1
|
Park KS, Lässer C, Lötvall J. Extracellular vesicles and the lung: from disease pathogenesis to biomarkers and treatments. Physiol Rev 2025; 105:1733-1821. [PMID: 40125970 DOI: 10.1152/physrev.00032.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 10/14/2024] [Accepted: 03/12/2025] [Indexed: 03/25/2025] Open
Abstract
Nanosized extracellular vesicles (EVs) are released by all cells to convey cell-to-cell communication. EVs, including exosomes and microvesicles, carry an array of bioactive molecules, such as proteins and RNAs, encapsulated by a membrane lipid bilayer. Epithelial cells, endothelial cells, and various immune cells in the lung contribute to the pool of EVs in the lung microenvironment and carry molecules reflecting their cellular origin. EVs can maintain lung health by regulating immune responses, inducing tissue repair, and maintaining lung homeostasis. They can be detected in lung tissues and biofluids such as bronchoalveolar lavage fluid and blood, offering information about disease processes, and can function as disease biomarkers. Here, we discuss the role of EVs in lung homeostasis and pulmonary diseases such as asthma, chronic obstructive pulmonary disease, cystic fibrosis, pulmonary fibrosis, and lung injury. The mechanistic involvement of EVs in pathogenesis and their potential as disease biomarkers are discussed. Finally, the pulmonary field benefits from EVs as clinical therapeutics in severe pulmonary inflammatory disease, as EVs from mesenchymal stem cells attenuate severe respiratory inflammation in multiple clinical trials. Further, EVs can be engineered to carry therapeutic molecules for enhanced and broadened therapeutic opportunities, such as the anti-inflammatory molecule CD24. Finally, we discuss the emerging opportunity of using different types of EVs for treating severe respiratory conditions.
Collapse
Affiliation(s)
- Kyong-Su Park
- Krefting Research Centre, Institute of Medicine at the Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| | - Cecilia Lässer
- Krefting Research Centre, Institute of Medicine at the Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| | - Jan Lötvall
- Krefting Research Centre, Institute of Medicine at the Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| |
Collapse
|
2
|
Zhang X, Fan H, Su L, Wang Y, Chen G. Mdivi-1 Attenuates Sepsis-Associated Acute Lung Injury by Inhibiting M1 Alveolar Macrophage Polarization and Pyroptosis. Mediators Inflamm 2025; 2025:3675276. [PMID: 40196168 PMCID: PMC11972854 DOI: 10.1155/mi/3675276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 01/21/2025] [Indexed: 04/09/2025] Open
Abstract
Background: Dynamin-related protein 1 (DRP1)-dependent mitochondrial fission is a novel target for mitigating inflammatory diseases. This study aims to explore the effects of the DRP1 inhibitor Mdivi-1 on sepsis-induced acute lung injury (ALI). Methods: C57BL/6 mice were intraperitoneally injected with lipopolysaccharide (LPS) and then treated with or without Mdivi-1 2 h post-injection. RAW264.7 alveolar macrophages were stimulated with LPS and treated with or without NLRP3 inhibitors, Mito-TEMPO, or Mdivi-1. Hematoxylin and eosin (H&E) staining was used to observe pathological changes in lung tissues. The levels of inflammatory cytokines in lung tissue homogenates, serum, and cell culture medium were detected using enzyme-linked immunosorbent assays (ELISA). The mRNA expression of macrophage polarization markers, NLRP3 activation, and phosphorylation status of DRP1 were assessed. Flow cytometry was employed to evaluate the levels of macrophage apoptosis. Immunofluorescence was utilized to detect the levels of in vivo and in vitro macrophage polarization markers. Mitochondrial reactive oxygen species (Mito-ROS) were measured using a Mito-SOX assay kit. Results: Our results suggested that Mdivi-1 reduced lung tissue pathological injury, M1 alveolar macrophage polarization, NLRP3 activation, and DRP1 Ser616 phosphorylation. In vitro, LPS triggered abnormal accumulation of M1 polarization, NLRP3 activation, and excessive increase in Mito-ROS. NLRP3 inhibitors and Mito-TEMPO inhibited M1 alveolar macrophage polarization and pyroptosis-mediated tissue damage. Mito-TEMPO significantly inhibited NLRP3 activation. Furthermore, Mdivi-1 reduced ALI by inhibiting M1 polarization and pyroptosis. The mechanism of Mdivi-1 in reducing M1 alveolar macrophage polarization and pyroptosis may be related to the inhibition of DRP1-mediated mitochondrial fission, thus suppressing the Mito-ROS/NLRP3 pathway. Similar results were observed in vitro by knocking down DRP1. Conclusion: Inhibition of DRP1 by Mdivi-1 alleviates ALI by hindering Mito-ROS/NLRP3-mediated M1 alveolar macrophage polarization and pyroptosis, suggesting that DRP1-dependent mitochondrial fission is a potential therapeutic target for ALI.
Collapse
Affiliation(s)
- Xiaoyu Zhang
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Hui Fan
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Li Su
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Yanni Wang
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Guozhong Chen
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| |
Collapse
|
3
|
Yang X, Sun Z, Liu Z, Chen H, Fang Y, Tao W, Zhao N, Ouyang X, Liu F, Qian K. From Gene to Intervention: NLRC4 and WIPI1 Regulate Septic Acute Lung Injury Through Autophagy. J Inflamm Res 2025; 18:3639-3656. [PMID: 40093959 PMCID: PMC11910914 DOI: 10.2147/jir.s510691] [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: 12/06/2024] [Accepted: 03/04/2025] [Indexed: 03/19/2025] Open
Abstract
Background Septic Acute Lung Injury (SALI)-induced severe respiratory dysfunction has been established to significantly increase patient mortality rates and socioeconomic costs. To mitigate cellular damage, autophagy -a conserved biological process in organisms -degrades damaged cellular components, such as proteins and organelles. Although autophagy is crucially involved in the inflammatory response, its precise molecular mechanisms in SALI remain unclear, forming the basis of this study. Methods Herein, two microarray datasets (GSE33118 and GSE131761) and three single-cell sequencing datasets (SCP43, SCP548, and SCP2156) derived from human samples were used to ascertain the interrelationship between Differentially Expressed Autophagy-Related Genes (DEARGs) and SALI. The relationship between key DEARGs and SALI was validated both in vitro and in vivo using various techniques, including flow cytometry, Immunofluorescence (IF), Quantitative Polymerase Chain Reaction (qPCR), Western Blotting (WB), and small interfering RNA (siRNA). Results Herein, we found that autophagy activation attenuated SALI, with NLRC4 and WIPI1 as the two key DEARGs involved. Specifically, NLRC4 and WIPI1 downregulation mitigated SALI via autophagy activation. Compared to NLRC4, WIPI1 was more closely associated with noncanonical autophagic flux in SALI. Furthermore, immune infiltration analysis and single-cell data showed a close relationship between NLRC4, WIPI1, and immune cells. Conclusion Our findings revealed that SALI correlated strongly with autophagy, with the downregulation of the two key DEARGs, NLRC4 and WIPI1, attenuating sepsis lung injury via autophagy regulation, highlighting their therapeutic significance in SALI.
Collapse
Affiliation(s)
- Xinyi Yang
- Department of Critical Care Medicine, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People's Republic of China
- Medical Innovation Center, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People's Republic of China
| | - Zhijian Sun
- Department of Critical Care Medicine, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People's Republic of China
- Medical Innovation Center, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People's Republic of China
| | - Zhuohui Liu
- Department of Critical Care Medicine, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People's Republic of China
- Medical Innovation Center, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People's Republic of China
| | - Hui Chen
- Department of Critical Care Medicine, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People's Republic of China
- Medical Innovation Center, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People's Republic of China
| | - Yang Fang
- Department of Critical Care Medicine, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People's Republic of China
- Medical Innovation Center, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People's Republic of China
| | - Wenqiang Tao
- Department of Critical Care Medicine, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People's Republic of China
- Medical Innovation Center, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People's Republic of China
| | - Ning Zhao
- Department of Critical Care Medicine, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People's Republic of China
- Medical Innovation Center, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People's Republic of China
| | - Xiufang Ouyang
- Department of Critical Care Medicine, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People's Republic of China
- Medical Innovation Center, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People's Republic of China
| | - Fen Liu
- Department of Critical Care Medicine, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People's Republic of China
- Jiangxi Medical Center for Critical Public Health Events, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330052, People's Republic of China
| | - Kejian Qian
- Department of Critical Care Medicine, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People's Republic of China
| |
Collapse
|
4
|
Ali-Khiavi P, Mohammadi M, Masoumi S, Saffarfar H, Kheradmand R, Mobed A, Hatefnia F. The Therapeutic Potential of Exosome Therapy in Sepsis Management: Addressing Complications and Improving Outcomes". Cell Biochem Biophys 2025; 83:307-326. [PMID: 39363035 DOI: 10.1007/s12013-024-01564-7] [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] [Accepted: 09/17/2024] [Indexed: 10/05/2024]
Abstract
Infection occurs when pathogens penetrate tissues, reproduce, and trigger a host response to both the infectious agents and their toxins. A diverse array of pathogens, including viruses and bacteria, can cause infections. The host's immune system employs several mechanisms to combat these infections, typically involving an innate inflammatory response. Inflammation is a complex biological reaction that can affect various parts of the body and is a key component of the response to harmful stimuli. Sepsis arises when the body's response to infection leads to widespread damage to tissues and organs, potentially resulting in severe outcomes or death. The initial phase of sepsis involves immune system suppression. Early identification and targeted management are crucial for improving sepsis outcomes. Common treatment approaches include antibiotics, intravenous fluids, blood cultures, and monitoring urine output. This study explores the potential of exosome therapy in enhancing the management and alleviation of sepsis symptoms.
Collapse
Affiliation(s)
- Payam Ali-Khiavi
- Medical faculty, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahya Mohammadi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sajjad Masoumi
- Department of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Hossein Saffarfar
- Cardiovascular Research Center, Tehran, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Kheradmand
- Social Determinants of Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahmad Mobed
- Social Determinants of Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Faezeh Hatefnia
- Social Determinants of Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
5
|
You B, Yang Y, Wei J, Zhou C, Dong S. Pathogenic and therapeutic roles of extracellular vesicles in sepsis. Front Immunol 2025; 16:1535427. [PMID: 39967672 PMCID: PMC11832720 DOI: 10.3389/fimmu.2025.1535427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 01/17/2025] [Indexed: 02/20/2025] Open
Abstract
Sepsis is a systemic injury resulting in vascular dysfunction, which can lead to multiple organ dysfunction, even shock and death. Extracellular vesicles (EVs) released by mammalian cells and bacteria have been shown to play important roles in intercellular communication and progression of various diseases. In past decades, the functional role of EVs in sepsis and its complications has been well explored. EVs are one of the paracrine components of cells. By delivering bioactive materials, EVs can promote immune responses, particularly the development of inflammation. In addition, EVs can serve as beneficial tools for delivering therapeutic cargos. In this review, we discuss the dual role of EVs in the progression and treatment of sepsis, exploring their intricate involvement in both inflammation and tissue repair processes. Specifically, the remarkable role of engineered strategies based on EVs in the treatment of sepsis is highlighted. The engineering EVs-mediated drug delivery and release strategies offer broad prospects for the effective treatment of sepsis. EVs-based approaches provide a novel avenue for diagnosing sepsis and offer opportunities for more precise intervention.
Collapse
Affiliation(s)
- Benshuai You
- Clinical Laboratory Center, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou, Jiangsu, China
| | - Yang Yang
- Clinical Laboratory Center, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou, Jiangsu, China
| | - Jing Wei
- Department of Obstetrics and Gynecology, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou, Jiangsu, China
| | - Chenglin Zhou
- Clinical Laboratory Center, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou, Jiangsu, China
| | - Surong Dong
- Clinical Laboratory Center, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou, Jiangsu, China
| |
Collapse
|
6
|
Shirani N, Abdi N, Chehelgerdi M, Yaghoobi H, Chehelgerdi M. Investigating the role of exosomal long non-coding RNAs in drug resistance within female reproductive system cancers. Front Cell Dev Biol 2025; 13:1485422. [PMID: 39925739 PMCID: PMC11802832 DOI: 10.3389/fcell.2025.1485422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 01/02/2025] [Indexed: 02/11/2025] Open
Abstract
Exosomes, as key mediators of intercellular communication, have been increasingly recognized for their role in the oncogenic processes, particularly in facilitating drug resistance. This article delves into the emerging evidence linking exosomal lncRNAs to the modulation of drug resistance mechanisms in cancers such as ovarian, cervical, and endometrial cancer. It synthesizes current research findings on how these lncRNAs influence cancer cell survival, tumor microenvironment, and chemotherapy efficacy. Additionally, the review highlights potential therapeutic strategies targeting exosomal lncRNAs, proposing a new frontier in overcoming drug resistance. By mapping the interface of exosomal lncRNAs and drug resistance, this article aims to provide a comprehensive understanding that could pave the way for innovative treatments and improved patient outcomes in female reproductive system cancers.
Collapse
Affiliation(s)
- Nooshafarin Shirani
- Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Neda Abdi
- Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Matin Chehelgerdi
- Novin Genome (NG) Lab, Research and Development Center for Biotechnology, Shahrekord, Iran
- Young Researchers and Elite Club, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Hajar Yaghoobi
- Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mohammad Chehelgerdi
- Novin Genome (NG) Lab, Research and Development Center for Biotechnology, Shahrekord, Iran
- Young Researchers and Elite Club, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| |
Collapse
|
7
|
Lv K, Liang Q. Macrophages in sepsis-induced acute lung injury: exosomal modulation and therapeutic potential. Front Immunol 2025; 15:1518008. [PMID: 39840035 PMCID: PMC11746006 DOI: 10.3389/fimmu.2024.1518008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 12/16/2024] [Indexed: 01/23/2025] Open
Abstract
Sepsis-induced acute lung injury (ALI) remains a leading cause of mortality in critically ill patients. Macrophages, key modulators of immune responses, play a dual role in both promoting and resolving inflammation. Exosomes, small extracellular vesicles released by various cells, carry bioactive molecules that influence macrophage polarization and immune responses. Emerging researchers have identified exosomes as crucial mediators that modulate macrophage activity during sepsis-induced ALI. This review explores the role of exosomes in modulating macrophage functions, focusing on the cellular interactions within the lung microenvironment and their potential as therapeutic targets. It highlights the regulation of macrophages by exosomes derived from pathogenic germs, neutrophils, alveolar epithelial cells, and mesenchymal stromal cells. By understanding these mechanisms, it aims to uncover innovative therapeutic strategies for sepsis-induced ALI.
Collapse
Affiliation(s)
- Kaiying Lv
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Qun Liang
- Department of Critical Care Medicine, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
8
|
Saleem M, Shahzad KA, Marryum M, Singh S, Zhou Q, Du S, Wang S, Shao C, Shaikh II. Exosome-based therapies for inflammatory disorders: a review of recent advances. Stem Cell Res Ther 2024; 15:477. [PMID: 39695750 DOI: 10.1186/s13287-024-04107-2] [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: 10/26/2024] [Accepted: 12/09/2024] [Indexed: 12/20/2024] Open
Abstract
Exosomes, small extracellular vesicles secreted by cells, have emerged as focal mediators in intercellular communication and therapeutic interventions across diverse biomedical fields. Inflammatory disorders, including inflammatory bowel disease, acute liver injury, lung injury, neuroinflammation, and myocardial infarction, are complex conditions that require innovative therapeutic approaches. This review summarizes recent advances in exosome-based therapies for inflammatory disorders, highlighting their potential as diagnostic biomarkers and therapeutic agents. Exosomes have shown promise in reducing inflammation, promoting tissue repair, and improving functional outcomes in preclinical models of inflammatory disorders. However, further research is needed to overcome the challenges associated with exosome isolation, characterization, and delivery, as well as to fully understand their mechanisms of action. Current limitations and future directions in exosome research underscore the need for enhanced isolation techniques and deeper mechanistic insights to harness exosomes' full therapeutic potential in clinical applications. Despite these challenges, exosome-based therapies hold great potential for the treatment of inflammatory disorders and may offer a new paradigm for personalized medication.
