1
|
Sha HX, Liu YB, Qiu YL, Zhong WJ, Yang NSY, Zhang CY, Duan JX, Xiong JB, Guan CX, Zhou Y. Neutrophil extracellular traps trigger alveolar epithelial cell necroptosis through the cGAS-STING pathway during acute lung injury in mice. Int J Biol Sci 2024; 20:4713-4730. [PMID: 39309425 PMCID: PMC11414388 DOI: 10.7150/ijbs.99456] [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: 06/10/2024] [Accepted: 08/25/2024] [Indexed: 09/25/2024] Open
Abstract
Extensive loss of alveolar epithelial cells (AECs) undergoing necroptosis is a crucial mechanism of acute lung injury (ALI), but its triggering mechanism needs to be thoroughly investigated. Neutrophil extracellular traps (NETs) play a significant role in ALI. However, the effect of NETs on AECs' death has not been clarified. Our study found that intratracheal instillation of NETs disrupted lung tissue structure, suggesting that NETs could induce ALI in mice. Moreover, we observed that NETs could trigger necroptosis of AECs in vivo and in vitro. The phosphorylation levels of RIPK3 and MLKL were increased in MLE12 cells after NETs treatment (P < 0.05). Mechanistically, NETs taken up by AECs through endocytosis activated the cGAS-STING pathway and triggered AECs necroptosis. The expression of cGAS, STING, TBK1 and IRF3 were increased in MLE12 cells treated with NETs (P < 0.05). Furthermore, the cGAS inhibitor RU.521 inhibited NETs-triggered AECs necroptosis and alleviated the pulmonary damage induced by NETs in mice. In conclusion, our study demonstrates that NETs taken up by AECs via endocytosis can activate the cGAS-STING pathway and trigger AECs necroptosis to promote ALI in mice. Our findings indicate that targeting the NETs/cGAS-STING/necroptosis pathway in AECs is an effective strategy for treating ALI.
Collapse
Affiliation(s)
- Han-Xi Sha
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan 410078, China
- National Experimental Teaching Demonstration Center for Medical Function, Changsha, Hunan 410013, China
| | - Yu-Biao Liu
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan 410078, China
- National Experimental Teaching Demonstration Center for Medical Function, Changsha, Hunan 410013, China
| | - Yan-Ling Qiu
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan 410078, China
- National Experimental Teaching Demonstration Center for Medical Function, Changsha, Hunan 410013, China
| | - Wen-Jing Zhong
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan 410078, China
- National Experimental Teaching Demonstration Center for Medical Function, Changsha, Hunan 410013, China
| | - Nan-Shi-Yu Yang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan 410078, China
- National Experimental Teaching Demonstration Center for Medical Function, Changsha, Hunan 410013, China
| | - Chen-Yu Zhang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan 410078, China
- National Experimental Teaching Demonstration Center for Medical Function, Changsha, Hunan 410013, China
| | - Jia-Xi Duan
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan 410078, China
- National Experimental Teaching Demonstration Center for Medical Function, Changsha, Hunan 410013, China
| | - Jian-Bing Xiong
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan 410078, China
- National Experimental Teaching Demonstration Center for Medical Function, Changsha, Hunan 410013, China
| | - Cha-Xiang Guan
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan 410078, China
- National Experimental Teaching Demonstration Center for Medical Function, Changsha, Hunan 410013, China
| | - Yong Zhou
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan 410078, China
- National Experimental Teaching Demonstration Center for Medical Function, Changsha, Hunan 410013, China
| |
Collapse
|
2
|
Jia H, He W, Wu B, Zhong Z, Chang Y, Liu Y, Wang M, Xia S. Cigarette smoke-induced exosomal miR-221-3p facilitates M1 macrophage polarization via the STAT3 pathway in chronic obstructive pulmonary disease. Aging (Albany NY) 2024; 16:12379-12391. [PMID: 39213192 PMCID: PMC11424577 DOI: 10.18632/aging.206095] [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: 01/09/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024]
Abstract
AIMS Chronic obstructive pulmonary disease (COPD) is marked by irreversible airflow limitations stemming from small airway constriction and lung emphysema. The advancement of COPD is greatly influenced by the M1 polarization of macrophages. The mechanisms governing macrophage polarization in inflammation conditions in COPD are not yet fully understood. METHODS To investigate the interplay between exosomes triggered by cigarette smoke and the polarization of macrophages, we utilized a combination of flow cytometry, quantitative real-time reverse transcription PCR, and western blot analysis. RESULTS Our research reveals that cigarette smoke (CS) exposure induces the secretion of exosomes from human bronchial epithelial cells, with exosomal miR-221-3p identified as a key player in modulating the polarization of M1 macrophages. The evidence indicates that cigarette smoke promotes exosome secretion in these cells, with exosomal miR-221-3p targeting SOCS3 and regulating the STAT3 signaling pathway to facilitate M1 macrophage polarization. CONCLUSIONS This research delves into the molecular pathways through which miR-221-3p facilitates the polarization of M1 macrophages, presenting a groundbreaking approach for potential targeted therapy in COPD.
Collapse
Affiliation(s)
- Hui Jia
- Graduate School of Dalian Medical University, Dalian, China
- Department of Respiratory and Critical Care Medicine, Central Hospital Affiliated to Shenyang Medical College, Shenyang, China
| | - Wei He
- Department of Respiratory and Critical Care Medicine, Central Hospital Affiliated to Shenyang Medical College, Shenyang, China
| | - Bo Wu
- Department of Respiratory Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhaoshuang Zhong
- Department of Respiratory and Critical Care Medicine, Central Hospital Affiliated to Shenyang Medical College, Shenyang, China
| | - Yuele Chang
- Department of Respiratory and Critical Care Medicine, Central Hospital Affiliated to Shenyang Medical College, Shenyang, China
| | - Yang Liu
- Department of Respiratory and Critical Care Medicine, Central Hospital Affiliated to Shenyang Medical College, Shenyang, China
| | - Min Wang
- Department of Respiratory and Critical Care Medicine, Central Hospital Affiliated to Shenyang Medical College, Shenyang, China
| | - Shuyue Xia
- Department of Respiratory and Critical Care Medicine, Central Hospital Affiliated to Shenyang Medical College, Shenyang, China
| |
Collapse
|
3
|
Su J, Song Y, Zhu Z, Huang X, Fan J, Qiao J, Mao F. Cell-cell communication: new insights and clinical implications. Signal Transduct Target Ther 2024; 9:196. [PMID: 39107318 PMCID: PMC11382761 DOI: 10.1038/s41392-024-01888-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 05/09/2024] [Accepted: 06/02/2024] [Indexed: 09/11/2024] Open
Abstract
Multicellular organisms are composed of diverse cell types that must coordinate their behaviors through communication. Cell-cell communication (CCC) is essential for growth, development, differentiation, tissue and organ formation, maintenance, and physiological regulation. Cells communicate through direct contact or at a distance using ligand-receptor interactions. So cellular communication encompasses two essential processes: cell signal conduction for generation and intercellular transmission of signals, and cell signal transduction for reception and procession of signals. Deciphering intercellular communication networks is critical for understanding cell differentiation, development, and metabolism. First, we comprehensively review the historical milestones in CCC studies, followed by a detailed description of the mechanisms of signal molecule transmission and the importance of the main signaling pathways they mediate in maintaining biological functions. Then we systematically introduce a series of human diseases caused by abnormalities in cell communication and their progress in clinical applications. Finally, we summarize various methods for monitoring cell interactions, including cell imaging, proximity-based chemical labeling, mechanical force analysis, downstream analysis strategies, and single-cell technologies. These methods aim to illustrate how biological functions depend on these interactions and the complexity of their regulatory signaling pathways to regulate crucial physiological processes, including tissue homeostasis, cell development, and immune responses in diseases. In addition, this review enhances our understanding of the biological processes that occur after cell-cell binding, highlighting its application in discovering new therapeutic targets and biomarkers related to precision medicine. This collective understanding provides a foundation for developing new targeted drugs and personalized treatments.
