1
|
Bai Q, Wang C, Ding N, Wang Z, Liu R, Li L, Piao H, Song Y, Yan G. Eupalinolide B targets DEK and PANoptosis through E3 ubiquitin ligases RNF149 and RNF170 to negatively regulate asthma. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156657. [PMID: 40120540 DOI: 10.1016/j.phymed.2025.156657] [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: 01/16/2025] [Revised: 02/27/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025]
Abstract
PURPOSE We investigated the mechanism by which eupalinolide B (EB) regulates DEK protein ubiquitination and degradation, and its impact on DEK-mediated receptor-interacting protein kinase 1 (RIPK)-PANoptosis pathway in allergic asthma. STUDY DESIGN AND METHODS In vitro studies were conducted on human bronchial epithelial cells (BEAS-2B) treated with EB and human-recombinant DEK. Mass spectrometry analysis, RNA sequencing, molecular docking, and functional assays were used to assess the interactions and effects of EB, DEK, and ring finger protein 149 and 170 (RNF149 and RNF170). In vivo experiments involved a house dust mite-induced asthma model in mice and evaluation of airway inflammation, DEK expression, and PANoptosis markers. RESULTS In vitro, EB could bind to DEK. RNF149 and RNF170 were identified as regulatory factors of DEK, polyubiquitinating the K349 site in the DEK coding DNA sequence region 270-350 through K48 linkages and leading to its degradation. RNA sequencing showed that DEK overexpression upregulated the expression of genes such as RIPK1, FADD, and Caspase 8. Treatment with DEK siRNA or EB reduced the activation of the RIPK1-PANoptosis pathway in BEAS-2B-DEK cells. In vivo, EB significantly reduced the levels of DEK in house dust mite-induced mice and alleviated pulmonary inflammatory cell infiltration, goblet cell hyperplasia, collagen fiber deposition, and eosinophil proportion in BALF. Knocking out the DEK gene reduced RIPK1-induced PANoptosis, and inhibited airway inflammation and cell apoptosis. CONCLUSION EB promotes the degradation of DEK by RNF149 and RNF170, inhibits the RIPK1-PANoptosis pathway, and may effectively suppress asthma. EB may become a potential drug for treating airway inflammation in asthma.
Collapse
Affiliation(s)
- Qiaoyun Bai
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji 133002, PR China; Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji 133002, PR China
| | - Chongyang Wang
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji 133002, PR China; Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji 133002, PR China
| | - Ningpo Ding
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji 133002, PR China; Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji 133002, PR China
| | - Zhiguang Wang
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji 133002, PR China; Department of Respiratory Medicine, Affiliated Hospital of Yanbian University, Yanji 133000, PR China
| | - Ruobai Liu
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji 133002, PR China; Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji 133002, PR China
| | - Liangchang Li
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji 133002, PR China; Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji 133002, PR China
| | - Hongmei Piao
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji 133002, PR China; Department of Respiratory Medicine, Affiliated Hospital of Yanbian University, Yanji 133000, PR China
| | - Yilan Song
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji 133002, PR China; Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji 133002, PR China; Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji 133002, PR China.
| | - Guanghai Yan
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji 133002, PR China; Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji 133002, PR China; Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji 133002, PR China.
