Shaanxi, China
| | - Xiaofei Zhang
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, Shaanxi, China
| | - Deng Wang
- Department of Pharmacy, Xi'an No. 3 Hospital, The Affiliated Hospital of Northwest University, Xi'an, Shaanxi, China
| | - Fei Luan
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, Shaanxi, China
| |
Collapse
|
44
|
Zhan T, Zou Y, Han Z, Tian X, Chen M, Liu J, Yang X, Zhu Q, Liu M, Chen W, Chen M, Huang X, Tan J, Liu W, Tian X. Single-cell sequencing combined with spatial transcriptomics reveals that the IRF7 gene in M1 macrophages inhibits the occurrence of pancreatic cancer by regulating lipid metabolism-related mechanisms. Clin Transl Med 2024; 14:e1799. [PMID: 39118300 PMCID: PMC11310283 DOI: 10.1002/ctm2.1799] [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: 07/24/2024] [Revised: 07/24/2024] [Accepted: 07/26/2024] [Indexed: 08/10/2024] Open
Abstract
AIM The main focus of this study is to explore the molecular mechanism of IRF7 regulation on RPS18 transcription in M1-type macrophages in pancreatic adenocarcinoma (PAAD) tissue, as well as the transfer of RPS18 by IRF7 via exosomes to PAAD cells and the regulation of ILF3 expression. METHODS By utilising single-cell RNA sequencing (scRNA-seq) data and spatial transcriptomics (ST) data from the Gene Expression Omnibus database, we identified distinct cell types with significant expression differences in PAAD tissue. Among these cell types, we identified those closely associated with lipid metabolism. The differentially expressed genes within these cell types were analysed, and target genes relevant to prognosis were identified. Flow cytometry was employed to assess the expression levels of target genes in M1 and M2 macrophages. Cell lines with target gene knockout were constructed using CRISPR/Cas9 editing technology, and cell lines with target gene knockdown and overexpression were established using lentiviral vectors. Additionally, a co-culture model of exosomes derived from M1 macrophages with PAAD cells was developed. The impact of M1 macrophage-derived exosomes on the lipid metabolism of PAAD cells in the model was evaluated through metabolomics analysis. The effects of M1 macrophage-derived exosomes on the viability, proliferation, division, migration and apoptosis of PAAD cells were assessed using MTT assay, flow cytometry, EdU assay, wound healing assay, Transwell assay and TUNEL staining. Furthermore, a mouse PAAD orthotopic implantation model was established, and bioluminescence imaging was utilised to assess the influence of M1 macrophage-derived exosomes on the intratumoural formation capacity of PAAD cells, as well as measuring tumour weight and volume. The expression of proliferation-associated proteins in tumour tissues was examined using immunohistochemistry. RESULTS Through combined analysis of scRNA-seq and ST technologies, we discovered a close association between M1 macrophages in PAAD samples and lipid metabolism signals, as well as a negative correlation between M1 macrophages and cancer cells. The construction of a prognostic risk score model identified RPS18 and IRF7 as two prognostically relevant genes in M1 macrophages, exhibiting negative and positive correlations, respectively. Mechanistically, it was found that IRF7 in M1 macrophages can inhibit the transcription of RPS18, reducing the transfer of RPS18 to PAAD cells via exosomes, consequently affecting the expression of ILF3 in PAAD cells. IRF7/RPS18 in M1 macrophages can also suppress lipid metabolism, cell viability, proliferation, migration, invasion and intratumoural formation capacity of PAAD cells, while promoting cell apoptosis. CONCLUSION Overexpression of IRF7 in M1 macrophages may inhibit RPS18 transcription, reduce the transfer of RPS18 from M1 macrophage-derived exosomes to PAAD cells, thereby suppressing ILF3 expression in PAAD cells, inhibiting the lipid metabolism pathway, and curtailing the viability, proliferation, migration, invasion of PAAD cells, as well as enhancing cell apoptosis, ultimately inhibiting tumour formation in PAAD cells in vivo. Targeting IRF7/RPS18 in M1 macrophages could represent a promising immunotherapeutic approach for PAAD in the future.