Collapse
Affiliation(s)
- Jimeng Su
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Cancer Center, Peking University Third Hospital, Beijing, China
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China
| | - Ying Song
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Cancer Center, Peking University Third Hospital, Beijing, China
| | - Zhipeng Zhu
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Cancer Center, Peking University Third Hospital, Beijing, China
| | - Xinyue Huang
- Biomedical Research Institute, Shenzhen Peking University-the Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Jibiao Fan
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China
| | - Jie Qiao
- State Key Laboratory of Female Fertility Promotion, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China.
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China.
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China.
| | - Fengbiao Mao
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China.
- Cancer Center, Peking University Third Hospital, Beijing, China.
| |
Collapse
|
4
|
Zhang R, Yang A, Fu J, Zhang L, Yin L, Xu T, Dai C, Su W, Shen W. Budesonide and N-acetylcysteine inhibit activation of the NLRP3 inflammasome by regulating miR-381 to alleviate acute lung injury caused by the pyroptosis-mediated inflammatory response. Toxicol Res (Camb) 2024; 13:tfae115. [PMID: 39100861 PMCID: PMC11295220 DOI: 10.1093/toxres/tfae115] [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: 02/02/2024] [Revised: 06/04/2024] [Accepted: 07/24/2024] [Indexed: 08/06/2024] Open
Abstract
Background The anti-inflammatory effects of budesonide (BUN) and N-acetylcysteine (NAC) attenuate acute lung injury (ALI). The aim of this study was to investigate the effects of combination therapy consisting of BUN and NAC on ALI and the underlying mechanisms. Methods In vitro and in vivo models of ALI were generated by LPS induction. Western blotting was used to detect the expression levels of pyroptosis-related proteins and inflammation-related factors, and RT-qPCR was used to detect the expression of miR-381. Cell proliferation and apoptosis were detected by CCK-8 and flow cytometry, respectively. ELISA was used to detect the levels of inflammation-related factors. HE staining was used to detect lung injury. Results The results showed that LPS effectively induced pyroptosis in cells and promoted the expression of pyroptosis-related proteins (Caspase1, Gasdermin D and NLRP3) and inflammatory cytokines (TNF-α, IL-6 and IL-1β). The combination of BUN and NAC significantly alleviated LPS-induced pyroptosis and inflammation. In addition, the combination of BUN and NAC effectively promoted miR-381 expression. Transfection of miR-381 mimics effectively alleviated LPS-induced pyroptosis and inflammation, while transfection of miR-381 inhibitors had the opposite effect. miR-381 negatively regulates NLRP3 expression. Treatment with a miR-381 inhibitor or pc-NLRP3 reversed the effects of the combination of BUN and NAC. In a mouse model of ALI, the combination of BUN and NAC effectively improved lung injury, while treatment with a miR-381 inhibitor or pc-NLRP3 effectively reversed this effect. Conclusion Overall, this study revealed that BUN + NAC inhibits the activation of NLRP3 by regulating miR-381, thereby alleviating ALI caused by pyroptosis-mediated inflammation.
Collapse
Affiliation(s)
- Rongfang Zhang
- Department of Rehabilitation Medicine, Qujing No. 1 Hospital, No. 1 Garden Road, Qilin District, Qujing, Yunnan 655000, China
| | - Aiping Yang
- Department of Rehabilitation Medicine, Qujing No. 1 Hospital, No. 1 Garden Road, Qilin District, Qujing, Yunnan 655000, China
| | - Jin Fu
- Department of Rehabilitation Medicine, Qujing No. 1 Hospital, No. 1 Garden Road, Qilin District, Qujing, Yunnan 655000, China
| | - Li Zhang
- Department of Rehabilitation Medicine, Qujing No. 1 Hospital, No. 1 Garden Road, Qilin District, Qujing, Yunnan 655000, China
| | - Liyue Yin
- Department of Rehabilitation Medicine, Qujing No. 1 Hospital, No. 1 Garden Road, Qilin District, Qujing, Yunnan 655000, China
| | - Ting Xu
- Department of Rehabilitation Medicine, Qujing No. 1 Hospital, No. 1 Garden Road, Qilin District, Qujing, Yunnan 655000, China
| | - Chunhui Dai
- Department of Rehabilitation Medicine, Qujing No. 1 Hospital, No. 1 Garden Road, Qilin District, Qujing, Yunnan 655000, China
| | - Wenbing Su
- Department of Rehabilitation Medicine, Qujing No. 1 Hospital, No. 1 Garden Road, Qilin District, Qujing, Yunnan 655000, China
| | - Wanling Shen
- Department of Rehabilitation Medicine, Qujing No. 1 Hospital, No. 1 Garden Road, Qilin District, Qujing, Yunnan 655000, China
| |
Collapse
|
5
|
Karpurapu M, Nie Y, Chung S, Yan J, Dougherty P, Pannu S, Wisler J, Harkless R, Parinandi N, Berdyshev E, Pei D, Christman JW. The calcineurin-NFATc pathway modulates the lipid mediators in BAL fluid extracellular vesicles, thereby regulating microvascular endothelial cell barrier function. Front Physiol 2024; 15:1378565. [PMID: 38812883 PMCID: PMC11133699 DOI: 10.3389/fphys.2024.1378565] [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: 01/29/2024] [Accepted: 04/16/2024] [Indexed: 05/31/2024] Open
Abstract
Extracellular vesicles mediate intercellular communication by transporting biologically active macromolecules. Our prior studies have demonstrated that the nuclear factor of activated T cell cytoplasmic member 3 (NFATc3) is activated in mouse pulmonary macrophages in response to lipopolysaccharide (LPS). Inhibition of NFATc3 activation by a novel cell-permeable calcineurin peptide inhibitor CNI103 mitigated the development of acute lung injury (ALI) in LPS-treated mice. Although pro-inflammatory lipid mediators are known contributors to lung inflammation and injury, it remains unclear whether the calcineurin-NFATc pathway regulates extracellular vesicle (EV) lipid content and if this content contributes to ALI pathogenesis. In this study, EVs from mouse bronchoalveolar lavage fluid (BALF) were analyzed for their lipid mediators by liquid chromatography in conjunction with mass spectrometry (LC-MS/MS). Our data demonstrate that EVs from LPS-treated mice contained significantly higher levels of arachidonic acid (AA) metabolites, which were found in low levels by prior treatment with CNI103. The catalytic activity of lung tissue cytoplasmic phospholipase A2 (cPLA2) increased during ALI, correlating with an increased amount of arachidonic acid (AA) in the EVs. Furthermore, ALI is associated with increased expression of cPLA2, cyclooxygenase 2 (COX2), and lipoxygenases (5-LOX, 12-LOX, and 15-LOX) in lung tissue, and pretreatment with CNI103 inhibited the catalytic activity of cPLA2 and the expression of cPLA2, COX, and LOX transcripts. Furthermore, co-culture of mouse pulmonary microvascular endothelial cell (PMVEC) monolayer and NFAT-luciferase reporter macrophages with BALF EVs from LPS-treated mice increased the pulmonary microvascular endothelial cell (PMVEC) monolayer barrier permeability and luciferase activity in macrophages. However, EVs from CNI103-treated mice had no negative impact on PMVEC monolayer barrier integrity. In summary, BALF EVs from LPS-treated mice carry biologically active NFATc-dependent, AA-derived lipids that play a role in regulating PMVEC monolayer barrier function.