| |
Collapse
|
2
|
Duan H, Siadat SH, Jasim SA, Bansal P, Kaur H, Qasim MT, Abosaoda MK, Aboqader Al-Aouadi RF, Suliman M, Ali Khiavi P. Therapeutic Potential of Exosomal miRNAs: New Insights and Future Directions. J Biochem Mol Toxicol 2025; 39:e70270. [PMID: 40272032 DOI: 10.1002/jbt.70270] [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: 12/24/2024] [Revised: 03/13/2025] [Accepted: 04/10/2025] [Indexed: 04/25/2025]
Abstract
Modern advancements in medicine include developing targeted drug delivery systems in the medical field, which are designed to unravel the potential of therapeutic products and overcome the barriers to the effectiveness of current approaches. Various nanopolymer carrier systems have been introduced in this regard, and the simple characteristics of extracellular vesicles have drawn special attention to their application as an effective drug delivery tool. Exosomes are very similar to transport vesicles and have a lipid-biomembrane covering an aqueous core. They also contain both hydrophilic and lipophilic substances and deliver their cargo to the desired targets. These properties enable exosomes to overcome some of the limitations of liposomes. Exosomes can easily diffuse into body fluids and remain in the bloodstream for a long time, crossing physiological barriers and entering cells. Exosomes, which contain a large volume of biomolecules, do not stimulate immune responses and do not accumulate in the liver or lungs instead of target tissues. Recent advancements in regenerative medicine have enabled scientists to utilize exosomes extracted from mesenchymal stem cells (MSCs), which possess significant regenerative abilities, for treating various diseases. The contents of these exosomes are crucial for both diagnosis and treatment, as they influence disease progression. Numerous in vitro studies have confirmed the safety, effectiveness, and therapeutic promise of exosomes in conditions such as cancer, neurodegenerative disorders, cardiovascular issues, and orthopedic ailments. This article explores the therapeutic potential of MSC-derived exosomes and outlines the essential procedures for their preparation.
Collapse
Affiliation(s)
- Haili Duan
- China Three Gorges University, Yichang City, China
| | | | - Saade Abdalkareem Jasim
- Medical Laboratory Techniques department, College of Health and medical technology, University of Al-maarif, Anbar, Iraq
| | - Pooja Bansal
- Department of Biotechnology and Genetics, Jain (Deemed-to-be) University, Bengaluru, India
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, India
| | - Harpreet Kaur
- School of Basic & Applied Sciences, Shobhit University, Gangoh, India
- Department of Health & Allied Sciences, Arka Jain University, Jamshedpur, India
| | - Maytham T Qasim
- Immunology and Physiology, College of Health and Medical Technology, Al-Ayen University, Iraq
| | - Munther Kadhim Abosaoda
- College of Pharmacy, The Islamic University, Najaf, Iraq
- College of Pharmacy, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- College of Pharmacy, The Islamic University of Babylon, Al Diwaniyah, Iraq
| | | | - Muath Suliman
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Payam Ali Khiavi
- Medicine Faculty, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
3
|
Zhao M, Huang Z, Zheng J, Li W, Zhong Y, Ouyang T. MiR-340-5p alleviates AECOPD by targeting MAP3K2 via Qingjin Huatan decoction therapy. J Leukoc Biol 2025; 117:qiaf021. [PMID: 39973067 DOI: 10.1093/jleuko/qiaf021] [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: 10/08/2024] [Revised: 12/27/2024] [Accepted: 02/19/2025] [Indexed: 02/21/2025] Open
Abstract
Chronic obstructive pulmonary disease (COPD) features persistent inflammation and restricted airflow, with acute exacerbations of COPD (AECOPD) significantly worsening patient outcomes. This study aims to explore the role of Qingjin Huatan Decoction (QJHTT) on AECOPD with the syndrome of phlegm-heat obstruction of the lung. AECOPD was induced in male Sprague-Dawley rats using lipopolysaccharide and cigarette smoke exposure. Rats were treated with varying doses of QJHTT. miR-340-5p expression was quantified using qPCR. Lung histopathology was assessed with hematoxylin and eosin staining, and interleukin-6, interleukin-1 beta, and tumor necrosis factor-alpha were measured by enzyme-linked immunosorbent assay (ELISA). The effects on cell viability and apoptosis in primary airway epithelial cells were evaluated using Cell Counting Kit-8 and flow cytometry assays, respectively. The dual-luciferase reporter assay validated the interaction between miR-340-5p and mitogen-activated protein kinase kinase kinase 2 (MAP3K2), and protein expression was analyzed by Western blot. QJHTT improved lung histopathology, reducing inflammatory cell infiltration, and alveolar damage. ELISA results showed reduced inflammatory cytokine levels in QJHTT-treated groups (P < 0.05). qPCR analysis demonstrated that QJHTT upregulated miR-340-5p expression (P < 0.05). miR-340-5p mimic enhanced cell viability and reduced apoptosis in primary airway epithelial cells (P < 0.05). Dual-luciferase reporter assay confirmed that miR-340-5p directly targets MAP3K2, leading to its downregulation (P < 0.05). QJHTT exerts therapeutic effects in phlegm-heat obstructing the lung type of AECOPD through upregulating miR-340-5p and inhibiting MAP3K2. This study highlights the QJHTT and miR-340-5p/MAP3K2 pathway for this disease treatment.