Collapse
Affiliation(s)
- Mavra Saleem
- Department of Zoology, The Islamia University of Bahawalpur, Bahawalpur, 63100, Pakistan
| | - Khawar Ali Shahzad
- Department of Zoology, The Islamia University of Bahawalpur, Bahawalpur, 63100, Pakistan
- Department of ORL-HNS, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Munazzah Marryum
- Department of Zoology, The Islamia University of Bahawalpur, Bahawalpur, 63100, Pakistan
| | - Shekhar Singh
- Lishui People's Hospital, Central Laboratory of The Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui, 323000, Zhejiang, China
| | - Quan Zhou
- Lishui People's Hospital, Central Laboratory of The Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui, 323000, Zhejiang, China
| | - Siting Du
- Lishui People's Hospital, Central Laboratory of The Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui, 323000, Zhejiang, China
| | - Shuanghu Wang
- Lishui People's Hospital, Central Laboratory of The Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui, 323000, Zhejiang, China
| | - Chuxiao Shao
- Lishui People's Hospital, Central Laboratory of The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, Zhejiang, China
| | - Imran Ibrahim Shaikh
- Lishui People's Hospital, Central Laboratory of The Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui, 323000, Zhejiang, China.
| |
Collapse
|
9
|
Ye R, Wei Y, Li J, Xu M, Xie H, Huang J, Deng L, Li C. MiRNAs and Neutrophil-Related Membrane Proteins from Plasma-Derived Extracellular Vesicles for Early Prediction of Organ Dysfunction and Prognosis in Septic Patients. J Inflamm Res 2024; 17:10347-10369. [PMID: 39649421 PMCID: PMC11625425 DOI: 10.2147/jir.s492902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 11/28/2024] [Indexed: 12/10/2024] Open
Abstract
Purpose The pathogenesis of sepsis-induced organ dysfunction remains elusive, and the mortality remains alarmingly high. We sought to investigate the profile of extracellular vesicles (EVs)-mediated communication between plasma and polymorphonuclear neutrophils (PMNs) in sepsis, and to elucidate whether miRNAs and PMN-related membrane proteins from plasma-derived EVs (plasma-EVs) are associated with sepsis-induced organ dysfunction and prognosis. Methods PMN-derived EVs (PMN-EVs) were isolated from the blood samples of healthy controls (N=3) and patients with septic shock (N=3) after ICU admission. We performed miRNA sequencing of the isolated EVs, followed by bioinformatic analysis. A miRNA model for comparing PMN-EVs and plasma-EVs was successfully established in the training cohort. Furthermore, miRNAs and PMN-related membrane proteins from the plasma-EV model were confirmed in the validation cohort. A logistic regression model, receiver operating characteristic (ROC) curves, and Kaplan-Meier analyses were performed to evaluate the efficiency of diagnostic and/or prognostic performance. Further, in vivo and in vitro experiments were conducted to explore the involvement of plasma-EVs in PMNs autophagy. Results Fifty-five miRNAs from PMN-EVs differed significantly between the healthy controls and patients with septic shock. Furthermore, the plasma-EV model (six miRNAs and eight PMN-related membrane proteins) was confirmed in the validation cohort, demonstrating that miR-34a-5p, miR-503-5p, miR-4772-3p, ITGAM, MPO, and MMP9 serve as sepsis biomarkers for distinguishing lung, liver, and kidney dysfunction. Kaplan-Meier survival analysis showed that miR-34a-5p, miR-4772-3p, ITGAM, and MMP9 were potential prognostic predictors. Finally, we found that plasma-EVs from sepsis patients exert an inhibitory effect on PMNs autophagy, which can be reversed by EV inhibitors such as GW4869 and enoxaparin. Conclusion These findings suggest that miRNAs and PMN-related membrane proteins from plasma-EVs could be valuable diagnostic tools for identifying sepsis-induced organ dysfunction and predicting prognosis, enabling proactive management of sepsis by physicians and improving the prognosis of sepsis patients.
Collapse
Affiliation(s)
- Rongzong Ye
- Department of Emergency Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Yating Wei
- Department of Emergency Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Jingwen Li
- Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Meili Xu
- Department of Emergency Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Haiyang Xie
- Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Jiahao Huang
- Department of Critical Care Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524000, People’s Republic of China
| | - Liehua Deng
- Department of Critical Care Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524000, People’s Republic of China
| | - Chaoqian Li
- Department of Emergency Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| |
Collapse
|
10
|
Yuan Y, Xiao Y, Zhao J, Zhang L, Li M, Luo L, Jia Y, Wang K, Chen Y, Wang P, Wang Y, Wei J, Shen K, Hu D. Exosomes as novel biomarkers in sepsis and sepsis related organ failure. J Transl Med 2024; 22:1078. [PMID: 39609831 PMCID: PMC11604007 DOI: 10.1186/s12967-024-05817-0] [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: 07/19/2024] [Accepted: 10/29/2024] [Indexed: 11/30/2024] Open
Abstract
Sepsis, a severe and life-threatening condition arising from a dysfunctional host response to infection, presents considerable challenges to the health care system and is characterized by high mortality rates and substantial economic costs. Exosomes have garnered attention as potential diagnostic markers because of their capacity to mirror the pathophysiological milieu of sepsis. This discourse reviews the progression of sepsis classification from Sepsis 1.0 to Sepsis 3.0, highlighting the imperative for sensitive and specific biomarkers to facilitate timely diagnosis and optimize patient outcomes. Existing biomarkers, such as procalcitonin (PCT) and C-reactive protein (CRP), exhibit certain limitations, thereby prompting the quest for more dependable diagnostic indicators. Exosomal cargoes, which encompass proteins and miRNAs, present a trove of biomarker candidates, attributable to their stability, pervasive presence, and indicative nature of the disease status. The potential of exosomal biomarkers in the identification of sepsis-induced organ damage, including cardiomyopathy, acute kidney injury, and acute lung injury, is emphasized, as they provide real-time insights into cardiac and renal impairments. Despite promising prospects, hurdles persist in the standardization of exosome extraction and the need for extensive clinical trials to validate their efficacy. The combination of biomarker development and sophisticated exosome detection techniques represents a pioneering strategy in the realm of sepsis diagnosis and management, underscoring the significance of further research and clinical validation.
Collapse
Affiliation(s)
- Yixuan Yuan
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Chang-Le Xi Street #127, Xi'an, 710032, China
| | - Yujie Xiao
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Chang-Le Xi Street #127, Xi'an, 710032, China
| | - Jiazhen Zhao
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Lixia Zhang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Chang-Le Xi Street #127, Xi'an, 710032, China
| | - Mengyang Li
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Chang-Le Xi Street #127, Xi'an, 710032, China
| | - Liang Luo
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Chang-Le Xi Street #127, Xi'an, 710032, China
| | - Yanhui Jia
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Chang-Le Xi Street #127, Xi'an, 710032, China
| | - Kejia Wang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Chang-Le Xi Street #127, Xi'an, 710032, China
| | - Yuxi Chen
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Chang-Le Xi Street #127, Xi'an, 710032, China
| | - Peng Wang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Chang-Le Xi Street #127, Xi'an, 710032, China
| | - Yuhang Wang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Chang-Le Xi Street #127, Xi'an, 710032, China
| | - Jingtao Wei
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Chang-Le Xi Street #127, Xi'an, 710032, China
| | - Kuo Shen
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Chang-Le Xi Street #127, Xi'an, 710032, China.
- Air Force Hospital of Western Theater Command, Gongnongyuan Street #1, Chengdu, 610065, China.
| | - Dahai Hu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Chang-Le Xi Street #127, Xi'an, 710032, China.
| |
Collapse
|
11
|
Lin L, Liu H, Zhang D, Du L, Zhang H. Nanolevel Immunomodulators in Sepsis: Novel Roles, Current Perspectives, and Future Directions. Int J Nanomedicine 2024; 19:12529-12556. [PMID: 39606559 PMCID: PMC11600945 DOI: 10.2147/ijn.s496456] [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: 10/01/2024] [Accepted: 11/13/2024] [Indexed: 11/29/2024] Open
Abstract
Sepsis represents a profound challenge in critical care, characterized by a severe systemic inflammatory response which can lead to multi-organ failure and death. The intricate pathophysiology of sepsis involves an overwhelming immune reaction that disrupts normal host defense mechanisms, necessitating innovative approaches to modulation. Nanoscale immunomodulators, with their precision targeting and controlled release capabilities, have emerged as a potent solution to recalibrate immune responses in sepsis. This review explores the recent advancements in nanotechnology for sepsis management, emphasizing the integration of nanoparticulate systems to modulate immune function and inflammatory pathways. Discussions detail the development of the immune system, the distinct inflammatory responses triggered by sepsis, and the scientific principles underpinning nanoscale immunomodulation, including specific targeting mechanisms and delivery systems. The review highlights nanoformulation designs aimed at enhancing bioavailability, stability, and therapeutic efficacy, which shows promise in clinical settings by modulating key inflammatory pathways. Ultimately, this review synthesizes the current state of knowledge and projects future directions for research, underscoring the transformative potential of nanolevel immunomodulators for sepsis treatment through innovative technologies and therapeutic strategies.
Collapse
Affiliation(s)
- Liangkang Lin
- Department of Pediatrics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, People’s Republic of China
| | - Hanyou Liu
- Department of Pediatrics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, People’s Republic of China
| | - Dingshan Zhang
- Department of Intensive Care Unit, Public Health Clinical Center of Chengdu, Chengdu, People’s Republic of China
| | - Lijia Du
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, People’s Republic of China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, People’s Republic of China
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, People’s Republic of China
| | - Haiyang Zhang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, People’s Republic of China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, People’s Republic of China
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, People’s Republic of China
| |
Collapse
|
12
|
Xu M, Zeng X, Pan M, Chen R, Bai Y, He J, Wang C, Qi Y, Sun Q, Wang C, An N. MiR-92a-3p Promotes Renal Injury and Fibrosis Through Facilitating M1 Macrophage Polarization via Targeting LIN28A. Physiol Res 2024; 73:755-767. [PMID: 39545790 PMCID: PMC11629952 DOI: 10.33549/physiolres.935305] [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: 12/18/2023] [Accepted: 05/07/2024] [Indexed: 12/13/2024] Open
Abstract
Infiltrated and activated M1 macrophages play a role in kidney injury and fibrosis during chronic kidney disease (CKD) progression. However, the specific ways that M1 macrophage polarization contributes to renal fibrosis are not fully understood. The study seeks to investigate how miR-92a-3p regulates M1 macrophage polarization and its connection to renal fibrosis in the development of CKD. Our results revealed that miR-92a-3p overexpression increased M1-macrophage activation, iNOS, IL-6, and TNF-alpha expression in RAW264.7 upon LPS stimulation. LIN28A overexpression reversed these effects. Moreover, miR-92a-3p overexpression in RAW264.7 exacerbated NRK-52E cell apoptosis induced by LPS, but LIN28A overexpression counteracted this effect. MiR-92a-3p knockout in unilateral ureteral obstruction (UUO) C57BL/6 mice led to reduced renal infiltration and fibrosis, accompanied by decreased iNOS, alpha-SMA, IL-6, TNF-alpha, and increased LIN28A. In summary, our findings suggest that miR-92a-3p may play a role in promoting renal injury and fibrosis both in vitro and in vivo. This effect is potentially achieved by facilitating M1 macrophage polarization through the targeting of LIN28A.
Collapse
Affiliation(s)
- M Xu
- Blood Purification Center, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Xiuying District, Haikou, Hainan Province, China.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Xu H, Li X, Liu K, Huang P, Liu XJ. PM2.5 Promotes Macrophage-Mediated Inflammatory Response Through Airway Epithelial Cell-Derived Exosomal miR-155-5p. J Inflamm Res 2024; 17:8555-8567. [PMID: 39539727 PMCID: PMC11559224 DOI: 10.2147/jir.s482509] [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: 08/07/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
Background Airway epithelial cells (AECs) and alveolar macrophages are involved in airway inflammation. The direct effects of atmospheric fine-particulate-matter (PM2.5) on airway cells, such as AECs and alveolar macrophages, have been widely investigated, but the effect of cell-cell interaction on inflammatory response remains unclear. Exosomes play a crucial role in intercellular communication. However, the cellular interaction of exosomes in PM2.5-induced airway inflammation is unclear. Methods The PM2.5-induced human bronchial epithelial (BEAS-2B) cells and phorbol 12-myristate 13-acetate-induced macrophages (Mφ) were co-cultured and then the expression of IL-6, IL-1β, TNF-α and miRNA-155-5p were detected. Exosomes from PM2.5-exposed BEAS-2B cells were then co-cultured with Mφ to detect the expression of miR-155-5p and inflammatory cytokines, as well as cytokine signaling inhibitor-1 (SOCS1)/NFκB, and to detect the effect of the exosome inhibitor GW4869. Results After the co-culture of PM2.5-induced BEAS-2B cells and Mφ, the expression of Mφ-derived IL-6, IL-1β, and TNF-α, as well as miRNA-155-5p were upregulated. The expression of miRNA-155-5p was upregulated in BEAS-2B and BEAS-2B cell-derived exosomes after exposure to PM2.5. Furthermore, co-culturing exosomes derived from PM2.5-exposed BEAS-2B cells with Mφ, upregulated miR-155-5p and inflammatory cytokines, decreased cytokine signaling inhibitor-1 (SOCS1) expression, and activated NF-κB. In addition, adding exosome inhibitor GW4869 to PM2.5-interfered BEAS-2B cells co-culture with Mφ downregulated miRNA-155-5p expression, inhibited NF-κB, and reduced the levels of inflammatory factors. Conclusion PM2.5 promotes Mφ inflammation by upregulating miRNA-155-5P in exosomes obtained from BEAS-2B cells through miR-155-5P/SOCS1/NF-κB pathway. Exosomal miRNAs mediate cellular communication between BEAS-2B cells and Mφ, which may be a new mechanism of PM2.5-stimulated pulmonary inflammatory response.
Collapse
Affiliation(s)
- Hui Xu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, People’s Republic of China
- The First Hospital of Lanzhou University, Lanzhou, Gansu, People’s Republic of China
| | - Xin Li
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, People’s Republic of China
| | - Kai Liu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, People’s Republic of China
| | - Ping Huang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, People’s Republic of China
| | - Xiao-Ju Liu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, People’s Republic of China
- The First Hospital of Lanzhou University, Lanzhou, Gansu, People’s Republic of China
| |
Collapse
|
14
|
Jadamba B, Jin Y, Lee H. Harmonising cellular conversations: decoding the vital roles of extracellular vesicles in respiratory system intercellular communications. Eur Respir Rev 2024; 33:230272. [PMID: 39537245 PMCID: PMC11558538 DOI: 10.1183/16000617.0272-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 08/22/2024] [Indexed: 11/16/2024] Open
Abstract
Extracellular vesicles (EVs) released by various cells play crucial roles in intercellular communication within the respiratory system. This review explores the historical context and significance of research into extracellular vesicles. Categorised into exosomes (sized 30-150 nm), microvesicles (sized 50-1000 nm) and apoptotic bodies (sized 500-2000nm), based on their generation mechanisms, extracellular vesicles carry diverse cargoes of biomolecules, including proteins, lipids and nucleic acids. Respiratory ailments are the primary contributors to both mortality and morbidity across various populations globally, significantly impacting public health. Recent studies have underscored the pivotal role of extracellular vesicles, particularly their cargo content, in mediating intercellular communication between lung cells in respiratory diseases. This comprehensive review provides insights into extracellular vesicle mechanisms and emphasises their significance in major respiratory conditions, including acute lung injury, COPD, pulmonary hypertension, pulmonary fibrosis, asthma and lung cancer.