Collapse
Affiliation(s)
- Manjula Karpurapu
- Division of Pulmonary, Critical Care and Sleep Medicine, Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, OH, United States
| | - Yunjuan Nie
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Sangwoon Chung
- Division of Pulmonary, Critical Care and Sleep Medicine, Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, OH, United States
| | - Jiasheng Yan
- Department of Pharmacology, Ohio State University, Columbus, OH, United States
| | - Patrick Dougherty
- Department of Chemistry and Biochemistry, Ohio State University, Columbus, OH, United States
| | - Sonal Pannu
- Division of Pulmonary, Critical Care and Sleep Medicine, Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, OH, United States
| | - Jon Wisler
- Department of Surgery, Ohio State Wexner Medical Center, Columbus, OH, United States
| | - Ryan Harkless
- Department of Surgery, Ohio State Wexner Medical Center, Columbus, OH, United States
| | - Narasimham Parinandi
- Division of Pulmonary, Critical Care and Sleep Medicine, Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, OH, United States
| | - Evgeny Berdyshev
- Division of Pulmonary Critical Care and Sleep Medicine, National Jewish Health, Denver, CO, United States
| | - Dehua Pei
- Department of Chemistry and Biochemistry, Ohio State University, Columbus, OH, United States
| | - John W. Christman
- Division of Pulmonary, Critical Care and Sleep Medicine, Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, OH, United States
| |
Collapse
|
6
|
Wang Q, Wen W, Zhou L, Liu F, Ren X, Yu L, Chen H, Jiang Z. LL-37 improves sepsis-induced acute lung injury by suppressing pyroptosis in alveolar epithelial cells. Int Immunopharmacol 2024; 129:111580. [PMID: 38310763 DOI: 10.1016/j.intimp.2024.111580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/19/2024] [Accepted: 01/20/2024] [Indexed: 02/06/2024]
Abstract
BACKGROUND LL-37 (also known as murine CRAMP) is a human antimicrobial peptide that plays a crucial role in innate immune defence against sepsis through various mechanisms. However, its involvement in sepsis-induced lung injury remains unclear. OBJECTIVES This work investigates the impact of LL-37 on pyroptosis generated by LPS in alveolar epithelial cells. The research utilizes both in vivo and in vitro sepsis-associated acute lung injury (ALI) models to understand the underlying molecular pathways. METHODS In vivo, an acute lung injury model induced by sepsis was established by intratracheal administration of LPS in C57BL/6J mice, which were subsequently treated with low-dose CRAMP (recombinant murine cathelicidin, 2.5 mg.kg-1) and high-dose CRAMP (5.0 mg.kg-1). In vitro, pyroptosis was induced in a human alveolar epithelial cell line (A549) by stimulation with LPS and ATP. Treatment was carried out with recombinant human LL-37, or LL-37 was knocked out in A549 cells using small interfering RNA (siRNA). Subsequently, haematoxylin and eosin staining was performed to observe the histopathological changes in lung tissues in the control group and sepsis-induced lung injury group. TUNEL and PI staining were used to observe DNA fragmentation and pyroptosis in mouse lung tissues and cells in the different groups. An lactate dehydrogenase (LDH) assay was performed to measure the cell death rate. The expression levels of NLRP3, caspase1, caspase 1 p20, GSDMD, NT-GSDMD, and CRAMP were detected in mice and cells using Western blotting, qPCR, and immunohistochemistry. ELISA was used to assess the levels of interleukin (IL)-1β and IL-18 in mouse serum, bronchoalveolar lavage fluid (BALF) and lung tissue and cell culture supernatants. RESULTS The expression of NLRP3, caspase1 p20, NT-GSDMD, IL 18 and IL1β in the lung tissue of mice with septic lung injury was increased, which indicated activation of the canonical pyroptosis pathway and coincided with an increase in CRAMP expression. Treatment with recombinant CRAMP improved pyroptosis in mice with lung injury. In vitro, treatment with LPS and ATP upregulated these classic pyroptosis molecules, LL-37 knockdown exacerbated pyroptosis, and recombinant human LL-37 treatment alleviated pyroptosis in alveolar epithelial cells. CONCLUSION These findings indicate that LL-37 protects against septic lung injury by modulating the expression of classic pyroptotic pathway components, including NLRP3, caspase1, and GSDMD and downstream inflammatory factors in alveolar epithelial cells.
Collapse
Affiliation(s)
- Quanzhen Wang
- Department of Critical Care Medicine, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong 250014, China
| | - Wei Wen
- Department of Critical Care Medicine, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong 250014, China
| | - Lei Zhou
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, China; Department of Respiratory Intensive Care Unit, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong 250014, China
| | - Fen Liu
- Department of Critical Care Medicine, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong 250014, China
| | - Xiaoxu Ren
- Department of Critical Care Medicine, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong 250014, China
| | - Lifeng Yu
- Department of Critical Care Medicine, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong 250014, China
| | - Huanqin Chen
- Department of Gerontology, Qilu Hospital, Shandong University, Jinan, 250012 Shandong, China
| | - Zhiming Jiang
- Department of Critical Care Medicine, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong 250014, China.
| |
Collapse
|
7
|
Al-Humiari MA, Yu L, Liu LP, Nouri MZ, Tuna KM, Denslow ND, Alli AA. Extracellular vesicles from BALF of pediatric cystic fibrosis and asthma patients increase epithelial sodium channel activity in small airway epithelial cells. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2024; 1866:184219. [PMID: 37634857 DOI: 10.1016/j.bbamem.2023.184219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 08/20/2023] [Accepted: 08/23/2023] [Indexed: 08/29/2023]
Abstract
Extracellular Vesicles (EVs) are nanosized vesicles derived from all cell types. EV cargo allows for intercellular communication, intracellular signaling, and regulation of proteins in recipient cells. We tested the hypothesis that EVs isolated from the bronchoalveolar-lavage fluid (BALF) of pediatric cystic fibrosis (CF) or pediatric asthma patients increase epithelial sodium channel (ENaC) activity in normal human small airway epithelial cells (SAECs) and the mechanism involves specific EV lipids. We characterized EVs from BALF of pediatric CF and pediatric asthma patients by nanoparticle tracking analysis, transmission electron microscopy, and Western blotting. The CF and asthma pediatric groups were similar in BALF electrolytes concentration and cell count, except for neutrophils, which were higher in the CF group. Lipidomic analyses for each group of EVs were performed using targeted mass spectrometry. Phosphatidylethanolamine, sphingomyelins, and triacylglycerol were enriched in both groups, but phosphatidylcholine and phosphatidylinositol concentrations were greater in the CF group compared to the asthma group, and the opposite trend was found for phosphatidylserine. Endogenous ENaC activity, measured by the single-channel patch-clamp technique, increased in normal human SAECs after challenging SAEC with EVs from either the CF or asthma groups compared to control EVs. In conclusion, EVs isolated from BALF of pediatric patients with CF or asthma have unique lipid profiles. Despite the differences, both types of EVs increase ENaC activity in normal human SAECs compared to control EVs isolated from the conditioned media of these cells.