Collapse
Affiliation(s)
- Ting Zhan
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
| | - Yanli Zou
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
| | - Zheng Han
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
| | - XiaoRong Tian
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Mengge Chen
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Jiaxi Liu
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Xiulin Yang
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Qingxi Zhu
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
| | - Meng Liu
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
| | - Wei Chen
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
| | - Mingtao Chen
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
| | - Xiaodong Huang
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Jie Tan
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
| | - Weijie Liu
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
| | - Xia Tian
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
| |
Collapse
|
45
|
Wan M, Yu Q, Xu F, You LX, Liang X, Kang Ren K, Zhou J. Novel hypoxia-induced HIF-1αactivation in asthma pathogenesis. Respir Res 2024; 25:287. [PMID: 39061007 PMCID: PMC11282634 DOI: 10.1186/s12931-024-02869-0] [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: 03/12/2024] [Accepted: 06/06/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Asthma's complexity, marked by airway inflammation and remodeling, is influenced by hypoxic conditions. This study focuses on the role of Hypoxia-Inducible Factor-1 Alpha (HIF-1α) and P53 ubiquitination in asthma exacerbation. METHODS High-throughput sequencing and bioinformatics were used to identify genes associated with asthma progression, with an emphasis on GO and KEGG pathway analyses. An asthma mouse model was developed, and airway smooth muscle cells (ASMCs) were isolated to create an in vitro hypoxia model. Cell viability, proliferation, migration, and apoptosis were assessed, along with ELISA and Hematoxylin and Eosin (H&E) staining. RESULTS A notable increase in HIF-1α was observed in both in vivo and in vitro asthma models. HIF-1α upregulation enhanced ASMCs' viability, proliferation, and migration, while reducing apoptosis, primarily via the promotion of P53 ubiquitination through MDM2. In vivo studies showed increased inflammatory cell infiltration and airway structural changes, which were mitigated by the inhibitor IDF-11,774. CONCLUSION The study highlights the critical role of the HIF-1α-MDM2-P53 axis in asthma, suggesting its potential as a target for therapeutic interventions. The findings indicate that modulating this pathway could offer new avenues for treating the complex respiratory disorder of asthma.
Collapse
Affiliation(s)
- Mengzhi Wan
- Department of Respiratory Emergency and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang, Jiangxi Province, 330006, PR China
| | - Qi Yu
- Department of Respiratory Emergency and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang, Jiangxi Province, 330006, PR China
| | - Fei Xu
- Department of Respiratory Emergency and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang, Jiangxi Province, 330006, PR China
| | - Lu Xia You
- Department of Respiratory Emergency and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang, Jiangxi Province, 330006, PR China
| | - Xiao Liang
- Department of Respiratory Emergency and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang, Jiangxi Province, 330006, PR China
| | - Kang Kang Ren
- Department of Respiratory Emergency and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang, Jiangxi Province, 330006, PR China
| | - Jing Zhou
- Department of Respiratory Emergency and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang, Jiangxi Province, 330006, PR China.
| |
Collapse
|
46
|
Xu H, Ma H, Zha L, Li Q, Pan H, Zhang L. Engineered exosomes transporting the lncRNA, SVIL-AS1, inhibit the progression of lung cancer via targeting miR-21-5p. Am J Cancer Res 2024; 14:3335-3347. [PMID: 39113865 PMCID: PMC11301303 DOI: 10.62347/yrjk5888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 06/20/2024] [Indexed: 08/10/2024] Open
Abstract
In this study, we constructed engineered exosomes carrying the long non-coding RNA (lncRNA) SVIL-AS1 (SVIL-AS1 Exos), and explored its role and mechanism in lung cancer. After the construction of SVIL-AS1 Exos, their physicochemical characteristics were identified. Then, their function and effect in three different cell lines, A549, HeLa, and HepG2, were detected using western blot, the quantitative reverse transcriptase polymerase chain reaction, flow cytometry, 5-ethynyl-2'-deoxyuridine, and Cell Counting Kit-8 experiments. Finally, a mouse xenograft model was constructed to analyze tumor growth and explore the in vivo utility of SVIL-AS1 Exos using hematoxylin and eosin staining, immunohistochemistry, and the TdT-mediated dUTP nick end labeling assay. The results demonstrated that SVIL-AS1 Exos preferentially targeted A549 lung cancer cells over HeLa and HepG2 cells. SVIL-AS1 Exos promoted apoptosis and inhibited A549 cell proliferation by elevating expression of the lncRNA, SVIL-AS1. In vivo, SVIL-AS1 Exos effectively inhibited the growth of lung cancer A549 cells. Furthermore, SVIL-AS1 Exos suppressed the expression of miR-21-5p and upregulated the expression of caspase-9, indicating that SVIL-AS1 may regulate the development of lung cancer through the miR-21-5p/caspase-9 pathway. In conclusion, the engineered SVIL-AS1 Exos targeted lung cancer cells to inhibit the expression of miR-21-5p, upregulate the expression of caspase-9, and inhibit the development of lung cancer.