Collapse
Affiliation(s)
- Budjav Jadamba
- Department of Biology and Chemistry, Changwon National University, Changwon, Korea
| | - Yang Jin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Heedoo Lee
- Department of Biology and Chemistry, Changwon National University, Changwon, Korea
| |
Collapse
|
15
|
Zeng M, Liu M, Tao X, Yin X, Shen C, Wang X. Emerging Trends in the Application of Extracellular Vesicles as Novel Oral Delivery Vehicles for Therapeutics in Inflammatory Diseases. Int J Nanomedicine 2024; 19:8573-8601. [PMID: 39185348 PMCID: PMC11345024 DOI: 10.2147/ijn.s475532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 08/08/2024] [Indexed: 08/27/2024] Open
Abstract
Inflammation involves complex immune responses where cytokines such as TNF-α, IL-1, and IL-6 promote vasodilation and increased vascular permeability to facilitate immune cell migration to inflammation sites. Persistent inflammation is linked to diseases like cancer, arthritis, and neurodegenerative disorders. Although oral anti-inflammatory drugs are favored for their non-invasiveness and cost-effectiveness, their efficacy is often compromised due to gastrointestinal degradation and limited bioavailability. Recent advancements highlight the potential of extracellular vesicles (EVs) as nanocarriers that enhance drug delivery by encapsulating therapeutic agents, ensuring targeted release and reduced toxicity. These EVs, derived from dietary sources and cell cultures, exhibit excellent biocompatibility and stability, presenting a novel approach in anti-inflammatory therapies. This review discusses the classification and advantages of orally administered EVs (O-EVs), their mechanism of action, and their emerging role in treating inflammatory conditions, positioning them as promising vectors in the development of innovative anti-inflammatory drug delivery systems.
Collapse
Affiliation(s)
- Mingtang Zeng
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Maozhu Liu
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Xuelin Tao
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Xi Yin
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Chao Shen
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Xueyan Wang
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| |
Collapse
|
16
|
Yu T, Mi N, Song Y, Xie W. Exosomes miR-92a-3p from human exfoliated deciduous teeth inhibits periodontitis progression via the KLF4/PI3K/AKT pathway. J Periodontal Res 2024; 59:771-782. [PMID: 38616305 DOI: 10.1111/jre.13262] [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: 08/04/2023] [Revised: 03/07/2024] [Accepted: 03/19/2024] [Indexed: 04/16/2024]
Abstract
BACKGROUND Periodontitis is a chronic inflammatory disease mediated by dysbiosis of the oral microflora, resulting in the destruction of periodontal tissue. Increasing evidence suggested that mesenchymal stem cell (MSCs) and exosomes derived from MSCs play a critical role in periodontal tissue regeneration. However, whether stem cells from exfoliated deciduous teeth (SHED)-secreted exosomes can improve the therapeutic potential of periodontitis is largely unknown. OBJECTIVE Here, we aim to evaluate the effect of SHED-exosomes on inflammation, apoptosis and osteogenic differentiation in periodontitis. METHODS The periodontitis cell model was constructed by stimulating periodontal ligament stem cells (PDLSCs) with lipopolysaccharide (LPS), and the periodontitis rats were established by ligation. RESULTS First, we isolated exosomes from the SHED, and we figured out that exosomes secreted by SHED were enriched in miR-92a-3p and the exosomes enhanced proliferation and osteogenic differentiation and reduced apoptosis and inflammatory responses in PDLSCs. In addition, we found that SHED-exosomes alleviated inflammatory effect and elevated the expression of osteogenic-related genes in periodontitis rat model. Moreover, miR-92a-3p targeted downstream Krüppel-Like Transcription Factor 4 (KLF4) and regulated the PI3K/AKT pathway. Finally, our data indicated that upregulation of KLF4 or activation of PI3K/AKT by 740Y-P counteracted the inhibitory effect of SHED-exosomes on periodontitis progression. CONCLUSION Taken together, our finding revealed that exosomal miR-92a-3p derived from SHED contributed to the alleviation of periodontitis development and progression through inactivating the KLF4/PI3K/AKT signaling pathway, which may provide a potential target for the treatment of periodontitis.
Collapse
Affiliation(s)
- Tianliang Yu
- Department of Prosthodontics, The First Affiliated Clinical Hospital of Harbin Medical University, Harbin, China
| | - Na Mi
- Department of Endodontics, The First Affiliated Clinical Hospital of Harbin Medical University, Harbin, China
| | - Yingtao Song
- Department of Prosthodontics, The First Affiliated Clinical Hospital of Harbin Medical University, Harbin, China
| | - Weili Xie
- Department of Prosthodontics, The First Affiliated Clinical Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
17
|
Meng Q, Winston T, Ma J, Song Y, Wang C, Yang J, Ma Z, Cooney RN. INDUCED PLURIPOTENT STEM CELL-DERIVED MESENCHYMAL STEM CELLS-DERIVED EXTRACELLULAR VESICLES ATTENUATE LPS-INDUCED LUNG INJURY AND ENDOTOXEMIA IN MICE. Shock 2024; 62:294-303. [PMID: 38813932 PMCID: PMC11466509 DOI: 10.1097/shk.0000000000002381] [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] [Indexed: 05/31/2024]
Abstract
ABSTRACT Introduction: We hypothesized extracellular vesicles (EVs) from preconditioned human-induced pluripotent stem cell-derived mesenchymal stem cells (iMSCs) attenuate LPS-induced acute lung injury (ALI) and endotoxemia. Methods: iMSCs were incubated with cell stimulation cocktail (CSC) and EVs were isolated. iMSC-EVs were characterized by size and EV markers. Biodistribution of intratracheal (IT), intravenous, and intraperitoneal injection of iMSC-EVs in mice was examined using IVIS. Uptake of iMSC-EVs in lung tissue, alveolar macrophages, and RAW264.7 cells was also assessed. C57BL/6 mice were treated with IT/IP iMSC-EVs or vehicle ± IT/IP LPS to induce ALI/acute respiratory distress syndrome and endotoxemia. Lung tissues, plasma, and bronchoalveolar lavage fluid (BALF) were harvested at 24 h. Lung histology, BALF neutrophil/macrophage, cytokine levels, and total protein concentration were measured to assess ALI and inflammation. Survival studies were performed using IP LPS in mice for 3 days. Results: iMSC-EV route of administration resulted in differential tissue distribution. iMSC-EVs were taken up by alveolar macrophages in mouse lung and cultured RAW264.7 cells. IT LPS-treated mice demonstrated marked histologic ALI, increased BALF neutrophils/macrophages and protein, and increased BALF and plasma TNF-α/IL-6 levels. These parameters were attenuated by 2 h before or 2 h after treatment with IT iMSC-EVs in ALI mice. Interestingly, the IT LPS-induced increase in IL-10 was augmented by iMSC-EVs. Mice treated with IP LPS showed increases in TNF-α and IL-6 that were downregulated by iMSC-EVs and LPS-induced mortality was ameliorated by iMSC-EVs. Administration of IT iMSC-EVs 2 h after LPS downregulated the increase in proinflammatory cytokines (TNF-α/IL-6) by LPS and further increased IL-10 levels. Conclusions: iMSC-EVs attenuate the inflammatory effects of LPS on cytokine levels in ALI and IP LPS in mice. LPS-induced mortality was improved with administration of iMSC-EVs.
Collapse
Affiliation(s)
- Qinghe Meng
- Department of Surgery, State University of New York (SUNY), Upstate Medical University, Syracuse, New York
| | - Tackla Winston
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, New York
| | - Julia Ma
- Department of Medicine, State University of New York (SUNY), Upstate Medical University, Syracuse, New York
| | - Yuanhui Song
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, New York
| | - Chunyan Wang
- Department of Surgery, State University of New York (SUNY), Upstate Medical University, Syracuse, New York
| | - Junhui Yang
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, New York
| | - Zhen Ma
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, New York
| | - Robert N Cooney
- Department of Surgery, State University of New York (SUNY), Upstate Medical University, Syracuse, New York
| |
Collapse
|
18
|
Xiao Y, Yuan Y, Hu D, Wang H. Exosome-Derived microRNA: Potential Target for Diagnosis and Treatment of Sepsis. J Immunol Res 2024; 2024:4481452. [PMID: 39104595 PMCID: PMC11300089 DOI: 10.1155/2024/4481452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 06/23/2024] [Accepted: 07/06/2024] [Indexed: 08/07/2024] Open
Abstract
Exosome-derived microRNAs (miRNAs) are emerging as pivotal players in the pathophysiology of sepsis, representing a new frontier in both the diagnosis and treatment of this complex condition. Sepsis, a severe systemic response to infection, involves intricate immune and nonimmune mechanisms, where exosome-mediated communication can significantly influence disease progression and outcomes. During the progress of sepsis, the miRNA profile of exosomes undergoes notable alterations, is reflecting, and may affect the progression of the disease. This review comprehensively explores the biology of exosome-derived miRNAs, which originate from both immune cells (such as macrophages and dendritic cells) and nonimmune cells (such as endothelial and epithelial cells) and play a dynamic role in modulating pathways that affect the course of sepsis, including those related to inflammation, immune response, cell survival, and apoptosis. Taking into account these dynamic changes, we further discuss the potential of exosome-derived miRNAs as biomarkers for the early detection and prognosis of sepsis and advantages over traditional biomarkers due to their stability and specificity. Furthermore, this review evaluates exosome-based therapeutic miRNA delivery systems in sepsis, which may pave the way for targeted modulation of the septic response and personalized treatment options.
Collapse
Affiliation(s)
- Yujie Xiao
- Department of Burns and Cutaneous SurgeryXijing HospitalFourth Military Medical University, 127 West Chang-le Road, Xi'an 710032, Shaanxi, China
| | - Yixuan Yuan
- Department of Burns and Cutaneous SurgeryXijing HospitalFourth Military Medical University, 127 West Chang-le Road, Xi'an 710032, Shaanxi, China
| | - Dahai Hu
- Department of Burns and Cutaneous SurgeryXijing HospitalFourth Military Medical University, 127 West Chang-le Road, Xi'an 710032, Shaanxi, China
| | - Hongtao Wang
- Department of Burns and Cutaneous SurgeryXijing HospitalFourth Military Medical University, 127 West Chang-le Road, Xi'an 710032, Shaanxi, China
| |
Collapse
|
19
|
Wang G, Ma X, Huang W, Wang S, Lou A, Wang J, Tu Y, Cui W, Zhou W, Zhang W, Li Y, Geng S, Meng Y, Li X. Macrophage biomimetic nanoparticle-targeted functional extracellular vesicle micro-RNAs revealed via multiomics analysis alleviate sepsis-induced acute lung injury. J Nanobiotechnology 2024; 22:362. [PMID: 38910259 PMCID: PMC11194988 DOI: 10.1186/s12951-024-02597-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: 08/07/2023] [Accepted: 05/28/2024] [Indexed: 06/25/2024] Open
Abstract
Patients who suffer from sepsis typically experience acute lung injury (ALI). Extracellular vesicles (EVs) contain miRNAs, which are potentially involved in ALI. However, strategies to screen more effective EV-miRNAs as therapeutic targets are yet to be elucidated. In this study, functional EV-miRNAs were identified based on multiomics analysis of single-cell RNA sequencing of targeted organs and serum EV (sEV) miRNA profiles in patients with sepsis. The proportions of neutrophils and macrophages were increased significantly in the lungs of mice receiving sEVs from patients with sepsis compared with healthy controls. Macrophages released more EVs than neutrophils. MiR-125a-5p delivery by sEVs to lung macrophages inhibited Tnfaip3, while miR-221-3p delivery to lung neutrophils inhibited Fos. Macrophage membrane nanoparticles (MM NPs) loaded with an miR-125a-5p inhibitor or miR-221-3p mimic attenuated the response to lipopolysaccharide (LPS)-induced ALI. Transcriptome profiling revealed that EVs derived from LPS-stimulated bone marrow-derived macrophages (BMDMs) induced oxidative stress in neutrophils. Blocking toll-like receptor, CXCR2, or TNFα signaling in neutrophils attenuated the oxidative stress induced by LPS-stimulated BMDM-EVs. This study presents a novel method to screen functional EV-miRNAs and highlights the pivotal role of macrophage-derived EVs in ALI. MM NPs, as delivery systems of key sEV-miRNA mimics or inhibitors, alleviated cellular responses observed in sepsis-induced ALI. This strategy can be used to reduce septic organ damage, particularly lung damage, by targeting EVs.
Collapse
Affiliation(s)
- Guozhen Wang
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Southern Medical University, Guangzhou, 510515, Guangdong, China
- Department of Gastroenterology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China
| | - Xiaoxin Ma
- Department of Gastroenterology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China
| | - Weichang Huang
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Shuanghu Wang
- Central Laboratory, Wenzhou Medical University Lishui Hospital, Lishui People's Hospital, Lishui, Zhejiang 323000, China
| | - Anni Lou
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jun Wang
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Southern Medical University, Guangzhou, 510515, Guangdong, China
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yingfeng Tu
- School of Pharmaceutical Science, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| | - Wanfu Cui
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Wangmei Zhou
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Wenyong Zhang
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yue Li
- Department of Intensive Care Unit, General Hospital of Southern Theatre Command, Southern Medical University, Guangzhou 510515, China
| | - Shiyu Geng
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Ying Meng
- Department of Respiratory Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Xu Li
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
20
|
Li Y, Xu H, Wang Y, Zhu Y, Xu K, Yang Z, Li Y, Guo C. Epithelium-derived exosomes promote silica nanoparticles-induced pulmonary fibroblast activation and collagen deposition via modulating fibrotic signaling pathways and their epigenetic regulations. J Nanobiotechnology 2024; 22:331. [PMID: 38867284 PMCID: PMC11170844 DOI: 10.1186/s12951-024-02609-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: 10/27/2023] [Accepted: 05/30/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND In the context of increasing exposure to silica nanoparticles (SiNPs) and ensuing respiratory health risks, emerging evidence has suggested that SiNPs can cause a series of pathological lung injuries, including fibrotic lesions. However, the underlying mediators in the lung fibrogenesis caused by SiNPs have not yet been elucidated. RESULTS The in vivo investigation verified that long-term inhalation exposure to SiNPs induced fibroblast activation and collagen deposition in the rat lungs. In vitro, the uptake of exosomes derived from SiNPs-stimulated lung epithelial cells (BEAS-2B) by fibroblasts (MRC-5) enhanced its proliferation, adhesion, and activation. In particular, the mechanistic investigation revealed SiNPs stimulated an increase of epithelium-secreted exosomal miR-494-3p and thereby disrupted the TGF-β/BMPR2/Smad pathway in fibroblasts via targeting bone morphogenetic protein receptor 2 (BMPR2), ultimately resulting in fibroblast activation and collagen deposition. Conversely, the inhibitor of exosomes, GW4869, can abolish the induction of upregulated miR-494-3p and fibroblast activation in MRC-5 cells by the SiNPs-treated supernatants of BEAS-2B. Besides, inhibiting miR-494-3p or overexpression of BMPR2 could ameliorate fibroblast activation by interfering with the TGF-β/BMPR2/Smad pathway. CONCLUSIONS Our data suggested pulmonary epithelium-derived exosomes serve an essential role in fibroblast activation and collagen deposition in the lungs upon SiNPs stimuli, in particular, attributing to exosomal miR-494-3p targeting BMPR2 to modulate TGF-β/BMPR2/Smad pathway. Hence, strategies targeting exosomes could be a new avenue in developing therapeutics against lung injury elicited by SiNPs.
Collapse
Affiliation(s)
- Yan Li
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, No.10 Xitoutiao, You An Men, Beijing, 100069, China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, No.10 Xitoutiao, You An Men, Beijing, 100069, China
| | - Hailin Xu
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, No.10 Xitoutiao, You An Men, Beijing, 100069, China
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, No.10 Xitoutiao, You An Men, Beijing, 100069, China
| | - Ying Wang
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, No.10 Xitoutiao, You An Men, Beijing, 100069, China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, No.10 Xitoutiao, You An Men, Beijing, 100069, China
| | - Yurou Zhu
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, No.10 Xitoutiao, You An Men, Beijing, 100069, China
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, No.10 Xitoutiao, You An Men, Beijing, 100069, China
| | - Kun Xu
- School of Medicine, Hunan Normal University, Changsha, 410013, Hunan, China
| | - Zhu Yang
- State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong, China
| | - Yanbo Li
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, No.10 Xitoutiao, You An Men, Beijing, 100069, China.
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, No.10 Xitoutiao, You An Men, Beijing, 100069, China.
| | - Caixia Guo
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, No.10 Xitoutiao, You An Men, Beijing, 100069, China.