Collapse
Affiliation(s)
- Mei Zhao
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi University of Chinese Medicine, No. 89-9 Dongge Road, Nanning 530022, Guangxi Zhuang Autonomous Region, P. R. China
| | - Zhijian Huang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi University of Chinese Medicine, No. 89-9 Dongge Road, Nanning 530022, Guangxi Zhuang Autonomous Region, P. R. China
| | - Jinghui Zheng
- Department of Integrated Chinese and Western Medicine Clinical Prevention and Treatment of Cardiovascular and Endocrine Diseases, Guangxi University of Chinese Medicine, No. 13, Wuhe Aveneue, Qingxiu District Nanning 530000, Guangxi Zhuang Autonomous Region, P. R. China
| | - Wanying Li
- Department of Integrated Chinese and Western Medicine Clinical Prevention and Treatment of Cardiovascular and Endocrine Diseases, Guangxi University of Chinese Medicine, No. 13, Wuhe Aveneue, Qingxiu District Nanning 530000, Guangxi Zhuang Autonomous Region, P. R. China
| | - Yunqing Zhong
- Department of Pulmonary Disease, Guangxi International Zhuang Medicine Hospital Affiliated to Guangxi University of Chinese Medicine, No. 8 Qiyue Road, Wuxiang New District, Nanning 530001, Guangxi Zhuang Autonomous Region, P. R. China
| | - Tun Ouyang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi University of Chinese Medicine, No. 89-9 Dongge Road, Nanning 530022, Guangxi Zhuang Autonomous Region, P. R. China
| |
Collapse
|
4
|
Li S, Zou M, Wang Y, Guo Q, Lv S, Zhao W, Kabir MA, Peng X. Matrix metalloproteinase 7 (MMP7) as a molecular target for Mycoplasma gallisepticum (MG) resistance in chickens. Int J Biol Macromol 2025; 298:140110. [PMID: 39842573 DOI: 10.1016/j.ijbiomac.2025.140110] [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: 11/26/2024] [Revised: 01/12/2025] [Accepted: 01/18/2025] [Indexed: 01/24/2025]
Abstract
Mycoplasma gallisepticum (MG) causes chronic respiratory disease (CRD), posing a significant threat to global poultry production. Current preventive strategies face limitations, emphasizing the need for alternative approaches such as breeding for disease resistance. This study identifies the matrix metalloproteinase 7 (MMP7) gene as a key factor in CRD resistance. Analysis of high-throughput sequencing data revealed MMP7's association with MG infection at tissue and cellular levels. Overexpression of MMP7 in avian type II alveolar epithelial cells (AECII) and macrophages (HD11) inhibited MG adhesion, modulated immune responses, and suppressed MG-induced cell proliferation and apoptosis, though MG replication remained unaffected. Conversely, MMP7 inhibition enhanced MG infection. Experimental infections in commercial (Jingfen Layer No.6, Hy-Line White) and local Chinese chicken breeds (Guangxi Indigenous, Tianlu Partridge, Cyan Shank Partridge) validated Tianlu Partridge chickens' relative resistance and Jingfen Layers' susceptibility. MMP7 expression levels correlated positively with reduced chick weight, air sac damage, tracheal mucosal thickness, and MG lung loads. These findings highlight MMP7 as a molecular target for assessing MG susceptibility and breeding resistant chickens while demonstrating the utility of local Chinese breeds in resistance-focused breeding programs.