Collapse
Affiliation(s)
- Mohammed A Al-Humiari
- Department of Pediatrics, Pediatric Pulmonology, University of Florida, Gainesville, FL, United States of America
| | - Ling Yu
- Department of Physiology and Aging, University of Florida, Gainesville, FL, United States of America
| | - Lauren P Liu
- Department of Physiology and Aging, University of Florida, Gainesville, FL, United States of America
| | - Mohammad-Zaman Nouri
- Department of Physiological Sciences and Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL, United States of America
| | - Kubra M Tuna
- Department of Endocrinology, University of Florida, Gainesville, FL, United States of America
| | - Nancy D Denslow
- Department of Physiological Sciences and Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL, United States of America
| | - Abdel A Alli
- Department of Pediatrics, Pediatric Pulmonology, University of Florida, Gainesville, FL, United States of America; Department of Physiology and Aging, University of Florida, Gainesville, FL, United States of America; Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida, Gainesville, FL, United States of America.
| |
Collapse
|
8
|
An N, Chen Z, Zhao P, Yin W. Extracellular Vesicles in Sepsis: Pathogenic Roles, Organ Damage, and Therapeutic Implications. Int J Med Sci 2023; 20:1722-1731. [PMID: 37928875 PMCID: PMC10620861 DOI: 10.7150/ijms.86832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 10/05/2023] [Indexed: 11/07/2023] Open
Abstract
Despite significant advances in anti-infective treatment and organ function support technology in recent years, the mortality rate of sepsis remains high. In addition to the high costs of sepsis treatment, the increasing consumption of medical resources also aggravates economic pressure and social burden. Extracellular vesicles (EVs) are membrane vesicles released from different types of activated or apoptotic cells to mediate intercellular communication, which can be detected in both human and animal body fluids. A growing body of researches suggest that EVs play an important role in the pathogenesis of sepsis. In this review, we summarize the predominant roles of EVs in various pathological processes during sepsis and its related organ dysfunction.
Collapse
Affiliation(s)
- Ni An
- Department of Emergency, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Zhe Chen
- University College London, London, UK
| | - Peng Zhao
- Department of Emergency, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Wen Yin
- Department of Emergency, Xijing Hospital, Air Force Medical University, Xi'an, China
| |
Collapse
|
9
|
Park J, Woo SJ, Hong Y, Lee JJ, Hong JY. Association between the Respiratory Microbiome and Plasma Microbial Extracellular Vesicles in Intubated Patients. Microorganisms 2023; 11:2128. [PMID: 37763972 PMCID: PMC10537887 DOI: 10.3390/microorganisms11092128] [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: 07/21/2023] [Revised: 08/17/2023] [Accepted: 08/18/2023] [Indexed: 09/29/2023] Open
Abstract
Extracellular vesicles (EVs) regulate various cellular and immunological functions in human diseases. There is growing interest in the clinical role of microbial EVs in pneumonia. However, there is a lack of research on the correlation between lung microbiome with microbial EVs and the microbiome of other body sites in pneumonia. We investigated the co-occurrence of lung microbiome and plasma microbe-derived EVs (mEVs) in 111 samples obtained from 60 mechanically ventilated patients (41 pneumonia and 19 non-pneumonia cases). The microbial correlation between the two samples was compared between the pneumonia and non-pneumonia cases. Bacterial composition of the plasma mEVs was distinct from that of the lung microbiome. There was a significantly higher correlation between lung microbiome and plasma mEVs in non-pneumonia individuals compared to pneumonia patients. In particular, Acinetobacter and Lactobacillus genera had high correlation coefficients in non-pneumonia patients. This indicates a beneficial effect of mEVs in modulating host lung immune response through EV component transfer.
Collapse
Affiliation(s)
- Jinkyeong Park
- Department of Pulmonary, Allergy and Critical Care Medicine, Kyung Hee University Hospital at Gangdong, School of Medicine, Kyung Hee University, Seoul 05278, Republic of Korea;
| | - Seong Ji Woo
- Institute of New Frontier Research Team, Hallym University College of Medicine, Chuncheon 24253, Republic of Korea; (S.J.W.); (J.J.L.)
| | - Yoonki Hong
- Department of Internal Medicine, Kangwon National University Hospital, School of Medicine, Kangwon National University, Chuncheon 24289, Republic of Korea;
| | - Jae Jun Lee
- Institute of New Frontier Research Team, Hallym University College of Medicine, Chuncheon 24253, Republic of Korea; (S.J.W.); (J.J.L.)
| | - Ji Young Hong
- Institute of New Frontier Research Team, Hallym University College of Medicine, Chuncheon 24253, Republic of Korea; (S.J.W.); (J.J.L.)
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Chuncheon Sacred Heart Hospital, Hallym University Medical Center, Chuncheon 24253, Republic of Korea
| |
Collapse
|
10
|
Tapak M, Sadeghi S, Ghazanfari T, Mosaffa N. Chemical exposure and alveolar macrophages responses: 'the role of pulmonary defense mechanism in inhalation injuries'. BMJ Open Respir Res 2023; 10:e001589. [PMID: 37479504 PMCID: PMC10364189 DOI: 10.1136/bmjresp-2022-001589] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 04/28/2023] [Indexed: 07/23/2023] Open
Abstract
Epidemiological and clinical studies have indicated an association between particulate matter (PM) exposure and acute and chronic pulmonary inflammation, which may be registered as increased mortality and morbidity. Despite the increasing evidence, the pathophysiology mechanism of these PMs is still not fully characterised. Pulmonary alveolar macrophages (PAMs), as a predominant cell in the lung, play a critically important role in these pathological mechanisms. Toxin exposure triggers events associated with macrophage activation, including oxidative stress, acute damage, tissue disruption, remodelling and fibrosis. Targeting macrophage may potentially be employed to treat these types of lung inflammation without affecting the natural immune response to bacterial infections. Biological toxins, their sources of exposure, physical and other properties, and their effects on the individuals are summarised in this article. Inhaled particulates from air pollution and toxic gases containing chemicals can interact with alveolar epithelial cells and immune cells in the airways. PAMs can sense ambient pollutants and be stimulated, triggering cellular signalling pathways. These cells are highly adaptable and can change their function and phenotype in response to inhaled agents. PAMs also have the ability to polarise and undergo plasticity in response to tissue damage, while maintaining resistance to exposure to inhaled agents.
Collapse
Affiliation(s)
- Mahtab Tapak
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Alinasab Hospital, Labratory Department, Iranian Social Security Organization (ISSO), Tabriz, Iran
| | - Somaye Sadeghi
- Advanced Therapy Medicinal Product (ATMP) Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Tooba Ghazanfari
- Immunoregulation Research Centre, Shahed University, Tehran, Iran
- Department of Immunology, Shahed University, Tehran, Iran
| | - Nariman Mosaffa
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
11
|
Zheng D, Ruan H, Chen W, Zhang Y, Cui W, Chen H, Shen H. Advances in extracellular vesicle functionalization strategies for tissue regeneration. Bioact Mater 2023; 25:500-526. [PMID: 37056271 PMCID: PMC10087114 DOI: 10.1016/j.bioactmat.2022.07.022] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 07/06/2022] [Accepted: 07/19/2022] [Indexed: 11/02/2022] Open
Abstract
Extracellular vesicles (EVs) are nano-scale vesicles derived by cell secretion with unique advantages such as promoting cell proliferation, anti-inflammation, promoting blood vessels and regulating cell differentiation, which benefit their wide applications in regenerative medicine. However, the in vivo therapeutic effect of EVs still greatly restricted by several obstacles, including the off-targetability, rapid blood clearance, and undesired release. To address these issues, biomedical engineering techniques are vastly explored. This review summarizes different strategies to enhance EV functions from the perspective of drug loading, modification, and combination of biomaterials, and emphatically introduces the latest developments of functionalized EV-loaded biomaterials in different diseases, including cardio-vascular system diseases, osteochondral disorders, wound healing, nerve injuries. Challenges and future directions of EVs are also discussed.