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, No.10 Xitoutiao, You An Men, Beijing, 100069, China.
| |
Collapse
|
21
|
Wu X, Tang Y, Lu X, Liu Y, Liu X, Sun Q, Wang L, Huang W, Liu A, Liu L, Chao J, Zhang X, Qiu H. Endothelial cell-derived extracellular vesicles modulate the therapeutic efficacy of mesenchymal stem cells through IDH2/TET pathway in ARDS. Cell Commun Signal 2024; 22:293. [PMID: 38802896 PMCID: PMC11129421 DOI: 10.1186/s12964-024-01672-0] [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: 05/20/2024] [Indexed: 05/29/2024] Open
Abstract
BACKGROUND Acute respiratory distress syndrome (ARDS) is a severe and fatal disease. Although mesenchymal stem cell (MSC)-based therapy has shown remarkable efficacy in treating ARDS in animal experiments, clinical outcomes have been unsatisfactory, which may be attributed to the influence of the lung microenvironment during MSC administration. Extracellular vesicles (EVs) derived from endothelial cells (EC-EVs) are important components of the lung microenvironment and play a crucial role in ARDS. However, the effect of EC-EVs on MSC therapy is still unclear. In this study, we established lipopolysaccharide (LPS) - induced acute lung injury model to evaluate the impact of EC-EVs on the reparative effects of bone marrow-derived MSC (BM-MSC) transplantation on lung injury and to unravel the underlying mechanisms. METHODS EVs were isolated from bronchoalveolar lavage fluid of mice with LPS - induced acute lung injury and patients with ARDS using ultracentrifugation. and the changes of EC-EVs were analysed using nanoflow cytometry analysis. In vitro assays were performed to establish the impact of EC-EVs on MSC functions, including cell viability and migration, while in vivo studies were performed to validate the therapeutic effect of EC-EVs on MSCs. RNA-Seq analysis, small interfering RNA (siRNA), and a recombinant lentivirus were used to investigate the underlying mechanisms. RESULTS Compared with that in non-ARDS patients, the quantity of EC-EVs in the lung microenvironment was significantly greater in patients with ARDS. EVs derived from lipopolysaccharide-stimulated endothelial cells (LPS-EVs) significantly decreased the viability and migration of BM-MSCs. Furthermore, engrafting BM-MSCs pretreated with LPS-EVs promoted the release of inflammatory cytokines and increased pulmonary microvascular permeability, aggravating lung injury. Mechanistically, LPS-EVs reduced the expression level of isocitrate dehydrogenase 2 (IDH2), which catalyses the formation of α-ketoglutarate (α-KG), an intermediate product of the tricarboxylic acid (TCA) cycle, in BM-MSCs. α-KG is a cofactor for ten-eleven translocation (TET) enzymes, which catalyse DNA hydroxymethylation in BM-MSCs. CONCLUSIONS This study revealed that EC-EVs in the lung microenvironment during ARDS can affect the therapeutic efficacy of BM-MSCs through the IDH2/TET pathway, providing potential strategies for improving the therapeutic efficacy of MSC-based therapy in the clinic.
Collapse
Affiliation(s)
- Xiao Wu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Ying Tang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Xinxing Lu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Yigao Liu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Xu Liu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Qin Sun
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Lu Wang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Wei Huang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Airan Liu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Ling Liu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Jie Chao
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China
- Department of Physiology, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Xiwen Zhang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China.
| | - Haibo Qiu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| |
Collapse
|
22
|
Zhang Z, Zhang X, Gao X, Fang B, Tian S, Kang P, Zhao Y. MiR-150-5p Alleviates Renal Tubule Epithelial Cell Fibrosis via the Inhibition of Epithelial-Mesenchymal Transition by Targeting ZEB1. Int Arch Allergy Immunol 2024; 185:827-835. [PMID: 38763133 DOI: 10.1159/000538670] [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: 02/15/2024] [Accepted: 03/28/2024] [Indexed: 05/21/2024] Open
Abstract
INTRODUCTION Although microRNA (miR)-150-5p participates in the progression of renal fibrosis, its mechanism of action remains elusive. METHODS A mouse model of unilateral ureteral obstruction was used. The in vitro renal fibrosis model was established by stimulating human kidney 2 (HK-2) cells with transforming growth factor beta 1 (TGF-β1). The expression profiles of miR-150-5p, zinc finger E-box binding homeobox 1 (ZEB1), and other fibrosis- and epithelial-mesenchymal transition (EMT)-linked proteins were determined using Western blot and quantitative reverse transcription polymerase chain reaction. The relationship between miR-150-5p and ZEB1 in HK-2 cells was confirmed by a dual-luciferase reporter assay. RESULTS Both in vivo and in vitro renal fibrosis models revealed reduced miR-150-5p expression and elevated ZEB1 level. A significant decrease in E-cadherin levels, as well as increases in alpha smooth muscle actin (α-SMA) and collagen type I (Col-I) levels, was seen in TGF-β1-treated HK-2 cells. The overexpression of miR-150-5p ameliorated TGF-β1-mediated fibrosis and EMT. Notably, miR-150-5p acts by directly targeting ZEB1. A significant reversal of the inhibitory impact of miR-150-5p on TGF-β1-mediated fibrosis and EMT in HK-2 cells was observed upon ZEB1 overexpression. CONCLUSION MiR-150-5p suppresses TGF-β1-induced fibrosis and EMT by targeting ZEB1 in HK-2 cells, providing helpful insights into the therapeutic intervention of renal fibrosis.
Collapse
Affiliation(s)
- Zhizhong Zhang
- Department of Urology, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, China
| | - Xinyu Zhang
- Department of Stomatology, Yinchuan Guolong Hospital, Yinchuan, China
| | - Xiangming Gao
- Department of Obstetrics and Gynecology, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, China
| | - Bing Fang
- Department of General Medicine, Yinchuan Meinian Health Hospital, Yinchuan, China
| | - Shuyu Tian
- Internal Medicine, Yinchuan Guolong Hospital, Yinchuan, China
| | - Ping Kang
- Department of Surgery, Yinchuan Guolong Hospital, Yinchuan, China
| | - Yi Zhao
- Department of Urology, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, China
| |
Collapse
|
23
|
Liu J, Song K, Lin B, Chen Z, Liu Y, Qiu X, He Q, Zuo Z, Yao X, Huang X, Liu Z, Liu Z, Huang Q, Guo X. The suppression of HSPA8 attenuates NLRP3 ubiquitination through SKP2 to promote pyroptosis in sepsis-induced lung injury. Cell Biosci 2024; 14:56. [PMID: 38698431 PMCID: PMC11064404 DOI: 10.1186/s13578-024-01239-z] [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: 12/14/2023] [Accepted: 04/25/2024] [Indexed: 05/05/2024] Open
Abstract
BACKGROUND Acute lung injury (ALI) is strongly associated with hospitalization and mortality in patients with sepsis. Recent evidence suggests that pyroptosis mediated by NLRP3(NOD-, LRR- and pyrin domain-containing 3) inflammasome activation plays a key role in sepsis. However, the mechanism of NLRP3 inflammasome activation in sepsis-induced lung injury remains unclear. RESULTS in this study, we demonstrated that NLRP3 inflammasome was activated by the down-regulation of heat shock protein family A member 8 (HSPA8) in Lipopolysaccharide (LPS) and adenosine triphosphate (ATP)-treated mouse alveolar epithelial cells (AECs). Geranylgeranylacetone (GGA)-induced HSPA8 overexpression in cecum ligation and puncture (CLP) mice could significantly reduce systemic inflammatory response and mortality, effectively protect lung function, whilst HSPA8 inhibitor VER155008 aggravated this effect. The inhibition of HSPA8 was involved in sepsis induced acute lung injury by promoting pyroptosis of AECs. The down-regulation of HSPA8 activated NLRP3 inflammasome to mediate pyroptosis by promoting the degradation of E3 ubiquitin ligase S-phase kinase-associated protein 2 (SKP2). In addition, when stimulated by LPS and ATP, down-regulated SKP2 promoted pyroptosis of AECs by further attenuating ubiquitination of NLRP3. Adeno-associated virus 9-SKP2(AAV9-SKP2) could promote NLRP3 ubiquitination and degradation, alleviate lung injury and inhibit systemic inflammatory response in vivo. CONCLUSION in summary, our study shows there is strong statistical evidence that the suppression of HSPA8 mediates alveolar epithelial pyroptosis by promoting the degradation of E3 ubiquitin ligase SKP2 and subsequently attenuating the ubiquitination of NLRP3 to activate the NLRP3 inflammasome, which provides a new perspective and therapeutic target for the treatment of sepsis-induced lung injury.
Collapse
Affiliation(s)
- Jinlian Liu
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, School of Basic Medical Sciences, National Experimental Education Demonstration Center for Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Ke Song
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, School of Basic Medical Sciences, National Experimental Education Demonstration Center for Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Bingqi Lin
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, School of Basic Medical Sciences, National Experimental Education Demonstration Center for Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Zhenfeng Chen
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, School of Basic Medical Sciences, National Experimental Education Demonstration Center for Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Yan Liu
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, School of Basic Medical Sciences, National Experimental Education Demonstration Center for Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Xianshuai Qiu
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, School of Basic Medical Sciences, National Experimental Education Demonstration Center for Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Qi He
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, School of Basic Medical Sciences, National Experimental Education Demonstration Center for Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Zirui Zuo
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, School of Basic Medical Sciences, National Experimental Education Demonstration Center for Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Xiaodan Yao
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, School of Basic Medical Sciences, National Experimental Education Demonstration Center for Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Xiaoxia Huang
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, School of Basic Medical Sciences, National Experimental Education Demonstration Center for Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Zhuanhua Liu
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, School of Basic Medical Sciences, National Experimental Education Demonstration Center for Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Zhifeng Liu
- Department of Medicine intensive care unit , National Clinical Research Center for Geriatric Diseases (Chinese PLA General Hospital), General Hospital of Southern Theatre Command of PLA, Guangdong Branch Center, Guangzhou, Guangdong, China.
| | - Qiaobing Huang
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, School of Basic Medical Sciences, National Experimental Education Demonstration Center for Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
- School of Basic Medical Sciences, Southern Medical University, 1023 Shatai Road, Tonghe, Guangzhou, 510515, China.
| | - Xiaohua Guo
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, School of Basic Medical Sciences, National Experimental Education Demonstration Center for Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
- School of Basic Medical Sciences, Southern Medical University, 1023 Shatai Road, Tonghe, Guangzhou, 510515, China.
| |
Collapse
|
24
|
Munley JA, Willis ML, Gillies GS, Kannan KB, Polcz VE, Balch JA, Barrios EL, Wallet SM, Bible LE, Efron PA, Maile R, Mohr AM. Exosomal microRNA following severe trauma: Role in bone marrow dysfunction. J Trauma Acute Care Surg 2024; 96:548-556. [PMID: 38151766 PMCID: PMC10978306 DOI: 10.1097/ta.0000000000004225] [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] [Indexed: 12/29/2023]
Abstract
INTRODUCTION Severe trauma disrupts bone marrow function and is associated with persistent anemia and altered hematopoiesis. Previously, plasma-derived exosomes isolated after trauma have been shown to suppress in vitro bone marrow function. However, the cargo contained in these vesicles has not been examined. We hypothesized that trauma plasma-derived exosomes exhibit microRNA (miRNA) changes that impact bone marrow function after severe injury. METHODS Plasma was collected from a prospective cohort study of trauma patients (n = 15; 7 males, 8 females) with hip and/or femur fractures and an Injury Severity Score of ≥15; elective total hip arthroplasty (THA) patients (n = 8; 4 males, 4 females) served as operative controls. Exosomes were isolated from plasma with the Invitrogen Total Exosome Isolation Kit (Thermo Fisher Scientific, Waltham, MA), and RNA was isolated using a miRNeasy Mini Kit (Qiagen, Hilden, Germany). Direct quantification of miRNA was performed by NanoString Technologies on a human miRNA gene panel and analyzed with nSolver with significance defined as p < 0.05. RESULTS There were no differences in age or sex distribution between trauma and THA groups; the average Injury Severity Score was 23. Trauma plasma-derived exosomes had 60 miRNA identities that were significantly downregulated and 3 miRNAs that were upregulated when compared with THA ( p < 0.05). Twelve of the downregulated miRNAs have a direct role in hematopoiesis regulation. Furthermore, male trauma plasma-derived exosomes demonstrated downregulation of 150 miRNAs compared with male THA ( p < 0.05). Female trauma plasma-derived exosomes demonstrated downregulation of only four miRNAs and upregulation of two miRNAs compared with female THA ( p < 0.05). CONCLUSION We observed downregulation of 12 miRNAs linked to hematopoiesis along with sexual dimorphism in miRNA expression from plasma-derived exosomes following severe trauma. Understanding sexually dimorphic miRNA expression provides new insight into sex-based changes in postinjury systemic inflammation, immune system dysregulation, and bone marrow dysfunction and will aid us in more precise future potential therapeutic strategies. LEVEL OF EVIDENCE Prognostic and Epidemiological; Level III.
Collapse
Affiliation(s)
- Jennifer A. Munley
- Department of Surgery and Sepsis and Critical Illness Research Center, University of Florida College of Medicine, Gainesville, Florida
| | - Micah L. Willis
- Department of Surgery and Sepsis and Critical Illness Research Center, University of Florida College of Medicine, Gainesville, Florida
| | - Gwendolyn S. Gillies
- Department of Surgery and Sepsis and Critical Illness Research Center, University of Florida College of Medicine, Gainesville, Florida
| | - Kolenkode B. Kannan
- Department of Surgery and Sepsis and Critical Illness Research Center, University of Florida College of Medicine, Gainesville, Florida
| | - Valerie E. Polcz
- Department of Surgery and Sepsis and Critical Illness Research Center, University of Florida College of Medicine, Gainesville, Florida
| | - Jeremy A. Balch
- Department of Surgery and Sepsis and Critical Illness Research Center, University of Florida College of Medicine, Gainesville, Florida
| | - Evan L. Barrios
- Department of Surgery and Sepsis and Critical Illness Research Center, University of Florida College of Medicine, Gainesville, Florida
| | - Shannon M. Wallet
- Department of Oral Biology, University of Florida College of Medicine, Gainesville, Florida
| | - Letitia E. Bible
- Department of Surgery and Sepsis and Critical Illness Research Center, University of Florida College of Medicine, Gainesville, Florida
| | - Philip A. Efron
- Department of Surgery and Sepsis and Critical Illness Research Center, University of Florida College of Medicine, Gainesville, Florida
| | - Robert Maile
- Department of Surgery and Sepsis and Critical Illness Research Center, University of Florida College of Medicine, Gainesville, Florida
| | - Alicia M. Mohr
- Department of Surgery and Sepsis and Critical Illness Research Center, University of Florida College of Medicine, Gainesville, Florida
| |
Collapse
|
25
|
Chen Z, Zhang J, Pan Y, Hao Z, Li S. Extracellular vesicles as carriers for noncoding RNA-based regulation of macrophage/microglia polarization: an emerging candidate regulator for lung and traumatic brain injuries. Front Immunol 2024; 15:1343364. [PMID: 38558799 PMCID: PMC10978530 DOI: 10.3389/fimmu.2024.1343364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 02/22/2024] [Indexed: 04/04/2024] Open
Abstract
Macrophage/microglia function as immune defense and homeostatic cells that originate from bone marrow progenitor cells. Macrophage/microglia activation is historically divided into proinflammatory M1 or anti-inflammatory M2 states based on intracellular dynamics and protein production. The polarization of macrophages/microglia involves a pivotal impact in modulating the development of inflammatory disorders, namely lung and traumatic brain injuries. Recent evidence indicates shared signaling pathways in lung and traumatic brain injuries, regulated through non-coding RNAs (ncRNAs) loaded into extracellular vesicles (EVs). This packaging protects ncRNAs from degradation. These vesicles are subcellular components released through a paracellular mechanism, constituting a group of nanoparticles that involve exosomes, microvesicles, and apoptotic bodies. EVs are characterized by a double-layered membrane and are abound with proteins, nucleic acids, and other bioactive compounds. ncRNAs are RNA molecules with functional roles, despite their absence of coding capacity. They actively participate in the regulation of mRNA expression and function through various mechanisms. Recent studies pointed out that selective packaging of ncRNAs into EVs plays a role in modulating distinct facets of macrophage/microglia polarization, under conditions of lung and traumatic brain injuries. This study will explore the latest findings regarding the role of EVs in the progression of lung and traumatic brain injuries, with a specific focus on the involvement of ncRNAs within these vesicles. The conclusion of this review will emphasize the clinical opportunities presented by EV-ncRNAs, underscoring their potential functions as both biomarkers and targets for therapeutic interventions.