Collapse
Affiliation(s)
- Hao Xu
- Department of Respiratory, The People's Hospital of Danyang, Affiliated Danyang Hospital of Nantong University Danyang 212300, Jiangsu, China
| | - Hongda Ma
- Department of Respiratory, The People's Hospital of Danyang, Affiliated Danyang Hospital of Nantong University Danyang 212300, Jiangsu, China
| | - Lifen Zha
- Department of Respiratory, The People's Hospital of Danyang, Affiliated Danyang Hospital of Nantong University Danyang 212300, Jiangsu, China
| | - Qian Li
- Department of Respiratory, The People's Hospital of Danyang, Affiliated Danyang Hospital of Nantong University Danyang 212300, Jiangsu, China
| | - Huiming Pan
- Department of Respiratory, The People's Hospital of Danyang, Affiliated Danyang Hospital of Nantong University Danyang 212300, Jiangsu, China
| | - Ladi Zhang
- Department of Respiratory, The People's Hospital of Danyang, Affiliated Danyang Hospital of Nantong University Danyang 212300, Jiangsu, China
| |
Collapse
|
47
|
Zhao W, Li K, Li L, Wang R, Lei Y, Yang H, Sun L. Mesenchymal Stem Cell-Derived Exosomes as Drug Delivery Vehicles in Disease Therapy. Int J Mol Sci 2024; 25:7715. [PMID: 39062956 PMCID: PMC11277139 DOI: 10.3390/ijms25147715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/09/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
Exosomes are small vesicles containing proteins, nucleic acids, and biological lipids, which are responsible for intercellular communication. Studies have shown that exosomes can be utilized as effective drug delivery vehicles to accurately deliver therapeutic substances to target tissues, enhancing therapeutic effects and reducing side effects. Mesenchymal stem cells (MSCs) are a class of stem cells widely used for tissue engineering, regenerative medicine, and immunotherapy. Exosomes derived from MSCs have special immunomodulatory functions, low immunogenicity, the ability to penetrate tumor tissues, and high yield, which are expected to be engineered into efficient drug delivery systems. Despite the promising promise of MSC-derived exosomes, exploring their optimal preparation methods, drug-loading modalities, and therapeutic potential remains challenging. Therefore, this article reviews the related characteristics, preparation methods, application, and potential risks of MSC-derived exosomes as drug delivery systems in order to find potential therapeutic breakthroughs.
Collapse
Affiliation(s)
- Wenzhe Zhao
- School of Life Sciences, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment in Special Environment, Northwestern Polytechnical University, Xi’an 710072, China; (W.Z.); (K.L.); (L.L.); (R.W.); (Y.L.)
- Dongguan Sanhang Innovation Institute, Dongguan 523808, China
| | - Kaixuan Li
- School of Life Sciences, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment in Special Environment, Northwestern Polytechnical University, Xi’an 710072, China; (W.Z.); (K.L.); (L.L.); (R.W.); (Y.L.)
- Dongguan Sanhang Innovation Institute, Dongguan 523808, China
| | - Liangbo Li
- School of Life Sciences, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment in Special Environment, Northwestern Polytechnical University, Xi’an 710072, China; (W.Z.); (K.L.); (L.L.); (R.W.); (Y.L.)
- Dongguan Sanhang Innovation Institute, Dongguan 523808, China
| | - Ruichen Wang
- School of Life Sciences, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment in Special Environment, Northwestern Polytechnical University, Xi’an 710072, China; (W.Z.); (K.L.); (L.L.); (R.W.); (Y.L.)
- Dongguan Sanhang Innovation Institute, Dongguan 523808, China
| | - Yang Lei
- School of Life Sciences, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment in Special Environment, Northwestern Polytechnical University, Xi’an 710072, China; (W.Z.); (K.L.); (L.L.); (R.W.); (Y.L.)