Collapse
Affiliation(s)
- Shiying Li
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education; College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Mengyun Zou
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education; College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; Institute of animal husbandry and veterinary medicine, Jiangxi Academy of Agricultural Sciences, Jiangxi poultry Engineering Technology Research Center, Jiangxi poultry breeding Engineering Laboratory, Nanchang, Jiangxi, China
| | - Yingjie Wang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education; College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan Province 611130, China
| | - Qiao Guo
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education; College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Shan Lv
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education; College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Wenqing Zhao
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education; College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Md Ahsanul Kabir
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education; College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Xiuli Peng
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education; College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
5
|
Su X, Zhu L, Zhuo J, Zhang S. Diagnostic value of miR-193a-5p in severe pneumonia and its correlation with prognosis. J Cardiothorac Surg 2025; 20:87. [PMID: 39849492 PMCID: PMC11756221 DOI: 10.1186/s13019-024-03256-y] [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: 09/13/2024] [Accepted: 12/24/2024] [Indexed: 01/25/2025] Open
Abstract
BACKGROUND Severe pneumonia is a common disease in children, with rapid progression and easy complications of respiratory failure, endangering the lives of children. This study aimed to elucidate the clinical significance of miR-193a-5p in severe pneumonia and to provide a new biomarker for the disease. METHODS A total of 150 children with severe pneumonia and an equal number of healthy children were selected for analysis. Serum miR-193a-5p levels were detected by RT-qPCR. The correlation of miR-193a-5p with CRP, WBC, neutrophil count, and NLR was assessed by Spearman analysis. The diagnostic and prognostic value of miR-193a-5p in severe pneumonia was analyzed using ROC curves. The relationship between miR-193a-5p and the prognosis of severe pneumonia was evaluated using the Kaplan-Meier curve and a multivariate logistic analysis. RESULTS Serum levels of miR-193a-5p were markedly elevated in children with severe pneumonia and exhibited a positive correlation with CRP, WBC, neutrophil count, and NLR. miR-193a-5p could effectively distinguish children with severe pneumonia from healthy children, with an AUC, sensitivity, and specificity of 0.862, 70.67%, and 88.67%, respectively. Serum miR-193a-5p expression was increased in children with poor prognosis and had a predictive value for patient prognosis. High expression of miR-193a-5p was linked to survival in children with severe pneumonia and was a risk factor for adverse prognosis. CONCLUSION Serum levels of miR-193a-5p were markedly elevated in children with severe pneumonia, which may be of significance for the early diagnosis of the disease and prognostic assessment.
Collapse
Affiliation(s)
- Xiaoji Su
- Clinical Laboratory, Anhui Hospital, Children's Hospital of Fudan University, No. 39, Wangjiang East Road, Hefei, Anhui, 230011, China
| | - Lijuan Zhu
- Clinical Laboratory, Anhui Hospital, Children's Hospital of Fudan University, No. 39, Wangjiang East Road, Hefei, Anhui, 230011, China
| | - Jiajia Zhuo
- Clinical Laboratory, Anhui Hospital, Children's Hospital of Fudan University, No. 39, Wangjiang East Road, Hefei, Anhui, 230011, China
| | - Shihai Zhang
- Clinical Laboratory, Anhui Hospital, Children's Hospital of Fudan University, No. 39, Wangjiang East Road, Hefei, Anhui, 230011, China.