Collapse
Affiliation(s)
- Zhihong Chen
- Department of Respiratory Medicine, The Third People’s Hospital of Longgang District, Shenzhen, China
| | - Jingang Zhang
- Department of Orthopedic, The Third People’s Hospital of Longgang District, Shenzhen, China
| | - Yongli Pan
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Zhongnan Hao
- Department of Neurology, University Medical Center of Göttingen, Georg-August-University of Göttingen, Göttingen, Lower Saxony, Germany
| | - Shuang Li
- Department of Respiratory Medicine, The Third People’s Hospital of Longgang District, Shenzhen, China
| |
Collapse
|
26
|
Zhang X, Cheng Z, Zeng M, He Z. The efficacy of extracellular vesicles for acute lung injury in preclinical animal models: a meta-analysis. BMC Pulm Med 2024; 24:128. [PMID: 38481171 PMCID: PMC10935944 DOI: 10.1186/s12890-024-02910-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 02/15/2024] [Indexed: 03/17/2024] Open
Abstract
BACKGROUND With the increasing research on extracellular vesicles (EVs), EVs have received widespread attention as biodiagnostic markers and therapeutic agents for a variety of diseases. Stem cell-derived EVs have also been recognized as a new viable therapy for acute lung injury (ALI) and acute respiratory distress syndrome (ARDS). To assess their efficacy, we conducted a meta-analysis of existing preclinical experimental animal models of EVs for ALI treatment. METHODS The database was systematically interrogated for pertinent data encompassing the period from January 2010 to April 2022 concerning interventions involving extracellular vesicles (EVs) in animal models of acute lung injury (ALI). The lung injury score was selected as the primary outcome measure for statistical analysis. Meta-analyses were executed utilizing RevMan 5.3 and State15.1 software tools. RESULTS The meta-analyses comprised 31 studies, exclusively involving animal models of acute lung injury (ALI), categorized into two cohorts based on the presence or absence of extracellular vesicle (EV) intervention. The statistical outcomes from these two study groups revealed a significant reduction in lung injury scores with the administration of stem and progenitor cell-derived EVs (SMD = -3.63, 95% CI [-4.97, -2.30], P < 0.05). Conversely, non-stem cell-derived EVs were associated with an elevation in lung injury scores (SMD = -4.34, 95% CI [3.04, 5.63], P < 0.05). EVs originating from stem and progenitor cells demonstrated mitigating effects on alveolar neutrophil infiltration, white blood cell counts, total cell counts in bronchoalveolar lavage fluid (BALF), lung wet-to-dry weight ratios (W/D), and total protein in BALF. Furthermore, pro-inflammatory mediators exhibited down-regulation, while anti-inflammatory mediators demonstrated up-regulation. Conversely, non-stem cell-derived EVs exacerbated lung injury. CONCLUSION In preclinical animal models of acute lung injury (ALI), the administration of extracellular vesicles (EVs) originating from stem and progenitor cells demonstrably enhances pulmonary function. This ameliorative effect is attributed to the mitigation of pulmonary vascular permeability and the modulation of immune homeostasis, collectively impeding the progression of inflammation. In stark contrast, the utilization of EVs derived from non-stem progenitor cells exacerbates the extent of lung injury. These findings substantiate the potential utility of EVs as a novel therapeutic avenue for addressing acute lung injury.
Collapse
Affiliation(s)
- Xuefeng Zhang
- The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zongyong Cheng
- The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Menghao Zeng
- The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhihui He
- Department of Critical Care Medicine, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China.
- 138 Tongzibo Road, Yuelu District, Changsha, Hunan, 410013, China.
| |
Collapse
|
27
|
Zhou BW, Liu HM, Xu F, Jia XH. The role of macrophage polarization and cellular crosstalk in the pulmonary fibrotic microenvironment: a review. Cell Commun Signal 2024; 22:172. [PMID: 38461312 PMCID: PMC10924385 DOI: 10.1186/s12964-024-01557-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 03/03/2024] [Indexed: 03/11/2024] Open
Abstract
Pulmonary fibrosis (PF) is a progressive interstitial inflammatory disease with a high mortality rate. Patients with PF commonly experience a chronic dry cough and progressive dyspnoea for years without effective mitigation. The pathogenesis of PF is believed to be associated with dysfunctional macrophage polarization, fibroblast proliferation, and the loss of epithelial cells. Thus, it is of great importance and necessity to explore the interactions among macrophages, fibroblasts, and alveolar epithelial cells in lung fibrosis, as well as in the pro-fibrotic microenvironment. In this review, we discuss the latest studies that have investigated macrophage polarization and activation of non-immune cells in the context of PF pathogenesis and progression. Next, we discuss how profibrotic cellular crosstalk is promoted in the PF microenvironment by multiple cytokines, chemokines, and signalling pathways. And finally, we discuss the potential mechanisms of fibrogenesis development and efficient therapeutic strategies for the disease. Herein, we provide a comprehensive summary of the vital role of macrophage polarization in PF and its profibrotic crosstalk with fibroblasts and alveolar epithelial cells and suggest potential treatment strategies to target their cellular communication in the microenvironment.
Collapse
Affiliation(s)
- Bo-Wen Zhou
- The First School of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Hua-Man Liu
- Department of General Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Fei Xu
- Department of Pneumology and Critical Care Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Xin-Hua Jia
- Department of Pneumology and Critical Care Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China.
| |
Collapse
|
28
|
Cao M, Shi M, Zhou B, Jiang H. An overview of the mechanisms and potential roles of extracellular vesicles in septic shock. Front Immunol 2024; 14:1324253. [PMID: 38343439 PMCID: PMC10853337 DOI: 10.3389/fimmu.2023.1324253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 12/29/2023] [Indexed: 02/15/2024] Open
Abstract
Septic shock, a subset of sepsis, is a fatal condition associated with high morbidity and mortality. However, the pathophysiology of septic shock is not fully understood. Moreover, the diagnostic markers employed for identifying septic shock lack optimal sensitivity and specificity. Current treatment protocols for septic shock have not been effective in lowering the mortality rate of patients. Most cells exhibit the capability to release extracellular vesicles (EVs), nanoscale vesicles that play a vital role in intercellular communication. In recent years, researchers have investigated the potential role of EVs in the pathogenesis, diagnosis, and treatment of different diseases, such as oncological, neurological, and cardiovascular diseases, as well as diabetes and septic shock. In this article, we present an overview of the inhibitory and facilitative roles that EVs play in the process of septic shock, the potential role of EVs in the diagnosis of septic shock, and the potential therapeutic applications of both native and engineered EVs in the management of septic shock.
Collapse
Affiliation(s)
- Meiling Cao
- Department of Neonatology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Mingyue Shi
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Boru Zhou
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Hongkun Jiang
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
29
|
Pei X, Liu L, Wang J, Guo C, Li Q, Li J, Ren Q, Ma R, Zheng Y, Zhang Y, Liu L, Zheng D, Wang P, Jiang P, Feng X, Jiang E, Wang Y, Feng S. Exosomal secreted SCIMP regulates communication between macrophages and neutrophils in pneumonia. Nat Commun 2024; 15:691. [PMID: 38263143 PMCID: PMC10805922 DOI: 10.1038/s41467-024-44714-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 01/02/2024] [Indexed: 01/25/2024] Open
Abstract
In pneumonia, the deficient or delayed pathogen clearance can lead to pathogen proliferation and subsequent overactive immune responses, inducing acute lung injury (ALI). While screening human genome coding genes using our peripheral blood cell chemotactic platform, we unexpectedly find SLP adaptor and CSK interacting membrane protein (SCIMP), a protein with neutrophil chemotactic activity secreted during ALI. However, the specific role of SCIMP in ALI remains unclear. In this study, we investigate the secretion of SCIMP in exosomes (SCIMPexo) by macrophages after bacterial stimulation, both in vitro and in vivo. We observe a significant increase in the levels of SCIMPexo in bronchoalveolar lavage fluid and serum of pneumonia patients. We also find that bronchial perfusion with SCIMPexo or SCIMP N-terminal peptides increases the survival rate of the ALI model. This occurs due to the chemoattraction and activation of peripheral neutrophils dependent on formyl peptide receptor 1/2 (FPR1/2). Conversely, exosome suppressors and FPR1/2 antagonists decrease the survival rate in the lethal ALI model. Scimp-deficient and Fpr1/2-deficient mice also have lower survival rates and shorter survival times than wild-type mice. However, bronchial perfusion of SCIMP rescues Scimp-deficient mice but not Fpr1/2-deficient mice. Collectively, our findings suggest that the macrophage-SCIMP-FPRs-neutrophil axis plays a vital role in the innate immune process underlying ALI.
Collapse
Affiliation(s)
- Xiaolei Pei
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Hematopoietic Stem Cell Transplantation Center, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, P. R. China.
- Tianjin Institutes of Health Science, Tianjin, 301600, P. R. China.
| | - Li Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Hematopoietic Stem Cell Transplantation Center, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, P. R. China
- Tianjin Institutes of Health Science, Tianjin, 301600, P. R. China
| | - Jieru Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Hematopoietic Stem Cell Transplantation Center, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, P. R. China
- Tianjin Institutes of Health Science, Tianjin, 301600, P. R. China
| | - Changyuan Guo
- Department of Immunology, School of Basic Medical Sciences and NHC Key Laboratory of Medical Immunology, Peking University, Beijing, 100191, P. R. China
| | - Qingqing Li
- Department of Immunology, School of Basic Medical Sciences and NHC Key Laboratory of Medical Immunology, Peking University, Beijing, 100191, P. R. China
| | - Jia Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Hematopoietic Stem Cell Transplantation Center, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, P. R. China
- Tianjin Institutes of Health Science, Tianjin, 301600, P. R. China
| | - Qian Ren
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Hematopoietic Stem Cell Transplantation Center, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, P. R. China
- Tianjin Institutes of Health Science, Tianjin, 301600, P. R. China
| | - Runzhi Ma
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Hematopoietic Stem Cell Transplantation Center, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, P. R. China
- Tianjin Institutes of Health Science, Tianjin, 301600, P. R. China
| | - Yi Zheng
- Department of Immunology, School of Basic Medical Sciences and NHC Key Laboratory of Medical Immunology, Peking University, Beijing, 100191, P. R. China
| | - Yan Zhang
- Department of Immunology, School of Basic Medical Sciences and NHC Key Laboratory of Medical Immunology, Peking University, Beijing, 100191, P. R. China
| | - Li Liu
- Tianjin First Central Hospital, Tianjin Medical University, Tianjin, 300192, P. R. China
| | - Danfeng Zheng
- Department of Immunology, School of Basic Medical Sciences and NHC Key Laboratory of Medical Immunology, Peking University, Beijing, 100191, P. R. China
| | - Pingzhang Wang
- Department of Immunology, School of Basic Medical Sciences and NHC Key Laboratory of Medical Immunology, Peking University, Beijing, 100191, P. R. China
| | - Ping Jiang
- Tianjin First Central Hospital, Tianjin Medical University, Tianjin, 300192, P. R. China
| | - Xiaoming Feng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Hematopoietic Stem Cell Transplantation Center, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, P. R. China
- Tianjin Institutes of Health Science, Tianjin, 301600, P. R. China
| | - Erlie Jiang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Hematopoietic Stem Cell Transplantation Center, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, P. R. China
- Tianjin Institutes of Health Science, Tianjin, 301600, P. R. China
| | - Ying Wang
- Department of Immunology, School of Basic Medical Sciences and NHC Key Laboratory of Medical Immunology, Peking University, Beijing, 100191, P. R. China.
| | - Sizhou Feng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Hematopoietic Stem Cell Transplantation Center, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, P. R. China.
- Tianjin Institutes of Health Science, Tianjin, 301600, P. R. China.
| |
Collapse
|
30
|
Huang Q, Le Y, Li S, Bian Y. Signaling pathways and potential therapeutic targets in acute respiratory distress syndrome (ARDS). Respir Res 2024; 25:30. [PMID: 38218783 PMCID: PMC10788036 DOI: 10.1186/s12931-024-02678-5] [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: 09/30/2023] [Accepted: 01/03/2024] [Indexed: 01/15/2024] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a common condition associated with critically ill patients, characterized by bilateral chest radiographical opacities with refractory hypoxemia due to noncardiogenic pulmonary edema. Despite significant advances, the mortality of ARDS remains unacceptably high, and there are still no effective targeted pharmacotherapeutic agents. With the outbreak of coronavirus disease 19 worldwide, the mortality of ARDS has increased correspondingly. Comprehending the pathophysiology and the underlying molecular mechanisms of ARDS may thus be essential to developing effective therapeutic strategies and reducing mortality. To facilitate further understanding of its pathogenesis and exploring novel therapeutics, this review provides comprehensive information of ARDS from pathophysiology to molecular mechanisms and presents targeted therapeutics. We first describe the pathogenesis and pathophysiology of ARDS that involve dysregulated inflammation, alveolar-capillary barrier dysfunction, impaired alveolar fluid clearance and oxidative stress. Next, we summarize the molecular mechanisms and signaling pathways related to the above four aspects of ARDS pathophysiology, along with the latest research progress. Finally, we discuss the emerging therapeutic strategies that show exciting promise in ARDS, including several pharmacologic therapies, microRNA-based therapies and mesenchymal stromal cell therapies, highlighting the pathophysiological basis and the influences on signal transduction pathways for their use.
Collapse
Affiliation(s)
- Qianrui Huang
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095, Jie Fang Avenue, Wuhan, 430030, China
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jie Fang Avenue, Wuhan, 430030, China
| | - Yue Le
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjia Bridge, Hunan Road, Gu Lou District, Nanjing, 210009, China
| | - Shusheng Li
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095, Jie Fang Avenue, Wuhan, 430030, China.
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jie Fang Avenue, Wuhan, 430030, China.
| | - Yi Bian
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095, Jie Fang Avenue, Wuhan, 430030, China.
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jie Fang Avenue, Wuhan, 430030, China.
| |
Collapse
|
31
|
Gao J, Chen H, Zhang Y, Yu S, Wu Y, Wang Q, Yu Z. METTL14 knockdown inhibits the pyroptosis in the sepsis-induced acute lung injury through regulating the m6A modification of NLRP3. Exp Lung Res 2023; 49:220-230. [PMID: 38047519 DOI: 10.1080/01902148.2023.2288182] [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: 08/12/2023] [Accepted: 11/17/2023] [Indexed: 12/05/2023]
Abstract
Background: Sepsis has become one of the main factors inducing the development of acute lung injury (ALI) in clinical practice. Currently, inhibiting the activation of NLRP3 mediated pyroptosis is the target of multiple drugs in the treatment of sepsis induced ALI. This study aimed to explore the effects of METTL14 on the pyroptosis in the sepsis induced ALI progression.Methods: LPS-stimulated A549 cells and cecal ligation and puncture (CLP)-treated mice were used to establish the ALI model in vitro and in vivo. Then, the cell viability was measured by CCK-8 assay. ELISA kits were used to determine the IL-18 and IL-1β contents. Pyroptosis rate was tested by flow cytometry. M6A dot blot was conducted to analyze the global m6A levels and MeRIP assay was performed to detect the m6A levels of NLRP3. The relationship between METTL14 and NLRP3 was confirmed by RIP and dual-luciferase report assays.Results: The global m6A levels were significantly increased in the LPS-stimulated A549 cells and CLP-treated mice. METTL14 knockdown decreased the cell viability, IL-18 and IL-1β contents, and pyroptosis rate of the LPS-stimulated A549 cells. Furthermore, the increase of pyroptosis-related proteins in LPS-stimulated A549 cells was significantly decreased after METTL14 knockdown. Additionally, METTL14 knockdown decreased the m6A and mRNA levels of NLRP3, and NLRP3 overexpression reversed the effects of METTL14 knockdown on the pyroptosis in the LPS-stimulated A549 cells. In CLP-treated mice, METTL14 knockdown relieved the injury and decreased the IL-18 and IL-1β contents in the lung tissues, serum and bronchoalveolar lavage fluid.Conclusion: This study demonstrated that METTL14 knockdown inhibited the pyroptosis in the sepsis-induced ALI progression through decreasing the NLRP3 levels dependent on m6A methylation modification.