- Dongguan Sanhang Innovation Institute, Dongguan 523808, China
| | - Hui Yang
- School of Life Sciences, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment in Special Environment, Northwestern Polytechnical University, Xi’an 710072, China; (W.Z.); (K.L.); (L.L.); (R.W.); (Y.L.)
| | - Leming Sun
- School of Life Sciences, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment in Special Environment, Northwestern Polytechnical University, Xi’an 710072, China; (W.Z.); (K.L.); (L.L.); (R.W.); (Y.L.)
- Dongguan Sanhang Innovation Institute, Dongguan 523808, China
| |
Collapse
|
48
|
Li H, Zhang J, Tan M, Yin Y, Song Y, Zhao Y, Yan L, Li N, Zhang X, Bai J, Jiang T, Li H. Exosomes based strategies for cardiovascular diseases: Opportunities and challenges. Biomaterials 2024; 308:122544. [PMID: 38579591 DOI: 10.1016/j.biomaterials.2024.122544] [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: 11/29/2023] [Revised: 03/11/2024] [Accepted: 03/19/2024] [Indexed: 04/07/2024]
Abstract
Exosomes, as nanoscale extracellular vesicles (EVs), are secreted by all types of cells to facilitate intercellular communication in living organisms. After being taken up by neighboring or distant cells, exosomes can alter the expression levels of target genes in recipient cells and thereby affect their pathophysiological outcomes depending on payloads encapsulated therein. The functions and mechanisms of exosomes in cardiovascular diseases have attracted much attention in recent years and are thought to have cardioprotective and regenerative potential. This review summarizes the biogenesis and molecular contents of exosomes and details the roles played by exosomes released from various cells in the progression and recovery of cardiovascular disease. The review also discusses the current status of traditional exosomes in cardiovascular tissue engineering and regenerative medicine, pointing out several limitations in their application. It emphasizes that some of the existing emerging industrial or bioengineering technologies are promising to compensate for these shortcomings, and the combined application of exosomes and biomaterials provides an opportunity for mutual enhancement of their performance. The integration of exosome-based cell-free diagnostic and therapeutic options will contribute to the further development of cardiovascular regenerative medicine.
Collapse
Affiliation(s)
- Hang Li
- Department of Cardiology, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, PR China
| | - Jun Zhang
- Department of Cardiology, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, PR China
| | - Mingyue Tan
- Department of Cardiology, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, PR China; Department of Geriatrics, Cardiovascular Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Yunfei Yin
- Department of Cardiology, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, PR China
| | - Yiyi Song
- Suzhou Medical College of Soochow University, Suzhou, Jiangsu, 215000, PR China
| | - Yongjian Zhao
- Department of Cardiology, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, PR China
| | - Lin Yan
- Department of Cardiology, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, PR China
| | - Ning Li
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230022, PR China
| | - Xianzuo Zhang
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230022, PR China
| | - Jiaxiang Bai
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230022, PR China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, PR China.
| | - Tingbo Jiang
- Department of Cardiology, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, PR China.
| | - Hongxia Li
- Department of Cardiology, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, PR China.