Collapse
Affiliation(s)
- Dandan Zheng
- Department of Spine Surgery, Renji Hospital, Shanghai JiaoTong University School of Medicine, 160 Pujian Road, Shanghai, 200127, PR China
| | - Huitong Ruan
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Wei Chen
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Yuhui Zhang
- Department of Spine Surgery, Renji Hospital, Shanghai JiaoTong University School of Medicine, 160 Pujian Road, Shanghai, 200127, PR China
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Hao Chen
- Department of Spine Surgery, Renji Hospital, Shanghai JiaoTong University School of Medicine, 160 Pujian Road, Shanghai, 200127, PR China
| | - Hongxing Shen
- Department of Spine Surgery, Renji Hospital, Shanghai JiaoTong University School of Medicine, 160 Pujian Road, Shanghai, 200127, PR China
| |
Collapse
|
12
|
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: 4] [Impact Index Per Article: 4.0] [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
|
13
|
Hamuro J, Yamashita T, Otsuki Y, Hiramoto N, Adachi M, Miyatani T, Tanaka H, Ueno M, Kinoshita S, Sotozono C. Spatiotemporal Coordination of RPE Cell Quality by Extracellular Vesicle miR-494-3p Via Competitive Interplays With SIRT3 or PTEN. Invest Ophthalmol Vis Sci 2023; 64:9. [PMID: 37163276 PMCID: PMC10179576 DOI: 10.1167/iovs.64.5.9] [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: 02/02/2023] [Accepted: 04/13/2023] [Indexed: 05/11/2023] Open
Abstract
Purpose To reveal the molecular mechanism underlying degeneration in human retinal pigment epithelial (hRPE) cells with dysfunctional mitochondrial homeostasis. Methods The expression of recently identified miR-494-3p in extracellular vesicles (EV) released from induced-pluripotential-stem-cell-derived human RPE (iPS-hRPE), during coculture with macrophages (Mps) was investigated in iPS-hRPE and ARPE cells differentiated in the presence of nicotinamide (Nic-ARPE). The expression of phosphatase and tensin homolog (PTEN), sirtuin3 (SIRT3), and mitochondrial marker proteins before and after the transfection of miR-494-3p inhibitor and mimic, and the changes in mitochondrial metabolism, membrane potential, and oxidative phosphorylation (OXPHOS) were monitored. Results Compared with senescent dedifferentiated ARPE19 cells, iPS-hRPE and Nic-ARPE cells expressed elevated levels of mitochondrial marker proteins but a repressed cellular miR-494-3p level. The expression of target proteins of miR-494-3p, PTEN, and SIRT3 was upregulated along with the differentiation disposition of these RPE cells. The ratio of PTEN/SIRT3 in de-differentiated ARPE19 cells was surprisingly elevated by around 20 times compared with that in iPS-hRPE and Nic-ARPE cells. The novel molecular interplay of EV miR-494-3p either with mitochondria selective SIRT3 or organelle nonselective PTEN was found to participate in the degeneration of hRPE cells by inducing mitochondrial dysfunctions and repressed OXPHOS, mitochondrial membrane potential, and ATP and NAD+ production. Conclusions Our results demonstrate a clear causal link between miR-494-3p and hRPE cell degeneration via the regulation of mitochondrial integrity. EV miR-494-3p may play a pivotal role in pathogenic spreading of degenerated hRPE cells from the local perifovea throughout the macula.
Collapse
Affiliation(s)
- Junji Hamuro
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tomoko Yamashita
- Department of Frontier Medical Science and Technology for Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yohei Otsuki
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Nao Hiramoto
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Mayuka Adachi
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takafumi Miyatani
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hiroshi Tanaka
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Morio Ueno
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Shigeru Kinoshita
- Department of Frontier Medical Science and Technology for Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Chie Sotozono
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
14
|
Yang Y, Hong S, Wang Q, Wang S, Xun Y. Exosome-mediated crosstalk between epithelial cells amplifies the cell injury cascade in CaOx stone formation. J Biol Eng 2023; 17:16. [PMID: 36855143 PMCID: PMC9976448 DOI: 10.1186/s13036-023-00324-0] [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: 10/08/2022] [Accepted: 01/04/2023] [Indexed: 03/02/2023] Open
Abstract
BACKGROUND Calcium oxalate (CaOx) stone disease is found worldwide. To explore the role of exosomes as a mediator of intercellular crosstalk during CaOx stone formation, we conducted this study, which may provide a new insight into the treatment and prevention of CaOx stones. METHODS Exosomes derived from HK2 cells with (EXO(S)) or without (EXO(C))CaOx crystal stimulation were cocultured with normal tubular epithelial cells and subcapsularly injected into rat kidneys. Then, oxidative stress levels, the MAPK signalling pathway and osteogenic changes were detected via qPCR, Western blotting, immunofluorescence and immunohistochemical staining. In vivo fluorescence imaging and exosome internalization assays showed the absorption and utilization of exosomes. RESULTS EXO(S) increased the reactive oxygen species (ROS) level and activated the expression of BMP2, OPN and OCN via the MAPK/P-38 pathway both in vivo and in vitro. In vivo experiments showed that preinjection of EXO(S) aggravated, while preinjection of EXO(C) ameliorated, these effects. Crystal depositions were significantly increased in SD rats injected with GAM when they were preinjected with EXO(S), and these effects could be reversed after preinjection with EXO(C). CONCLUSION Our study revealed that exosome-mediated intercellular crosstalk could accelerate the formation of CaOx stones by promoting oxidative stress and the osteogenic cascade in normal tubular epithelial cells. HK2 cells stimulated with CaOx crystals released more exosomal miR-223-3p and S100A8 comparing with normal HK2 cells. These exosomes derived from HK2 cells stimulated with CaOx (EXO(S)) could amplify the oxidative stress and osteogenic changes via MAPK/P-38 pathway, which finally led to the formation of Randall's plaque.
Collapse
Affiliation(s)
- Yuanyuan Yang
- grid.33199.310000 0004 0368 7223Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 Hubei China
| | - Senyuan Hong
- grid.33199.310000 0004 0368 7223Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 Hubei China
| | - Qing Wang
- grid.443382.a0000 0004 1804 268XDepartment of Urology, Guizhou Provincial People’s Hospital, Guizhou University, Guiyang, 550000 Guizhou China ,grid.443382.a0000 0004 1804 268XDepartment of Research Laboratory Center, Guizhou Provincial People’s Hospital, Guizhou University, Guiyang, 550000 Guizhou China
| | - Shaogang Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| | - Yang Xun
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| |
Collapse
|
15
|
Moirangthem A, Gondaliya P, Yan IK, Sayyed AA, Driscoll J, Patel T. Extracellular vesicle‑mediated miR‑126‑3p transfer contributes to inter‑cellular communication in the liver tumor microenvironment. Int J Oncol 2023; 62:31. [PMID: 36660950 PMCID: PMC9851126 DOI: 10.3892/ijo.2023.5479] [Citation(s) in RCA: 5] [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: 02/05/2022] [Accepted: 10/24/2022] [Indexed: 01/18/2023] Open
Abstract
Extracellular vesicles (EVs) and their contents are gaining recognition as important mediators of intercellular communication through the transfer of bioactive molecules, such as non‑coding RNA. The present study comprehensively assessed the microRNA (miRNA/miR) content within EVs released from HepG2 liver cancer (LC) cells and LX2 hepatic stellate cells (HSCs) and determined the contribution of EV miRNA to intercellular communication. Using both transwell and spheroid co‑cultures of LC cells and HSCs, miR‑126‑3p within EV was established as a mediator of HSC to LC cell communication that influenced tumor cell migration and invasion, as well as the growth of multicellular LC/HSC spheroids. Manipulation of miR‑126‑3p either by enforced expression using pre‑miR‑126‑3p or by inhibition using antimiR‑126‑3p did not alter tumor cell viability, proliferation or sensitivity to either sorafenib or regorafenib. By contrast, enforced expression of miR‑126‑3p decreased tumor‑cell migration. Knockdown of miR‑126‑3p in tumor cells increased disintegrin and metalloproteinase domain‑containing protein 9 (ADAM9) expression and in HSCs increased collagen‑1A1 accumulation with an increase in compactness of multicellular spheroids. Within LC/HSC spheroids, ADAM9 and vascular endothelial growth factor expression was increased by silencing of miR‑126‑3p but diminished with the restoration of miR‑126‑3p. These studies implicate miR‑126‑3p in functional effects on migration, invasion and spheroid growth of tumor cells in the presence of HSCs, and thereby demonstrate functional EV‑RNA‑based intercellular signaling between HSCs and LC cells that is directly relevant to tumor‑cell behavior.