Collapse
Affiliation(s)
- Jianting Gao
- Department of Intensive Care Unit, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Huizhen Chen
- Department of Intensive Care Unit, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yong Zhang
- Department of Geriatric Medicine, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Sun Yu
- Department of EICU, Changshu Hospital Affiliated to Soochow University, Changshu, Jiangsu, China
| | - Yiyi Wu
- Department of Intensive Care Unit, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Qiuyan Wang
- Department of Intensive Care Unit, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Zhenfei Yu
- Department of Intensive Care Unit, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| |
Collapse
|
32
|
Xu Y, Zhang C, Cai D, Zhu R, Cao Y. Exosomal miR-155-5p drives widespread macrophage M1 polarization in hypervirulent Klebsiella pneumoniae-induced acute lung injury via the MSK1/p38-MAPK axis. Cell Mol Biol Lett 2023; 28:92. [PMID: 37953267 PMCID: PMC10641976 DOI: 10.1186/s11658-023-00505-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 10/24/2023] [Indexed: 11/14/2023] Open
Abstract
BACKGROUND Hypervirulent Klebsiella pneumoniae (hvKp) infection-induced sepsis-associated acute lung injury (ALI) has emerged as a significant clinical challenge. Increasing evidence suggests that activated inflammatory macrophages contribute to tissue damage in sepsis. However, the underlying causes of widespread macrophage activation remain unclear. METHODS BALB/c mice were intravenously injected with inactivated hvKp (iHvKp) to observe lung tissue damage, inflammation, and M1 macrophage polarization. In vitro, activated RAW264.7 macrophage-derived exosomes (iHvKp-exo) were isolated and their role in ALI formation was investigated. RT-PCR was conducted to identify changes in exosomal miRNA. Bioinformatics analysis and dual-luciferase reporter assays were performed to validate MSK1 as a direct target of miR-155-5p. Further in vivo and in vitro experiments were conducted to explore the specific mechanisms involved. RESULTS iHvKp successfully induced ALI in vivo and upregulated the expression of miR-155-5p. In vivo, injection of iHvKp-exo induced inflammatory tissue damage and macrophage M1 polarization. In vitro, iHvKp-exo was found to promote macrophage inflammatory response and M1 polarization through the activation of the p38-MAPK pathway. RT-PCR revealed exposure time-dependent increased levels of miR-155-5p in iHvKp-exo. Dual-luciferase reporter assays confirmed the functional role of miR-155-5p in mediating iHvKp-exo effects by targeting MSK1. Additionally, inhibition of miR-155-5p reduced M1 polarization of lung macrophages in vivo, resulting in decreased lung injury and inflammation induced by iHvKp-exo or iHvKp. CONCLUSIONS The aforementioned results indicate that exosomal miR-155-5p drives widespread macrophage inflammation and M1 polarization in hvKp-induced ALI through the MSK1/p38-MAPK Axis.
Collapse
Affiliation(s)
- Yihan Xu
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, Fuzhou, 350001, People's Republic of China
| | - Chunying Zhang
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, Fuzhou, 350001, People's Republic of China
| | - Danni Cai
- Central Laboratory, Fujian Medical University Union Hospital, Fuzhou, 350001, People's Republic of China
| | - Rongping Zhu
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, Fuzhou, 350001, People's Republic of China
| | - Yingping Cao
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, Fuzhou, 350001, People's Republic of China.
| |
Collapse
|
33
|
Jin X, Sun H, Yang L. How Extracellular Nano-Vesicles Can Play a Role in Sepsis? An Evidence-Based Review of the Literature. Int J Nanomedicine 2023; 18:5797-5814. [PMID: 37869065 PMCID: PMC10588718 DOI: 10.2147/ijn.s427116] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 10/08/2023] [Indexed: 10/24/2023] Open
Abstract
Sepsis is a systemic inflammatory reaction caused by infection. Severe sepsis can lead to multiple organ dysfunction, with a high incidence rate and mortality. The molecular pathogenesis of sepsis is complex and diverse. In recent years, with further study of the role of extracellular vesicles (EVs) in inflammatory diseases, it has been found that EVs play a dual role in the imbalance of inflammatory response in sepsis. Due to the great advantages such as lower toxicity, lower immunogenicity compared with stem cells and better circulation stability, EVs are increasingly used for the diagnosis and treatment of sepsis. The roles of EVs in the pathogenesis, diagnosis and treatment of sepsis were summarized to guide further clinical studies.
Collapse
Affiliation(s)
- Xiaolin Jin
- Department of International Physical Examination Center, The First Hospital of China Medical University, Shengyang, People’s Republic of China
| | - Haiyan Sun
- Department of Endodontics, School of Stomatology, China Medical University, Shenyang, People’s Republic of China
| | - Lina Yang
- Department of International Physical Examination Center, The First Hospital of China Medical University, Shengyang, People’s Republic of China
- Department of Geriatrics, The First Hospital of China Medical University, Shenyang, People’s Republic of China
| |
Collapse
|
34
|
Lan B, Dong X, Yang Q, Luo Y, Wen H, Chen Z, Chen H. Exosomal MicroRNAs: An Emerging Important Regulator in Acute Lung Injury. ACS OMEGA 2023; 8:35523-35537. [PMID: 37810708 PMCID: PMC10551937 DOI: 10.1021/acsomega.3c04955] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 09/11/2023] [Indexed: 10/10/2023]
Abstract
Acute lung injury (ALI) is a clinically life-threatening form of respiratory failure with a mortality of 30%-40%. Acute respiratory distress syndrome is the aggravated form of ALI. Exosomes are extracellular lipid vesicles ubiquitous in human biofluids with a diameter of 30-150 nm. They can serve as carriers to convey their internal cargo, particularly microRNA (miRNA), to the target cells involved in cellular communication. In disease states, the quantities of exosomes and the cargo generated by cells are altered. These exosomes subsequently function as autocrine or paracrine signals to nearby or distant cells, regulating various pathogenic processes. Moreover, exosomal miRNAs from multiple stem cells can provide therapeutic value for ALI by regulating different signaling pathways. In addition, changes in exosomal miRNAs of biofluids can serve as biomarkers for the early diagnosis of ALI. This study aimed to review the role of exosomal miRNAs produced by different sources participating in various pathological processes of ALI and explore their potential significance in the treatment and diagnosis.
Collapse
Affiliation(s)
- Bowen Lan
- Department
of General Surgery, The First Affiliated
Hospital of Dalian Medical University, Dalian 116000, China
- Laboratory
of Integrative Medicine, The First Affiliated
Hospital of Dalian Medical University, Dalian 116000, China
| | - Xuanchi Dong
- Department
of General Surgery, The First Affiliated
Hospital of Dalian Medical University, Dalian 116000, China
- Laboratory
of Integrative Medicine, The First Affiliated
Hospital of Dalian Medical University, Dalian 116000, China
| | - Qi Yang
- Department
of General Surgery, The First Affiliated
Hospital of Dalian Medical University, Dalian 116000, China
- Laboratory
of Integrative Medicine, The First Affiliated
Hospital of Dalian Medical University, Dalian 116000, China
- Department
of Traditional Chinese Medicine, The Second
Affiliated Hospital of Dalian Medical University, Dalian 116023, China
| | - Yalan Luo
- Department
of General Surgery, The First Affiliated
Hospital of Dalian Medical University, Dalian 116000, China
- Laboratory
of Integrative Medicine, The First Affiliated
Hospital of Dalian Medical University, Dalian 116000, China
- Institute
(College) of Integrative Medicine, Dalian
Medical University, Dalian 116044, China
| | - Haiyun Wen
- Department
of General Surgery, The First Affiliated
Hospital of Dalian Medical University, Dalian 116000, China
- Laboratory
of Integrative Medicine, The First Affiliated
Hospital of Dalian Medical University, Dalian 116000, China
- Institute
(College) of Integrative Medicine, Dalian
Medical University, Dalian 116044, China
| | - Zhe Chen
- Department
of General Surgery, The First Affiliated
Hospital of Dalian Medical University, Dalian 116000, China
- Laboratory
of Integrative Medicine, The First Affiliated
Hospital of Dalian Medical University, Dalian 116000, China
| | - Hailong Chen
- Department
of General Surgery, The First Affiliated
Hospital of Dalian Medical University, Dalian 116000, China
- Laboratory
of Integrative Medicine, The First Affiliated
Hospital of Dalian Medical University, Dalian 116000, China
- Institute
(College) of Integrative Medicine, Dalian
Medical University, Dalian 116044, China
| |
Collapse
|
35
|
Xu L, Chen Y, Feng S, Liu Z, Ye Y, Zhou R, Liu L. PEDF inhibits LPS-induced acute lung injury in rats and promotes lung epithelial cell survival by upregulating PPAR-γ. BMC Pulm Med 2023; 23:359. [PMID: 37740176 PMCID: PMC10517507 DOI: 10.1186/s12890-023-02666-3] [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: 01/20/2023] [Accepted: 09/19/2023] [Indexed: 09/24/2023] Open
Abstract
BACKGROUND The progression of acute lung injury (ALI) involves numerous pathological factors and complex mechanisms, and cause the destruction of epithelial and endothelial barriers. Pigment epithelium-derived factor (PEDF) is an angiogenesis inhibitor and a potential anti-inflammatory factor. The purpose of this study was to investigate the effect of PEDF on lipopolysaccharide (LPS)-induced ALI in rats. METHODS In vivo, pathological and injury related factors examination were performed on rat lung to investigate the effect of PEDF on ALI. In vitro, the effect of PEDF on inflammatory injury and apoptosis of lung epithelial type II RLE-6TN cell was evaluated, and the expression of inflammatory factors and related pathway proteins and PPAR-γ (in the presence or absence of PPAR-γ inhibitors) were analyzed. RESULTS In vivo results showed that PEDF inhibited the inflammatory factor expression (TNF-α, IL-6 and IL-1β) and progression of ALI and reduced lung cell apoptosis in rats. In vitro results showed that PEDF could effectively inhibit LPS-stimulated inflammatory damage and apoptosis of RLE-6TN cells. PEDF inhibited the RLE-6TN cell injury by enhancing the expression of PPAR-γ. CONCLUSIONS PEDF is an anti-inflammatory factor, which can inhibit apoptosis of lung epithelial cells by upregulating the expression of PPAR-γ and reducing LPS-induced ALI in rats.
Collapse
Affiliation(s)
- Lei Xu
- Department of Emergency Medicine, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China
- Department of Emergency Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China
| | - Yifei Chen
- Department of Emergency Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou, 225001, Jiangsu, China
| | - Shoujie Feng
- Thoracic Surgery Laboratory, Xuzhou Medical University, Xuzhou, China
- Department of Thoracic Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China
| | - Zeyan Liu
- Department of Emergency Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China
| | - Ying Ye
- Department of Emergency Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China
| | - Ranran Zhou
- Department of Emergency Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China
| | - Lijun Liu
- Department of Emergency Medicine, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China.
| |
Collapse
|
36
|
Peng W, Yang Y, Chen J, Xu Z, Lou Y, Li Q, Zhao N, Qian K, Liu F. Small Extracellular Vesicles Secreted by iPSC-Derived MSCs Ameliorate Pulmonary Inflammation and Lung Injury Induced by Sepsis through Delivery of miR-125b-5p. J Immunol Res 2023; 2023:8987049. [PMID: 37425491 PMCID: PMC10329558 DOI: 10.1155/2023/8987049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 12/07/2022] [Accepted: 05/18/2023] [Indexed: 07/11/2023] Open
Abstract
Background Sepsis-induced acute lung injury is a common critical illness in intensive care units with no effective treatment is currently available. Small extracellular vesicles, secreted by mesenchymal stem cells (MSCs), derived from human-induced pluripotent stem cells (iMSC-sEV), possess striking advantages when incorporated MSCs and iPSCs, which are considered extremely promising cell-free therapeutic agents. However, no studies have yet been conducted to systemically examine the effects and underlying mechanisms of iMSC-sEV application on attenuated lung injury under sepsis conditions. Method iMSC-sEV were intraperitoneally administered in a rat septic lung injury model induced by cecal ligation and puncture (CLP). The efficacy of iMSC-sEV was assessed by histology, immunohistochemistry, and pro-inflammatory cytokines of bronchoalveolar lavage fluid. We also evaluated the in vitro effects of iMSC-sEV on the activation of the inflammatory response in alveolar macrophages (AMs). Small RNA sequencing was utilized to detect changes in the miRNA expression profile in lipopolysaccharide (LPS)-treated AMs after iMSC-sEV administration. The effects of miR-125b-5p on the function of AMs were studied. Results iMSC-sEV were able to attenuate pulmonary inflammation and lung injury following CLP-induced lung injury. iMSC-sEV were internalized by AMs and alleviated the release of inflammatory factors by inactivating the NF-κB signaling pathway. Moreover, miR-125b-5p showed a fold-change in LPS-treated AMs after iMSC-sEV administration and was enriched in iMSC-sEV. Mechanistically, iMSC-sEV transmitted miR-125b-5p into LPS-treated AMs to target TRAF6. Conclusion Our findings demonstrated that iMSC-sEV treatment protects against septic lung injury and exerts anti-inflammatory effects on AMs at least partially through miR-125b-5p, suggesting that iMSC-sEV may provide a novel cell-free strategy for the treatment of septic lung injury.
Collapse
Affiliation(s)
- Wei Peng
- Department of Critical Care Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yun Yang
- Department of Critical Care Medicine, The People's Hospital of Fengcheng City, Yichun, Jiangxi, China
| | - Jiaquan Chen
- Department of Critical Care Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Zeyao Xu
- Department of Critical Care Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yuanlei Lou
- Institute of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Qi Li
- Department of Critical Care Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Ning Zhao
- Department of Critical Care Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Kejian Qian
- Department of Critical Care Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Fen Liu
- Department of Critical Care Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
37
|
Xiong C, Huang X, Chen S, Li Y. Role of Extracellular microRNAs in Sepsis-Induced Acute Lung Injury. J Immunol Res 2023; 2023:5509652. [PMID: 37378068 PMCID: PMC10292948 DOI: 10.1155/2023/5509652] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 05/13/2023] [Accepted: 06/05/2023] [Indexed: 06/29/2023] Open
Abstract
Acute lung injury (ALI) is a life-threatening pathological disease characterized by the damage of pulmonary endothelial cells and epithelial cell barriers by uncontrolled inflammation. During sepsis-induced ALI, multiple cells cooperate and communicate with each other to respond to the stimulation of inflammatory factors. However, the underlying mechanisms of action have not been fully identified, and the modes of communication therein are also being investigated. Extracellular vesicles (EVs) are a heterogeneous population of spherical membrane structures released by almost all types of cells, containing various cellular components. EVs are primary transport vehicles for microRNAs (miRNAs), which play essential roles in physiological and pathological processes in ALI. EV miRNAs from different sources participated in regulating the biological function of pulmonary epithelial cells, endothelial cells, and phagocytes by transferring miRNA through EVs during ALI induced by sepsis, which has great potential diagnostic and therapeutic values. This study aims to summarize the role and mechanism of extracellular vesicle miRNAs from different cells in the regulation of sepsis-induced ALI. It provides ideas for further exploring the role of extracellular miRNA secreted by different cells in the ALI induced by sepsis, to make up for the deficiency of current understanding, and to explore the more optimal scheme for diagnosis and treatment of ALI.