| |
Collapse
|
49
|
Imam RAEN, Aboulhoda BE, Amer MM, Hassan FE, Alghamdi MA, Abdel-Hamed MR. Role of mesenchymal stem cells-derived exosomes on inflammation, apoptosis, fibrosis and telocyte modulation in doxorubicin-induced cardiotoxicity: A closer look at the structural level. Microsc Res Tech 2024; 87:1598-1614. [PMID: 38441397 DOI: 10.1002/jemt.24544] [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: 10/19/2023] [Revised: 01/13/2024] [Accepted: 02/23/2024] [Indexed: 06/03/2024]
Abstract
Cardiotoxicity induced by doxorubicin (Dox) is a major complication in cancer patients. Exosomes (Ex) derived from mesenchymal cells could be a promising therapeutic for various heart diseases. This study investigated the role of Ex in Dox-induced cardiotoxicity and its mechanistic insights, using Sacubitril/valsartan (S/V) as a reference drug widely recommended in heart failure management. The study involved 24 Wistar rats, divided into a control, Dox, Dox + S/V, and Dox + Ex groups. The rats were assessed for cardiac enzymes, inflammatory and oxidative stress markers. Immunohistochemical expression of caspase-1, nuclear factor erythroid 2-related factor 2 (NrF2), E-Cadherin, CD117/c-kit, and Platelet-derived growth factor-α (PDGFα) was evaluated. P53 and Annexin V were assessed by PCR. Histological examination was performed using hematoxylin and eosin and Sirius red stains. Ex ameliorated the adverse cardiac pathological changes and significantly decreased the cardiac enzymes and inflammatory and oxidative stress markers. Ex also exerted antifibrotic and antiapoptotic effect in heart tissue. Ex treatment also improved NrF2 immunohistochemistry, up-regulated E-Cadherin immune expression, and restored the telocyte markers CD117/c-kit and PDGFα. Ex can mitigate Dox-induced cardiotoxicity by acting as an anti-inflammatory, antioxidant, antiapoptotic, and antifibrotic agents, restoring telocytes and modulating epithelial mesenchymal transition. RESEARCH HIGHLIGHTS: Exosomes exhibit positive expression for CD90 and CD105 whereas showing -ve expression for CD 34 by flow cytometry. Exosomes restore the immunohistochemical expression of the telocytes markers CD117/c-kit and PDGFα. Exosomes alleviate myocardial apoptosis, oxidative stress and fibrosis.
Collapse
Affiliation(s)
- Reda A El Nasser Imam
- Department of Anatomy and Embryology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Basma Emad Aboulhoda
- Department of Anatomy and Embryology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Maha M Amer
- Department of Anatomy and Embryology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Fatma E Hassan
- Medical Physiology Department, Kasr Alainy, Faculty of Medicine, Cairo University, Giza, Egypt
- General Medicine Practice Program, Department of Physiology, Batterjee Medical College, Jeddah, Saudi Arabia
| | - Mansour A Alghamdi
- College of Medicine, King Khalid University, Abha, Saudi Arabia
- Genomics and Personalized Medicine Unit, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Mohamed R Abdel-Hamed
- Department of Anatomy and Embryology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
50
|
Wang Y, Shi X. The potential mechanisms and treatment effects of stem cell-derived exosomes in cardiac reengineering. NANOTECHNOLOGY 2024; 35:362005. [PMID: 38834043 DOI: 10.1088/1361-6528/ad53d1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 06/04/2024] [Indexed: 06/06/2024]
Abstract
Exosomes are extracellular vesicles of diverse compositions that are secreted by numerous cell types. Exosomes contain significant bioactive components, including lipids, proteins, mRNA, and miRNA. Exosomes play an important role in regulating cellular signaling and trafficking under both normal physiological and pathological circumstances. A multitude of factors, including thermal stress, ribosomal stress, endoplasmic reticulum stress, and oxidative stress influence the concentrations of exosomal mRNA, miRNA, proteins, and lipids. It has been stated that exosomes derived from stem cells (SCs) modulate a range of stresses by preventing or fostering cell balance. Exosomes derived from SCs facilitate recovery by facilitating cross-cellular communication via the transmission of information in the form of proteins, lipids, and other components. For this reason, exosomes are used as biomarkers to diagnose a wide variety of diseases. The focus of this review is the bioengineering of artificial exosomal cargoes. This process encompasses the control and transportation of particular exosomal cargoes, including but not limited to small molecules, recombinant proteins, immune modulators, and therapeutic medications. Therapeutic approaches of this nature have the potential to deliver therapeutic medications precisely to the intended site for the cure of a variety of disorders. Notably, our attention has been directed towards the therapeutic implementations of exosomes derived from SCs in the cure of cardiovascular ailments, including but not limited to ischemic heart disease, myocardial infarction, sepsis, heart failure, cardiomyopathy, and cardiac fibrosis. In general, researchers employ two methodologies when it comes to exosomal bioengineering. This review aims to explain the function of exosomes derived from SCs in the regulation of stress and present a novel therapeutic approach for cardiovascular disorders.
Collapse
Affiliation(s)
- Yibin Wang
- Department of Cardiology, Hangzhou Ninth People's Hospital, Hangzhou 311225, People's Republic of China
| | - Xiulian Shi
- Emergency Department, Chun'an First People's Hospital, Hangzhou 311700, People's Republic of China
| |
Collapse
|