Collapse
Affiliation(s)
| | | | - Irene K. Yan
- Departments of Transplantation and Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Adil Ali Sayyed
- Departments of Transplantation and Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Julia Driscoll
- Departments of Transplantation and Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Tushar Patel
- Departments of Transplantation and Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA
| |
Collapse
|
16
|
Burke H, Cellura D, Freeman A, Hicks A, Ostridge K, Watson A, Williams NP, Spalluto CM, Staples KJ, Wilkinson TMA. Pulmonary EV miRNA profiles identify disease and distinct inflammatory endotypes in COPD. Front Med (Lausanne) 2022; 9:1039702. [PMID: 36590967 PMCID: PMC9797812 DOI: 10.3389/fmed.2022.1039702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 11/28/2022] [Indexed: 12/16/2022] Open
Abstract
Introduction Chronic obstructive pulmonary disease (COPD) is a heterogeneous condition without effective disease modifying therapies. Identification of novel inflammatory endotype markers such as extracellular vesicles (EVs), which are important intercellular messengers carrying microRNA (miRNA), may enable earlier diagnosis and disease stratification for a targeted treatment approach. Our aim was to identify differentially expressed EV miRNA in the lungs of COPD patients compared with healthy ex-smokers and determine whether they can help define inflammatory COPD endotypes. Methods EV miRNA were isolated and sequenced from ex-smoking COPD patients and healthy ex-smoker bronchoalveolar lavage fluid. Results were validated with RT-qPCR and compared to differential inflammatory cell counts. Results Expression analysis identified five upregulated miRNA in COPD (miR-223-3p, miR-2110, miR-182-5p, miR-200b-5p and miR-625-3p) and three downregulated miRNA (miR-138-5p, miR-338-3p and miR-204-5p), all with a log2 fold change of >1/-1, FDR < 0.05. These miRNAs correlated with disease defining characteristics such as FEF 25-75% (a small airways disease measure) and DLCO% (a surrogate measure of emphysema). Receiver operator curve analysis demonstrated miR-2110, miR-223-3p, and miR-182-5p showed excellent combinatory predictive ability (AUC 0.91, p < 0.0001) in differentiating between health and mild COPD. Furthermore, miR-223-3p and miR-338-3p correlated with airway eosinophilia and were able to distinguish "pure eosinophilic" COPD from other airway inflammatory subtypes (AUC 0.94 and 0.85, respectively). Discussion This is the first study to identify differentially expressed miRNA in COPD bronchoalveolar lavage fluid EVs. These findings suggest specific lung derived EV miRNA are a strong predictor of disease presence even in mild COPD. Furthermore, specific miRNA correlated with inflammatory cell numbers in COPD, and may have a role in defining inflammatory endotypes for future treatment stratification.
Collapse
Affiliation(s)
- Hannah Burke
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, United Kingdom
| | - Doriana Cellura
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, United Kingdom
| | - Anna Freeman
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, United Kingdom
| | - Alex Hicks
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, United Kingdom
| | - Kris Ostridge
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Alastair Watson
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, United Kingdom
| | - Nicholas P. Williams
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, United Kingdom
| | - C. Mirella Spalluto
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, United Kingdom
| | - Karl J. Staples
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, United Kingdom
| | - Tom M. A. Wilkinson
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, United Kingdom
| |
Collapse
|
17
|
Yang HH, Jiang HL, Tao JH, Zhang CY, Xiong JB, Yang JT, Liu YB, Zhong WJ, Guan XX, Duan JX, Zhang YF, Liu SK, Jiang JX, Zhou Y, Guan CX. Mitochondrial citrate accumulation drives alveolar epithelial cell necroptosis in lipopolysaccharide-induced acute lung injury. Exp Mol Med 2022; 54:2077-2091. [PMID: 36443565 PMCID: PMC9722936 DOI: 10.1038/s12276-022-00889-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 09/09/2022] [Accepted: 09/14/2022] [Indexed: 11/29/2022] Open
Abstract
Necroptosis is the major cause of death in alveolar epithelial cells (AECs) during acute lung injury (ALI). Here, we report a previously unrecognized mechanism for necroptosis. We found an accumulation of mitochondrial citrate (citratemt) in lipopolysaccharide (LPS)-treated AECs because of the downregulation of Idh3α and citrate carrier (CIC, also known as Slc25a1). shRNA- or inhibitor-mediated inhibition of Idh3α and Slc25a1 induced citratemt accumulation and necroptosis in vitro. Mice with AEC-specific Idh3α and Slc25a1 deficiency exhibited exacerbated lung injury and AEC necroptosis. Interestingly, the overexpression of Idh3α and Slc25a1 decreased citratemt levels and rescued AECs from necroptosis. Mechanistically, citratemt accumulation induced mitochondrial fission and excessive mitophagy in AECs. Furthermore, citratemt directly interacted with FUN14 domain-containing protein 1 (FUNDC1) and promoted the interaction of FUNDC1 with dynamin-related protein 1 (DRP1), leading to excessive mitophagy-mediated necroptosis and thereby initiating and promoting ALI. Importantly, necroptosis induced by citratemt accumulation was inhibited in FUNDC1-knockout AECs. We show that citratemt accumulation is a novel target for protection against ALI involving necroptosis.
Collapse
Affiliation(s)
- Hui-Hui Yang
- grid.216417.70000 0001 0379 7164Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan China
| | - Hui-Ling Jiang
- grid.216417.70000 0001 0379 7164Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan China
| | - Jia-Hao Tao
- grid.216417.70000 0001 0379 7164Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan China
| | - Chen-Yu Zhang
- grid.216417.70000 0001 0379 7164Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan China
| | - Jian-Bing Xiong
- grid.216417.70000 0001 0379 7164Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan China
| | - Jin-Tong Yang
- grid.216417.70000 0001 0379 7164Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan China
| | - Yu-Biao Liu
- grid.216417.70000 0001 0379 7164Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan China
| | - Wen-Jing Zhong
- grid.216417.70000 0001 0379 7164Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan China
| | - Xin-Xin Guan
- grid.216417.70000 0001 0379 7164Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan China
| | - Jia-Xi Duan
- grid.216417.70000 0001 0379 7164Department of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan China
| | - Yan-Feng Zhang
- grid.216417.70000 0001 0379 7164Department of Cardiovascular Surgery, Xiangya Hospital, Central South University, Changsha, Hunan China
| | - Shao-Kun Liu
- grid.216417.70000 0001 0379 7164Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan China
| | - Jian-Xin Jiang
- grid.410570.70000 0004 1760 6682State Key Laboratory of Trauma, Burns, and Combined Injury, Department of Trauma Medical Center, Daping Hospital, Army Medical University, Chongqing, China
| | - Yong Zhou
- grid.216417.70000 0001 0379 7164Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan China
| | - Cha-Xiang Guan
- grid.216417.70000 0001 0379 7164Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan China
| |
Collapse
|
18
|
Liao K, Niu F, Hu G, Buch S. Morphine-mediated release of astrocyte-derived extracellular vesicle miR-23a induces loss of pericyte coverage at the blood-brain barrier: Implications for neuroinflammation. Front Cell Dev Biol 2022; 10:984375. [PMID: 36478740 PMCID: PMC9720401 DOI: 10.3389/fcell.2022.984375] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 11/07/2022] [Indexed: 08/08/2023] Open
Abstract
Opioids such as morphine are the most potent and efficacious drugs currently available for pain management. Paradoxically, opioids have also been implicated in inducing neuroinflammation and associated neurocognitive decline. Pericytes, a critical component of the neurovascular unit (NVU), are centrally positioned between endothelial cells and astrocytes, maintaining function of the blood-brain barrier (BBB) nd regulating neuroinflammation by controlling monocyte influx under various pathological conditions. The role of pericytes in morphine-mediated neuroinflammation however, has received less attention, especially in the context of how pericytes crosstalk with other central nervous system (CNS) cells. The current study was undertaken to examine the effect of miRNAs released from morphine-stimulated human primary astrocyte-derived extracellular vesicles (morphine-ADEVs) in mediating pericyte loss at the blood-brain barrier, leading, in turn, to increased influx of peripheral monocytes. Our findings suggest that the heterogeneous nuclear ribonucleoprotein complex A2/B1 (hnRNP A2/B1) plays role in morphine-mediated upregulation and release of miR-23a in ADEVs, and through action of morphine via mu opioid receptor.We further demonstrated that miR-23a in morphine-ADEVs could be taken up by pericytes, resulting in downregulation of PTEN expression, ultimately leading to increased pericyte migration. Furthermore, both overexpression of PTEN and blocking the miR-23a target site at PTEN 3UTR (by transfecting miR-23a-PTEN target protector), attenuated morphine-ADEV-mediated pericyte migration. We also demonstrated that in the microvessels isolated from morphine-administered mice, there were fewer PDGFβR + pericytes co-localizing with CD31+ brain endothelial cells compared with those from saline mice. In line with these findings, we also observed increased loss of pericytes and a concomitantly increased influx of monocytes in the brains of morphine-administered pericyte-labeled NG2-DsRed mice compared with saline mice. In conclusion, our findings indicate morphine-ADEVs mediated loss of pericyte coverage at the brain endothelium, thereby increasing the influx of peripheral monocytes in the central nervous system, leading to neuroinflammation.