Collapse
Affiliation(s)
- Chenlu Xiong
- Department of Anesthesiology, Medical Center of Anesthesiology and Pain, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xuan Huang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, China
| | - Shibiao Chen
- Department of Anesthesiology, Medical Center of Anesthesiology and Pain, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yong Li
- Department of Anesthesiology, Medical Center of Anesthesiology and Pain, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
38
|
Zhang Q, Huang Y, Gong C, Tang Y, Xiong J, Wang D, Liu X. Dexmedetomidine attenuates inflammation and organ injury partially by upregulating Nur77 in sepsis. Immun Inflamm Dis 2023; 11:e883. [PMID: 37382273 PMCID: PMC10283499 DOI: 10.1002/iid3.883] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 05/03/2023] [Accepted: 05/10/2023] [Indexed: 06/30/2023] Open
Abstract
PURPOSE The aim of this study was to investigate the effect of dexmedetomidine (Dex) on inflammation and organ injury in sepsis, as well as the potential relationship between Dex and nuclear receptor 77 (Nur77). METHODS We investigated the effects of dexmedetomidine on lipopolysaccharide (LPS)-induced inflammation in RAW264.7 cells and organ injury in the cecal ligation and puncture (CLP) mouse model. Additionally, we examined the relationship between dexmedetomidine and Nur77. The expression levels of Nur77 in RAW264.7 cells were analyzed under various types of stimulation using quantitative reverse transcription polymerase chain reaction and western blot analysis. Inflammatory cytokine levels in the cells were evaluated using enzyme-linked immunoassay. Organ injuries were assessed by examining tissue histology and pathology of the lung, liver, and kidney. RESULTS Dexmedetomidine increased the expression of Nur77 and IL-10, and downregulated inflammatory cytokines (IL-1β and TNF-α) in LPS-treated RAW264.7 cells. The effect of dexmedetomidine on inhibiting inflammation in LPS-treated RAW264.7 cells was promoted by overexpressing Nur77, while it was reversed by downregulating Nur77. Additionally, dexmedetomidine promoted the expression of Nur77 in the lung and CLP-induced pathological changes in the lung, liver, and kidney. Activation of Nur77 with the agonist Cytosporone B (CsnB) significantly suppressed the production of IL-1β and TNF-α in LPS-treated RAW264.7 cells. In contrast, knockdown of Nur77 augmented IL-1β and TNF-α production in LPS-treated RAW264.7 cells. CONCLUSION Dexmedetomidine can attenuate inflammation and organ injury, at least partially, via upregulating Nur77 in sepsis.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Critical Care MedicineGuizhou Medical UniversityGuiyangGuizhouPeople's Republic of China
| | - Yun Huang
- Department of NephrologyFirst People's HospitalGuiyangGuizhouPeople's Republic of China
| | - Chenchen Gong
- Department of Thoracic and Cardiovascular SurgeryThe Children's Hospital of Zhejiang University School of MedicineZhejiangPeople's Republic of China
| | - Yan Tang
- Department of Critical Care MedicineAffiliated Hospital of Guizhou Medical UniversityGuiyangGuizhouPeople's Republic of China
| | - Jie Xiong
- Department of HematologyThe Affiliated Hospital of Guizhou Medical UniversityGuiyangGuizhouPeople's Republic of China
| | - Difen Wang
- Department of Critical Care MedicineAffiliated Hospital of Guizhou Medical UniversityGuiyangGuizhouPeople's Republic of China
| | - Xu Liu
- Department of Critical Care MedicineAffiliated Hospital of Guizhou Medical UniversityGuiyangGuizhouPeople's Republic of China
| |
Collapse
|
39
|
Lai X, Zhong J, Zhang B, Zhu T, Liao R. Exosomal Non-Coding RNAs: Novel Regulators of Macrophage-Linked Intercellular Communication in Lung Cancer and Inflammatory Lung Diseases. Biomolecules 2023; 13:536. [PMID: 36979471 PMCID: PMC10046066 DOI: 10.3390/biom13030536] [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: 02/07/2023] [Revised: 03/08/2023] [Accepted: 03/13/2023] [Indexed: 03/18/2023] Open
Abstract
Macrophages are innate immune cells and often classified as M1 macrophages (pro-inflammatory states) and M2 macrophages (anti-inflammatory states). Exosomes are cell-derived nanovesicles that range in diameter from 30 to 150 nm. Non-coding RNAs (ncRNAs), including microRNAs (miRNAs), long noncoding RNAs (lncRNAs), and circular RNAs (circRNAs), are abundant in exosomes and exosomal ncRNAs influence immune responses. Exosomal ncRNAs control macrophage-linked intercellular communication via their targets or signaling pathways, which can play positive or negative roles in lung cancer and inflammatory lung disorders, including acute lung injury (ALI), asthma, and pulmonary fibrosis. In lung cancer, exosomal ncRNAs mediated intercellular communication between lung tumor cells and tumor-associated macrophages (TAMs), coordinating cancer proliferation, migration, invasion, metastasis, immune evasion, and therapy resistance. In inflammatory lung illnesses, exosomal ncRNAs mediate macrophage activation and inflammation to promote or inhibit lung damage. Furthermore, we also discussed the possible applications of exosomal ncRNA-based therapies for lung disorders.
Collapse
Affiliation(s)
- Xingning Lai
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China
- Research Unit for Perioperative Stress Assessment and Clinical Decision, Chinese Academy of Medical Sciences (2018RU012), West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jie Zhong
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China
- Research Unit for Perioperative Stress Assessment and Clinical Decision, Chinese Academy of Medical Sciences (2018RU012), West China Hospital, Sichuan University, Chengdu 610041, China
| | - Boyi Zhang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China
- Research Unit for Perioperative Stress Assessment and Clinical Decision, Chinese Academy of Medical Sciences (2018RU012), West China Hospital, Sichuan University, Chengdu 610041, China
| | - Tao Zhu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China
- Research Unit for Perioperative Stress Assessment and Clinical Decision, Chinese Academy of Medical Sciences (2018RU012), West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ren Liao
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China
- Research Unit for Perioperative Stress Assessment and Clinical Decision, Chinese Academy of Medical Sciences (2018RU012), West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
40
|
Weber B, Henrich D, Hildebrand F, Marzi I, Leppik L. THE ROLES OF EXTRACELLULAR VESICLES IN SEPSIS AND SYSTEMIC INFLAMMATORY RESPONSE SYNDROME. Shock 2023; 59:161-172. [PMID: 36730865 PMCID: PMC9940838 DOI: 10.1097/shk.0000000000002010] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 08/29/2022] [Accepted: 10/05/2022] [Indexed: 02/04/2023]
Abstract
ABSTRACT Sepsis is a life-threatening organ dysfunction, caused by dysregulation of the host response to infection. To understand the underlying mechanisms of sepsis, the vast spectrum of extracellular vesicles (EVs) is gaining importance in this research field. A connection between EVs and sepsis was shown in 1998 in an endotoxemia pig model. Since then, the number of studies describing EVs as markers and mediators of sepsis increased steadily. Extracellular vesicles in sepsis could be friends and foes at the same time depending on their origin and cargo. On the one hand, transfer of EVs or outer membrane vesicles can induce sepsis or systemic inflammatory response syndrome with comparable efficiency as well-established methods, such as cecal ligation puncture or lipopolysaccharide injection. On the other hand, EVs could provide certain therapeutic effects, mediated via reduction of reactive oxygen species, inflammatory cytokines and chemokines, influence on macrophage polarization and apoptosis, as well as increase of anti-inflammatory cytokines. Moreover, EVs could be helpful in the diagnosis of sepsis. Extracellular vesicles of different cellular origin, such as leucocytes, macrophages, platelets, and granulocytes, have been suggested as potential sepsis biomarkers. They ensure the diagnosis of sepsis earlier than classical clinical inflammation markers, such as C-reactive protein, leucocytes, or IL-6. This review summarizes the three roles of EVs in sepsis-mediator/inducer, biomarker, and therapeutic tool.
Collapse
Affiliation(s)
- Birte Weber
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Frankfurt, Goethe-University, Frankfurt am Main, Germany
| | - Dirk Henrich
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Frankfurt, Goethe-University, Frankfurt am Main, Germany
| | - Frank Hildebrand
- Department of Trauma and Reconstructive Surgery, University Hospital RWTH Aachen. Aachen, Germany
| | - Ingo Marzi
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Frankfurt, Goethe-University, Frankfurt am Main, Germany
| | - Liudmila Leppik
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Frankfurt, Goethe-University, Frankfurt am Main, Germany
| |
Collapse
|
41
|
Soto-Vázquez YM, Genschmer KR. Impact of extracellular vesicles on the pathogenesis, diagnosis, and potential therapy in cardiopulmonary disease. Front Pharmacol 2023; 14:1081015. [PMID: 36891265 PMCID: PMC9986338 DOI: 10.3389/fphar.2023.1081015] [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: 10/26/2022] [Accepted: 02/08/2023] [Indexed: 02/22/2023] Open
Abstract
Cardiopulmonary diseases span a wide breadth of conditions affecting both heart and lung, the burden of which is globally significant. Chronic pulmonary disease and cardiovascular disease are two of the leading causes of morbidity and mortality worldwide. This makes it critical to understand disease pathogenesis, thereby providing new diagnostic and therapeutic avenues to improve clinical outcomes. Extracellular vesicles provide insight into all three of these features of the disease. Extracellular vesicles are membrane-bound vesicles released by a multitude, if not all, cell types and are involved in multiple physiological and pathological processes that play an important role in intercellular communication. They can be isolated from bodily fluids, such as blood, urine, and saliva, and their contents include a variety of proteins, proteases, and microRNA. These vesicles have shown to act as effective transmitters of biological signals within the heart and lung and have roles in the pathogenesis and diagnosis of multiple cardiopulmonary diseases as well as demonstrate potential as therapeutic agents to treat said conditions. In this review article, we will discuss the role these extracellular vesicles play in the diagnosis, pathogenesis, and therapeutic possibilities of cardiovascular, pulmonary, and infection-related cardiopulmonary diseases.
Collapse
Affiliation(s)
- Yixel M Soto-Vázquez
- Department of Medicine, Division of Pulmonary, Allergy & Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Kristopher R Genschmer
- Department of Medicine, Division of Pulmonary, Allergy & Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
42
|
Hu Q, Zhang S, Yang Y, Yao JQ, Tang WF, Lyon CJ, Hu TY, Wan MH. Extracellular vesicles in the pathogenesis and treatment of acute lung injury. Mil Med Res 2022; 9:61. [PMID: 36316787 PMCID: PMC9623953 DOI: 10.1186/s40779-022-00417-9] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 09/19/2022] [Indexed: 11/05/2022] Open
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are common life-threatening lung diseases associated with acute and severe inflammation. Both have high mortality rates, and despite decades of research on clinical ALI/ARDS, there are no effective therapeutic strategies. Disruption of alveolar-capillary barrier integrity or activation of inflammatory responses leads to lung inflammation and injury. Recently, studies on the role of extracellular vesicles (EVs) in regulating normal and pathophysiologic cell activities, including inflammation and injury responses, have attracted attention. Injured and dysfunctional cells often secrete EVs into serum or bronchoalveolar lavage fluid with altered cargoes, which can be used to diagnose and predict the development of ALI/ARDS. EVs secreted by mesenchymal stem cells can also attenuate inflammatory reactions associated with cell dysfunction and injury to preserve or restore cell function, and thereby promote cell proliferation and tissue regeneration. This review focuses on the roles of EVs in the pathogenesis of pulmonary inflammation, particularly ALI/ARDS.
Collapse
Affiliation(s)
- Qian Hu
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Shu Zhang
- Department of Emergency Medicine, Emergency Medical Laboratory, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Yue Yang
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Jia-Qi Yao
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Wen-Fu Tang
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Christopher J Lyon
- Center of Cellular and Molecular Diagnosis, Tulane University School of Medicine, 1430 Tulane Ave., New Orleans, LA, 70112, USA.,Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, 1430 Tulane Ave., New Orleans, LA, 70112, USA
| | - Tony Ye Hu
- Center of Cellular and Molecular Diagnosis, Tulane University School of Medicine, 1430 Tulane Ave., New Orleans, LA, 70112, USA. .,Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, 1430 Tulane Ave., New Orleans, LA, 70112, USA.
| | - Mei-Hua Wan
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China. .,West China Hospital (Airport) of Sichuan University, Chengdu, 610299, China.
| |
Collapse
|
43
|
Sun G, Zeng Y, Luo F, Zhang L, Tan J, Tong J, Yang L, Liu D, Liu L, Zhou J. Electroacupuncture Preconditioning Alleviates Lipopolysaccharides-Induced Acute Lung Injury by Downregulating LC3-II/I and Beclin 1 Expression. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:8997173. [PMID: 36310624 PMCID: PMC9613389 DOI: 10.1155/2022/8997173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 10/05/2022] [Indexed: 11/15/2022]
Abstract
Our study aimed to investigate the effect of electroacupuncture pretreatment on the inflammatory response and expression levels of LC3-II/I and Beclin 1 using a model of lipopolysaccharide (LPS)-induced acute lung injury (ALI). Eighteen male Sprague-Dawley (SD) rats were randomly divided into three groups: normal control group (NC, n = 6), LSP modeling group (LM, n = 6), and electroacupuncture group (EA, n = 6). Rats in the EA group received electroacupuncture pretreatment at bilateral Zusanli (ST36) and Chize (LU5) points for five days (30 min each time daily, frequency; 3 Hz/15 Hz, intensity; 1 mA). Rats in the EA and LM groups were then injected with 5 mg/kg LPS (Beijing, Solarbio Company, concentration; 5 mg/mL) through the tail vein, while those in the NC group were injected with 5 mg/kg saline. The animals were sacrificed six hours after LPS or saline injection through cervical vertebrae by dislocation under deep anesthesia. Orbital blood was collected for the analysis of serum inflammatory factors including interleukin-1β (IL-1β) and transforming growth factor-β (TGF-β). The lower left lung was excised, stained with hematoxylin-eosin (HE), and subjected to histopathological analysis. The mRNA and protein expression of Beclin 1 and LC3 II/I in the lower right lung tissues were detected via RT-qPCR and Western blot analyses, respectively. The results showed that lung injury score was significantly higher in the LM group than that of the NC group (P < 0.01) and EA group (P < 0.01). The IL-1β contents were significantly decreased in the EA group (P < 0.01) than in the LM group. In contrast, the GF-β contents were increased in the EA group significantly when compared with the LM group (P < 0.01). RT-qPCR and Western blot detection showed that the relative gene expression of LC3-II/I and Beclin 1 was significantly lower in the EA group than in the LM group (P < 0.01). However, the relative protein expression level of LC3-II/I and Beclin 1 was slightly lower in the EA group than the in LM group (P > 0.05). These results show that electroacupuncture pretreatment reduces the inflammatory response in ALI and can protect lung tissue by inhibiting the gene and protein expression levels of LC3-II/I and Beclin 1.