| |
Collapse
|
6
|
Deng L, Liu Y, Wu Q, Lai S, Yang Q, Mu Y, Dong M. Exosomes to exosome-functionalized scaffolds: a novel approach to stimulate bone regeneration. Stem Cell Res Ther 2024; 15:407. [PMID: 39521993 PMCID: PMC11550564 DOI: 10.1186/s13287-024-04024-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Bone regeneration is a complex biological process that relies on the orchestrated interplay of various cellular and molecular events. Bone tissue engineering is currently the most promising method for treating bone regeneration. However, the immunogenicity, stable and cell quantity of seed cells limited their application. Recently, exosomes, which are small extracellular vesicles released by cells, have been found to effectively address these problems and better induce bone regeneration. Meanwhile, a growing line of research has shown the cargos of exosomes may provide effective therapeutic and biomarker tools for bone repair, including miRNA, lncRNA, and proteins. Moreover, engineered scaffolds loaded with exosomes can offer a cell-free bone repair strategy, addressing immunogenicity concerns and providing a more stable functional performance. Herein, we provide a comprehensive summary of the role played by scaffolds loaded with exosomes in bone regeneration, drawing on a systematic analysis of relevant literature available on PubMed, Scopus, and Google Scholar database.
Collapse
Affiliation(s)
- Li Deng
- Center for Medicine Research and Translation, Chengdu Fifth People's Hospital (The Second Clinical Medical College, Affiliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, 611135, Sichuan, China
| | - Yang Liu
- Center for Medicine Research and Translation, Chengdu Fifth People's Hospital (The Second Clinical Medical College, Affiliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, 611135, Sichuan, China
| | - Qian Wu
- Center for Medicine Research and Translation, Chengdu Fifth People's Hospital (The Second Clinical Medical College, Affiliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, 611135, Sichuan, China
| | - Shuang Lai
- Stomatology Department, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Qiu Yang
- Center for Medicine Research and Translation, Chengdu Fifth People's Hospital (The Second Clinical Medical College, Affiliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, 611135, Sichuan, China
| | - Yandong Mu
- Stomatology Department, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, China.
| | - Mingqing Dong
- Center for Medicine Research and Translation, Chengdu Fifth People's Hospital (The Second Clinical Medical College, Affiliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, 611135, Sichuan, China.
| |
Collapse
|
7
|
Lv J, Xiong X. Extracellular Vesicle microRNA: A Promising Biomarker and Therapeutic Target for Respiratory Diseases. Int J Mol Sci 2024; 25:9147. [PMID: 39273095 PMCID: PMC11395461 DOI: 10.3390/ijms25179147] [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: 06/29/2024] [Revised: 08/14/2024] [Accepted: 08/19/2024] [Indexed: 09/15/2024] Open
Abstract
Respiratory diseases, including chronic obstructive pulmonary disease (COPD), asthma, lung cancer, and coronavirus pneumonia, present a major global health challenge. Current diagnostic and therapeutic options for these diseases are limited, necessitating the urgent development of novel biomarkers and therapeutic strategies. In recent years, microRNAs (miRNAs) within extracellular vesicles (EVs) have received considerable attention due to their crucial role in intercellular communication and disease progression. EVs are membrane-bound structures released by cells into the extracellular environment, encapsulating a variety of biomolecules such as DNA, RNA, lipids, and proteins. Specifically, miRNAs within EVs, known as EV-miRNAs, facilitate intercellular communication by regulating gene expression. The expression levels of these miRNAs can reflect distinct disease states and significantly influence immune cell function, chronic airway inflammation, airway remodeling, cell proliferation, angiogenesis, epithelial-mesenchymal transition, and other pathological processes. Consequently, EV-miRNAs have a profound impact on the onset, progression, and therapeutic responses of respiratory diseases, with great potential for disease management. Synthesizing the current understanding of EV-miRNAs in respiratory diseases such as COPD, asthma, lung cancer, and novel coronavirus pneumonia, this review aims to explore the potential of EV-miRNAs as biomarkers and therapeutic targets and examine their prospects in the diagnosis and treatment of these respiratory diseases.