Collapse
Affiliation(s)
- Ke Liao
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Fang Niu
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| | - Guoku Hu
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| | - Shilpa Buch
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
19
|
Cai J, Wang YL, Sheng XD, Zhang L, Lv X. Shufeng Jiedu capsule inhibits inflammation and apoptosis by activating A2AAR and inhibiting NF-κB to alleviate LPS-induced ALI. JOURNAL OF ETHNOPHARMACOLOGY 2022; 298:115661. [PMID: 36002086 PMCID: PMC9392900 DOI: 10.1016/j.jep.2022.115661] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/31/2022] [Accepted: 08/17/2022] [Indexed: 05/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Shufeng Jiedu capsule (SFJDC) is a pure form of traditional Chinese medicine (TCM) that contains eight medicinal plants. Known for its anti-inflammatory and antipyretic effects, it is mostly used to treat upper respiratory tract infections and other infectious diseases, such as colds, pharyngitis, laryngitis, and tonsillitis. Both acute lung injury (ALI) and COVID-19 are closely related to lung damage, primarily manifesting as lung inflammation and epithelial cell damage. However, whether SFJDC can improve ALI and by what mechanism remain unclear. The purpose of this study was to explore whether SFJDC could be used as a prophylactic treatment for COVID-19 by improving acute lung injury. AIM OF THE STUDY The purpose of this study was to determine whether SFJDC could protect against ALI caused by lipopolysaccharide (LPS), and we wanted to determine how SFJDC reduces inflammation and apoptosis pharmacologically and molecularly. MATERIALS AND METHODS Preadministering SFJDC at 0.1 g/kg, 0.3 g/kg, or 0.5 g/kg for one week was followed by 5 mg/kg LPS to induce ALI in mice. Observations included the study of lung histomorphology, tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and interleukin-6 (IL-6) secretion, as well as the ratio of lung wet/dry weights. In addition, RAW264.7 cells were treated for 24 h with 1 μg/mL LPS after being pretreated for 1 h with 0.5 mg/mL SFJDC. In the samples, we detected TNF-α, IL-1β, and IL-6. Cell apoptosis was detected by stimulating A549 cells for 24 h with RAW264.7 supernatant. Both in vitro and in vivo, the levels of A2A adenosine receptor (A2AAR), PKA, IκB, p-IκB, NF-κB P65 (P65), p-NF-κB P65 (p-P65), cleaved caspases-3 (Cc3), Bcl-2 associated X protein (Bax), and B-cell lymphoma-2 (Bcl-2) proteins were determined using Western blot analysis. RESULTS Lung tissue morphology was improved as SFJDC decreased cytokine secretion, the ratio of lung wet/dry weights, and lung tissue secretion of proinflammatory cytokines. The expression of A2AAR was increased by SFJDC, and the phosphorylation of NF-κB was inhibited. TUNEL staining and flow cytometry showed that SFJDC inhibited apoptosis by reducing the expression of Cc3 and the ratio of Bax/Bcl-2. CONCLUSIONS According to the results of this study, SFJDC can reduce inflammation and inhibit apoptosis. A2AAR activation and regulation of NF-κB expression are thought to make SFJDC anti-inflammatory and anti-apoptotic. A wide range of active ingredients may result in an anti-inflammatory and antipyretic effect with SFJDC.
Collapse
Affiliation(s)
- Junnan Cai
- Institute of Liver Disease, Anhui Medical University, Hefei, Anhui, China; Center of Traditional Chinese Medicine Formula Granule, Anhui Medical University, Hefei, Anhui, China; Key Laboratory of Anti-inflammatory and Immunological Drugs, Ministry of Education, Hefei, China; Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China
| | - Yu-Lian Wang
- Institute of Liver Disease, Anhui Medical University, Hefei, Anhui, China; Center of Traditional Chinese Medicine Formula Granule, Anhui Medical University, Hefei, Anhui, China; Key Laboratory of Anti-inflammatory and Immunological Drugs, Ministry of Education, Hefei, China; Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China
| | - Xiao-Dong Sheng
- Institute of Liver Disease, Anhui Medical University, Hefei, Anhui, China; Center of Traditional Chinese Medicine Formula Granule, Anhui Medical University, Hefei, Anhui, China; Key Laboratory of Anti-inflammatory and Immunological Drugs, Ministry of Education, Hefei, China; Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China
| | - Lei Zhang
- Institute of Liver Disease, Anhui Medical University, Hefei, Anhui, China; Center of Traditional Chinese Medicine Formula Granule, Anhui Medical University, Hefei, Anhui, China; Key Laboratory of Anti-inflammatory and Immunological Drugs, Ministry of Education, Hefei, China; Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China
| | - Xiongwen Lv
- Institute of Liver Disease, Anhui Medical University, Hefei, Anhui, China; Center of Traditional Chinese Medicine Formula Granule, Anhui Medical University, Hefei, Anhui, China; Key Laboratory of Anti-inflammatory and Immunological Drugs, Ministry of Education, Hefei, China; Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China.