Collapse
Affiliation(s)
- Guanghua Sun
- The First Affiliated Hospital, Department of Rehabilitation, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
- The First Affiliated Hospital, Rehabilitation Medicine Center, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
- The First Affiliated Hospital, Rehabilitation Laboratory, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Yahua Zeng
- The First Affiliated Hospital, Department of Rehabilitation, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
- The First Affiliated Hospital, Rehabilitation Medicine Center, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
- The First Affiliated Hospital, Rehabilitation Laboratory, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Fu Luo
- The First Affiliated Hospital, Department of Rehabilitation, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
- The First Affiliated Hospital, Rehabilitation Medicine Center, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
- The First Affiliated Hospital, Rehabilitation Laboratory, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Lixian Zhang
- Children's Nerve and Development Center, Maternal and Child Health Hospital of Qingyuan City, Qingyuan 511500, Guangdong, China
| | - Jinqu Tan
- The First Affiliated Hospital, Department of Rehabilitation, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
- The First Affiliated Hospital, Rehabilitation Medicine Center, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
- The First Affiliated Hospital, Rehabilitation Laboratory, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Jie Tong
- The First Affiliated Hospital, Department of Rehabilitation, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
- The First Affiliated Hospital, Rehabilitation Medicine Center, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
- The First Affiliated Hospital, Rehabilitation Laboratory, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Lu Yang
- The First Affiliated Hospital, Department of Rehabilitation, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
- The First Affiliated Hospital, Rehabilitation Medicine Center, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
- The First Affiliated Hospital, Rehabilitation Laboratory, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Danni Liu
- The First Affiliated Hospital, Department of Rehabilitation, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
- The First Affiliated Hospital, Rehabilitation Medicine Center, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
- The First Affiliated Hospital, Rehabilitation Laboratory, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Liu Liu
- The First Affiliated Hospital, Department of Rehabilitation, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
- The First Affiliated Hospital, Rehabilitation Medicine Center, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
- The First Affiliated Hospital, Rehabilitation Laboratory, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Jun Zhou
- The First Affiliated Hospital, Department of Rehabilitation, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
- The First Affiliated Hospital, Rehabilitation Medicine Center, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
- The First Affiliated Hospital, Rehabilitation Laboratory, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| |
Collapse
|
44
|
Antonakos N, Gilbert C, Théroude C, Schrijver IT, Roger T. Modes of action and diagnostic value of miRNAs in sepsis. Front Immunol 2022; 13:951798. [PMID: 35990654 PMCID: PMC9389448 DOI: 10.3389/fimmu.2022.951798] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 07/08/2022] [Indexed: 11/13/2022] Open
Abstract
Sepsis is a clinical syndrome defined as a dysregulated host response to infection resulting in life-threatening organ dysfunction. Sepsis is a major public health concern associated with one in five deaths worldwide. Sepsis is characterized by unbalanced inflammation and profound and sustained immunosuppression, increasing patient susceptibility to secondary infections and mortality. microRNAs (miRNAs) play a central role in the control of many biological processes, and deregulation of their expression has been linked to the development of oncological, cardiovascular, neurodegenerative and metabolic diseases. In this review, we discuss the role of miRNAs in sepsis pathophysiology. Overall, miRNAs are seen as promising biomarkers, and it has been proposed to develop miRNA-based therapies for sepsis. Yet, the picture is not so straightforward because of the versatile and dynamic features of miRNAs. Clearly, more research is needed to clarify the expression and role of miRNAs in sepsis, and to promote the use of miRNAs for sepsis management.
Collapse
Affiliation(s)
| | | | | | | | - Thierry Roger
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Epalinges, Switzerland
| |
Collapse
|
45
|
Hu X, Chen M, Cao X, Yuan X, Zhang F, Ding W. TGF-β-Containing Small Extracellular Vesicles From PM2.5-Activated Macrophages Induces Cardiotoxicity. Front Cardiovasc Med 2022; 9:917719. [PMID: 35872905 PMCID: PMC9304575 DOI: 10.3389/fcvm.2022.917719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 06/14/2022] [Indexed: 11/29/2022] Open
Abstract
Numerous epidemiological and experimental studies have demonstrated that the exposure to fine particulate matter (aerodynamic diameter <2.5 μm, PM2.5) was closely associated with cardiovascular morbidity and mortality. Our previous studies revealed that PM2.5 exposure induced cardiac dysfunction and fibrosis. However, the corresponding underlying mechanism remains largely unaddressed. Here, PM2.5-induced cardiotoxicity is presented to directly promote collagen deposition in cardiomyocytes through the transforming growth factor-β (TGF-β)-containing small extracellular vesicles (sEV). The sEV transition may play an important role in PM2.5-induced cardiac fibrosis. Firstly, long-term PM2.5 exposure can directly induce cardiac fibrosis and increase the level of serum sEV. Secondly, PM2.5 can directly activate macrophages and increase the release of tumor necrosis factor α (TNF-α), interleukin-6 (IL-6), and TGF-β-containing sEV. Thirdly, TGF-β-containing sEV increases the expression of α-smooth muscle actin (α-SMA), collagen I, and collagen III in mouse cardiac muscle HL-1 cells. Finally, TGF-β-containing sEV released from PM2.5-treated macrophages can increase collagen through the activation of the TGF-β-Smad2/3 signaling pathway in HL-1 cells from which some fibroblasts involved in cardiac fibrosis are thought to originate. These findings suggest that TGF-β-containing sEV from PM2.5-activated macrophages play a critical role in the process of increasing cardiac collagen content via activating the TGF-β-Smad2/3 signaling pathway.
Collapse
Affiliation(s)
- Xiaoqi Hu
- Laboratory of Environment and Health, College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- Sino-Danish Center for Education and Research, Sino-Danish College, University of Chinese Academy of Sciences, Beijing, China
| | - Mo Chen
- Laboratory of Environment and Health, College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- Sino-Danish Center for Education and Research, Sino-Danish College, University of Chinese Academy of Sciences, Beijing, China
| | - Xue Cao
- Laboratory of Environment and Health, College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Xinyi Yuan
- Laboratory of Environment and Health, College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Fang Zhang
- Laboratory of Environment and Health, College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- Fang Zhang
| | - Wenjun Ding
- Laboratory of Environment and Health, College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- Sino-Danish Center for Education and Research, Sino-Danish College, University of Chinese Academy of Sciences, Beijing, China
- *Correspondence: Wenjun Ding
| |
Collapse
|
46
|
Cai B, Yang L, Do Jung Y, Zhang Y, Liu X, Zhao P, Li J. PTEN: An Emerging Potential Target for Therapeutic Intervention in Respiratory Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4512503. [PMID: 35814272 PMCID: PMC9262564 DOI: 10.1155/2022/4512503] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 04/22/2022] [Accepted: 05/19/2022] [Indexed: 12/13/2022]
Abstract
Phosphatase and tensin homolog deleted on chromosome 10 (PTEN) is a potent tumor suppressor that regulates several key cellular processes, including proliferation, survival, genomic integrity, migration, and invasion, via PI3K-dependent and independent mechanisms. A subtle decrease in PTEN levels or catalytic activity is implicated not only in cancer but also in a wide spectrum of other diseases, including various respiratory diseases. A systemic overview of the advances in the molecular and cellular mechanisms of PTEN involved in the initiation and progression of respiratory diseases may offer novel targets for the development of effective therapeutics for the treatment of respiratory diseases. In the present review, we highlight the novel findings emerging from current research on the role of PTEN expression and regulation in airway pathological conditions such as asthma/allergic airway inflammation, pulmonary hypertension (PAH), chronic obstructive pulmonary disease (COPD), idiopathic pulmonary fibrosis (IPF), and other acute lung injuries (ALI). Moreover, we discuss the clinical implications of PTEN alteration and recently suggested therapeutic possibilities for restoration of PTEN expression and function in respiratory diseases.
Collapse
Affiliation(s)
- Bangrong Cai
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province & Education Ministry of P.R. Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, China
- Henan Research Center for Special Processing Technology of Chinese Medicine, School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Liu Yang
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province & Education Ministry of P.R. Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, China
| | - Young Do Jung
- Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju 501-190, Republic of Korea
| | - Ying Zhang
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xinguang Liu
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province & Education Ministry of P.R. Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, China
| | - Peng Zhao
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province & Education Ministry of P.R. Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, China
| | - Jiansheng Li
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province & Education Ministry of P.R. Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, China
- Department of Respiratory Diseases, the First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, China
| |
Collapse
|
47
|
Wang Y, Xie W, Feng Y, Xu Z, He Y, Xiong Y, Chen L, Li X, Liu J, Liu G, Wu Q. Epithelial‑derived exosomes promote M2 macrophage polarization via Notch2/SOCS1 during mechanical ventilation. Int J Mol Med 2022; 50:96. [PMID: 35616134 PMCID: PMC9170191 DOI: 10.3892/ijmm.2022.5152] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 05/10/2022] [Indexed: 11/24/2022] Open
Abstract
Alveolar macrophages (AMs) play an essential role in ventilator-induced lung injury (VILI). Exosomes and their cargo, including microRNAs (miRNAs/miRs) serve as regulators of the intercellular communications between macrophages and epithelial cells (ECs), and are involved in maintaining homeostasis in lung tissue. The present study found that exosomes released by ECs subjected to cyclic stretching promoted M2 macrophage polarization. It was demonstrated that miR-21a-5p, upregulated in epithelial-derived exosomes, increased the percentage of M2 macrophages by suppressing the expression of Notch2 and the suppressor of cytokine signaling 1 (SOCS1). The overexpression of Notch2 decreased the percentage of M2 macrophages. However, these effects were reversed by the downregulation of SOCS1. The percentage of M2 macrophages was increased in both short-term high- and low-tidal-volume mechanical ventilation, and the administration of exosomes-derived from cyclically stretched ECs had the same function. However, the administration of miR-21a-5p antagomir decreased M2 macrophage activation induced by cyclically stretched ECs or ventilation. Thus, the present study demonstrates that the intercellular transferring of exosomes from ECs to AMs promotes M2 macrophage polarization. Exosomes may prove to be a novel treatment for VILI.
Collapse
Affiliation(s)
- Yanting Wang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Wanli Xie
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Yiqi Feng
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Zhenzhen Xu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Yuyao He
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Yue Xiong
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Lu Chen
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Xia Li
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Jie Liu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Guoyang Liu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Qingping Wu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
48
|
Wang S, Duan Y. LncRNA OIP5-AS1 inhibits the lipopolysaccharide-induced inflammatory response and promotes osteogenic differentiation of human periodontal ligament cells by sponging miR-92a-3p. Bioengineered 2022; 13:12055-12066. [PMID: 35546327 PMCID: PMC9276041 DOI: 10.1080/21655979.2022.2067291] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 04/08/2022] [Accepted: 04/12/2022] [Indexed: 11/12/2022] Open
Abstract
Periodontitis is a chronic infectious disease that affects the oral health of adults. Long non-coding RNA OIP5 antisense RNA 1 (OIP5-AS1) has been reported to downregulated in the periodontal tissue of patients with periodontitis. Therefore, the study sought to look at the possible functions of OIP5-AS1 in periodontitis and the associated underlying mechanisms. In the present study, the expression level of OIP5-AS1 and microRNA-92a-3p were analyzed using reverse transcription-quantitative PCR. The levels of osteogenic proteins were determined using western blotting and inflammatory cytokines and oxidative stress were also examined. The proliferation of human periodontal ligament stem cells (hPDLSCs) was evaluated using MTT assays. Assay of osteogenic differentiation was undertaken by means of Alkaline phosphatase staining. The possible association between OIP5-AS1 and miR-92a-3p was determined applying dual-luciferase reporter assays and verified by RNA immunoprecipitation assay. We found that OIP5-AS1 was expressed at low levels in lipopolysaccharide (LPS)-stimulated hPDLSCs. OIP5-AS1 overexpression promoted proliferation and osteogenic differentiation ability and reduced LPS-induced inflammation in hPDLSCs. Furthermore, OIP5-AS1 directly targeted and reduced miR-92a-3p expression. The overexpression of miR-92a-3p partly abolished the effects of OIP5-AS1 on LPS-induced cell proliferation and osteogenic differentiation as well as inflammation in hPDLSCs. Collectively, the results indicated that OIP5-AS1 overexpression inhibited the LPS-induced inflammatory response and promoted the osteogenic differentiation of hPDLSCs by sponging miR-92a-3p. Thus, OIP5-AS1 is probably an essential objective for research during periodontitis treatment.
Collapse
Affiliation(s)
- Shiwei Wang
- Dental Department, The First Affiliated Hospital of Xi’an Medical University, Xi’an, Shanxi 710077, P.R. China
| | - Yao Duan
- Second Clinical Division, Peking University School and Hospital of Stomatology & National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology, Beijing 100081, P.R. China
| |
Collapse
|
49
|
Ren Y, Li L, Wang M, Yang Z, Sun Z, Zhang W, Cao L, Nie S. Knockdown of circRNA Paralemmin 2 Ameliorates Lipopolysaccharide-induced Murine Lung Epithelial Cell Injury by Sponging miR-330-5p to Reduce ROCK2 Expression. Immunol Invest 2022; 51:1707-1724. [PMID: 35171050 DOI: 10.1080/08820139.2022.2027961] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Previous data have reported the high expression of circRNA paralemmin 2 (circPALM2) in mice with acute lung injury (ALI). However, the role of circPALM2 in ALI pathogenesis remains unclear. The study aims to reveal the function of circPALM2 in ALI and the underlying mechanism. C57BL/6 J mice and murine lung epithelial-12 (MLE-12) cells were treated with lipopolysaccharide (LPS) to simulate ALI mouse and ALI cell models, respectively. Lung injury score and lung wet-to-dry ratio assays were used to evaluate the ALI mouse model. Quantitative real-time polymerase chain reaction and Western blot assays were implemented to analyze the expressions of circPALM2, microRNA-330-5p (miR-330-5p), rho-associated coiled-coil containing protein kinase 2 (ROCK2), and apoptosis-related markers. Cell viability, apoptosis, and the production of inflammatory cytokines were investigated by cell counting kit-8, flow cytometry, and enzyme-linked immunosorbent assays. The expressions of circPALM2 and ROCK2 were significantly increased, while miR-330-5p was decreased in ALI mice and LPS-induced MLE-12 cells compared with controls. LPS treatment inhibited cell viability but induced apoptosis, inflammatory cytokine production, and oxidative stress; however, these effects were attenuated after the combination of circPALM2 knockdown and LPS. CircPALM2 regulated LPS-caused MLE-12 cell damage by targeting miR-330-5p. Additionally, ROCK2, a target gene of miR-330-5p, participated in LPS-induced MLE-12 cell injury. Further, circPALM2 activated ROCK2 by associating with miR-330-5p. CircPALM2 modulated LPS-caused murine lung epithelial cell injury by the miR-330-5p/ROCK2 pathway, providing a therapeutic target for ALI.
Collapse
Affiliation(s)
- Yi Ren
- Department of Emergency Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, PR China
| | - Liang Li
- Department of Emergency Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, PR China
| | - Mengmeng Wang
- Department of Emergency Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, PR China
| | - Zhizhou Yang
- Department of Emergency Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, PR China
| | - Zhaorui Sun
- Department of Emergency Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, PR China
| | - Wei Zhang
- Department of Emergency Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, PR China
| | - Liping Cao
- Department of Emergency Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, PR China
| | - Shinan Nie
- Department of Emergency Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, PR China
| |
Collapse
|
50
|
Zheng F, Pan Y, Yang Y, Zeng C, Fang X, Shu Q, Chen Q. Novel biomarkers for acute respiratory distress syndrome: genetics, epigenetics and transcriptomics. Biomark Med 2022; 16:217-231. [PMID: 35026957 DOI: 10.2217/bmm-2021-0749] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) can be induced by multiple clinical factors, including sepsis, acute pancreatitis, trauma, intestinal ischemia/reperfusion and burns. However, these factors alone may poorly explain the risk and outcomes of ARDS. Emerging evidence suggests that genomic-based or transcriptomic-based biomarkers may hold the promise to establish predictive or prognostic stratification methods for ARDS, and also to help in developing novel therapeutic targets for ARDS. Notably, genetic/epigenetic variations correlated with susceptibility and prognosis of ARDS and circulating microRNAs have emerged as potential biomarkers for diagnosis or prognosis of ARDS. Although limited by sample size, ethnicity and phenotypic heterogeneity, ongoing genetic/transcriptomic research contributes to the characterization of novel biomarkers and ultimately helps to develop innovative therapeutics for ARDS patients.
Collapse
Affiliation(s)
- Fei Zheng
- Department of Clinical Research Center, The Children's Hospital, School of Medicine, Zhejiang University, National Clinical Research Center for Child Health, Hangzhou, 310052, China
| | - Yihang Pan
- Department of Clinical Research Center, The Children's Hospital, School of Medicine, Zhejiang University, National Clinical Research Center for Child Health, Hangzhou, 310052, China
| | - Yang Yang
- Department of Intensive Care Medicine, The Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Congli Zeng
- Department of Anesthesia, Critical Care & Pain Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Xiangming Fang
- Department of Anesthesiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Qiang Shu
- Department of Clinical Research Center, The Children's Hospital, School of Medicine, Zhejiang University, National Clinical Research Center for Child Health, Hangzhou, 310052, China
| | - Qixing Chen
- Department of Clinical Research Center, The Children's Hospital, School of Medicine, Zhejiang University, National Clinical Research Center for Child Health, Hangzhou, 310052, China
| |
Collapse
|