Collapse
Affiliation(s)
- Jiaxi Lv
- Department of Pulmonary and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Xianzhi Xiong
- Department of Pulmonary and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| |
Collapse
|
8
|
Wang Y, Li S, Wang T, Zou M, Peng X. Extracellular Vesicles From Mycoplasma gallisepticum: Modulators of Macrophage Activation and Virulence. J Infect Dis 2024; 229:1523-1534. [PMID: 37929888 DOI: 10.1093/infdis/jiad486] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 10/21/2023] [Accepted: 10/30/2023] [Indexed: 11/07/2023] Open
Abstract
Extracellular vesicles (EVs) mediate intercellular communication by transporting proteins. To investigate the pathogenesis of Mycoplasma gallisepticum, a major threat to the poultry industry, we isolated and characterized M. gallisepticum-produced EVs. Our study highlights the significant impact of M. gallisepticum-derived EVs on immune function and macrophage apoptosis, setting them apart from other M. gallisepticum metabolites. These EVs dose-dependently enhance M. gallisepticum adhesion and proliferation, simultaneously modulating Toll-like receptor 2 and interferon γ pathways and thereby inhibiting macrophage activation. A comprehensive protein analysis revealed 117 proteins in M. gallisepticum-derived EVs, including established virulence factors, such as GapA, CrmA, VlhA, and CrmB. Crucially, these EV-associated proteins significantly contribute to M. gallisepticum infection. Our findings advance our comprehension of M. gallisepticum pathogenesis, offering insights for preventive strategies and emphasizing the pivotal role of M. gallisepticum-derived EVs and their associated proteins. This research sheds light on the composition and crucial role of M. gallisepticum-derived EVs in M. gallisepticum pathogenesis, aiding our fight against M. gallisepticum infections.
Collapse
Affiliation(s)
- Yingjie Wang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Shiying Li
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Tengfei Wang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Mengyun Zou
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xiuli Peng
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
9
|
Feng Y, Guo K, Jiang J, Lin S. Mesenchymal stem cell-derived exosomes as delivery vehicles for non-coding RNAs in lung diseases. Biomed Pharmacother 2024; 170:116008. [PMID: 38071800 DOI: 10.1016/j.biopha.2023.116008] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 12/02/2023] [Accepted: 12/06/2023] [Indexed: 01/10/2024] Open
Abstract
The burden of lung diseases is gradually increasing with an increase in the average human life expectancy. Therefore, it is necessary to identify effective methods to treat lung diseases and reduce their social burden. Currently, an increasing number of studies focus on the role of mesenchymal stem cell-derived exosomes (MSC-Exos) as a cell-free therapy in lung diseases. They show great potential for application to lung diseases as a more stable and safer option than traditional cell therapies. MSC-Exos are rich in various substances, including proteins, nucleic acids, and DNA. Delivery of Non-coding RNAs (ncRNAs) enables MSC-Exos to communicate with target cells. MSC-Exos significantly inhibit inflammatory factors, reduce oxidative stress, promote normal lung cell proliferation, and reduce apoptosis by delivering ncRNAs. Moreover, MSC-Exos carrying specific ncRNAs affect the proliferation, invasion, and migration of lung cancer cells, thereby playing a role in managing lung cancer. The detailed mechanisms of MSC-Exos in the clinical treatment of lung disease were explored by developing standardized culture, isolation, purification, and administration strategies. In summary, MSC-Exo-based delivery methods have important application prospects for treating lung diseases.
Collapse
Affiliation(s)
- Yuqian Feng
- Hangzhou School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Kaibo Guo
- Department of Oncology, Hangzhou First People's Hospital, Hangzhou, Zhejiang 310003, China
| | - Jing Jiang
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Shengyou Lin
- Department of Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang 310006, China.
| |
Collapse
|