| |
Collapse
|
20
|
Marzec JM, Nadadur SS. Inflammation resolution in environmental pulmonary health and morbidity. Toxicol Appl Pharmacol 2022; 449:116070. [PMID: 35618031 PMCID: PMC9872158 DOI: 10.1016/j.taap.2022.116070] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 05/04/2022] [Accepted: 05/14/2022] [Indexed: 02/07/2023]
Abstract
Inflammation and resolution are dynamic processes comprised of inflammatory activation and neutrophil influx, followed by mediator catabolism and efferocytosis. These critical pathways ensure a return to homeostasis and promote repair. Over the past decade research has shown that diverse mediators play a role in the active process of resolution. Specialized pro-resolving mediators (SPMs), biosynthesized from fatty acids, are released during inflammation to facilitate resolution and are deficient in a variety of lung disorders. Failed resolution results in remodeling and cellular deposition through pro-fibrotic myofibroblast expansion that irreversibly narrows the airways and worsens lung function. Recent studies indicate environmental exposures may perturb and deregulate critical resolution pathways. Environmental xenobiotics induce lung inflammation and generate reactive metabolites that promote oxidative stress, injuring the respiratory mucosa and impairing gas-exchange. This warrants recognition of xenobiotic associated molecular patterns (XAMPs) as new signals in the field of inflammation biology, as many environmental chemicals generate free radicals capable of initiating the inflammatory response. Recent studies suggest that unresolved, persistent inflammation impacts both resolution pathways and endogenous regulatory mediators, compromising lung function, which over time can progress to chronic lung disease. Chronic ozone (O3) exposure overwhelms successful resolution, and in susceptible individuals promotes asthma onset. The industrial contaminant cadmium (Cd) bioaccumulates in the lung to impair resolution, and recurrent inflammation can result in chronic obstructive pulmonary disease (COPD). Persistent particulate matter (PM) exposure increases systemic cardiopulmonary inflammation, which reduces lung function and can exacerbate asthma, COPD, and idiopathic pulmonary fibrosis (IPF). While recurrent inflammation underlies environmentally induced pulmonary morbidity and may drive the disease process, our understanding of inflammation resolution in this context is limited. This review aims to explore inflammation resolution biology and its role in chronic environmental lung disease(s).
Collapse
Affiliation(s)
- Jacqui M Marzec
- National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Srikanth S Nadadur
- National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA.
| |
Collapse
|
21
|
Höglund N, Koho N, Rossi H, Karttunen J, Mustonen AM, Nieminen P, Rilla K, Oikari S, Mykkänen A. Isolation of Extracellular Vesicles From the Bronchoalveolar Lavage Fluid of Healthy and Asthmatic Horses. Front Vet Sci 2022; 9:894189. [PMID: 35799843 PMCID: PMC9255554 DOI: 10.3389/fvets.2022.894189] [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: 03/11/2022] [Accepted: 05/27/2022] [Indexed: 11/15/2022] Open
Abstract
Extracellular vesicles (EVs) are membrane-bound particles that engage in inflammatory reactions by mediating cell–cell interactions. Previously, EVs have been isolated from the bronchoalveolar lavage fluid (BALF) of humans and rodents. The aim of this study was to investigate the number and size distribution of EVs in the BALF of asthmatic horses (EA, n = 35) and healthy horses (n = 19). Saline was injected during bronchoscopy to the right lung followed by manual aspiration. The retrieved BALF was centrifuged twice to remove cells and biological debris. The supernatant was concentrated and EVs were isolated using size-exclusion chromatography. Sample fractions were measured with nanoparticle tracking analysis (NTA) for particle number and size, and transmission electron microscopy and confocal laser scanning microscopy were used to visualize EVs. The described method was able to isolate and preserve EVs. The mean EV size was 247 ± 35 nm (SD) in the EA horses and 261 ± 47 nm in the controls by NTA. The mean concentration of EVs was 1.38 × 1012 ± 1.42 × 1012 particles/mL in the EA horses and 1.33 × 1012 ± 1.07 × 1012 particles/mL in the controls with no statistically significant differences between the groups. With Western blotting and microscopy, these particles were documented to associate with EV protein markers (CD63, TSG101, HSP70, EMMPRIN, and actin) and hyaluronan. Equine BALF is rich in EVs of various sizes, and the described protocol is usable for isolating EVs. In the future, the role of EVs can be studied in horses with airway inflammation.
Collapse
Affiliation(s)
- Nina Höglund
- Department of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
- *Correspondence: Nina Höglund
| | - Ninna Koho
- Department of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Heini Rossi
- Department of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Jenni Karttunen
- Department of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Anne-Mari Mustonen
- Institute of Biomedicine, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
- Department of Environmental and Biological Sciences, Faculty of Science and Forestry, University of Eastern Finland, Joensuu, Finland
| | - Petteri Nieminen
- Institute of Biomedicine, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Kirsi Rilla
- Institute of Biomedicine, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Sanna Oikari
- Institute of Biomedicine, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Anna Mykkänen
- Department of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
22
|
Lam LM, Mangalmurti NS. ExRNA Takes a Toll in Sepsis-Associated Lung Injury. Am J Respir Cell Mol Biol 2022; 67:271-272. [PMID: 35728049 PMCID: PMC9447133 DOI: 10.1165/rcmb.2022-0237ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Lk Metthew Lam
- University of Pennsylvania Perelman School of Medicine, 14640, Philadelphia, Pennsylvania, United States
| | - Nilam S Mangalmurti
- University of Pennsylvania Perelman School of Medicine, 14640, Philadelphia, Pennsylvania, United States;
| |
Collapse
|
23
|
Abstract
Extracellular vesicles (EVs) are membranous nanoparticles secreted by nearly all cell types and play a critical role in cell-to-cell crosstalk. EVs can be categorized based on their size, surface markers, or the cell type from which they originate. EVs carry "cargo," including but not limited to, RNA, DNA, proteins, and small signaling molecules. To date, many methods have been developed to isolate EVs from biological fluids, such as blood plasma, urine, bronchoalveolar lavage fluid, and urine. Once isolated, EVs can be characterized by dynamic light scattering, nanotracking analysis, nanoscale flow cytometry, and transmission electron microscopy. Given the ability of EVs to transport cargo between cells, research has recently focused on understanding their role in various human diseases. As understanding of their significance to disease processes grows, insight into the mechanisms behind the physiological role of their cargo in target cells can facilitate the development of a new type of biomarker and therapeutic target for diseases in future. In addition, their ability to deliver their cargo selectively to target cells within the human body means that they could serve as therapeutic agents or methods of drug delivery. In this review, we will first introduce EVs and the cargo they carry, outline current methods for EV isolation and characterization, and discuss their potential use as biomarkers and therapeutic agents in the near future.
Collapse
Affiliation(s)
- Jonathan M Carnino
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University Medical Campus, Boston, MA, United States
| | - Heedoo Lee
- Department of Biology and Chemistry, Changwon National University, Changwon, South Korea.
| |
Collapse
|
24
|
Weber B, Franz N, Marzi I, Henrich D, Leppik L. Extracellular vesicles as mediators and markers of acute organ injury: current concepts. Eur J Trauma Emerg Surg 2022; 48:1525-1544. [PMID: 33533957 PMCID: PMC7856451 DOI: 10.1007/s00068-021-01607-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 01/19/2021] [Indexed: 12/15/2022]
Abstract
Due to the continued high incidence and mortality rate worldwide, there is a need to develop new strategies for the quick, precise, and valuable recognition of presenting injury pattern in traumatized and poly-traumatized patients. Extracellular vesicles (EVs) have been shown to facilitate intercellular communication processes between cells in close proximity as well as distant cells in healthy and disease organisms. miRNAs and proteins transferred by EVs play biological roles in maintaining normal organ structure and function under physiological conditions. In pathological conditions, EVs change the miRNAs and protein cargo composition, mediating or suppressing the injury consequences. Therefore, incorporating EVs with their unique protein and miRNAs signature into the list of promising new biomarkers is a logical next step. In this review, we discuss the general characteristics and technical aspects of EVs isolation and characterization. We discuss results of recent in vitro, in vivo, and patients study describing the role of EVs in different inflammatory diseases and traumatic organ injuries. miRNAs and protein signature of EVs found in patients with acute organ injury are also debated.
Collapse
Affiliation(s)
- Birte Weber
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt, Goethe-University, Frankfurt am Main, Germany
| | - Niklas Franz
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt, Goethe-University, Frankfurt am Main, Germany
| | - Ingo Marzi
- 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
| | - Liudmila Leppik
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt, Goethe-University, Frankfurt am Main, Germany.
| |
Collapse
|
25
|
Jiang HL, Yang HH, Liu YB, Zhang CY, Zhong WJ, Guan XX, Jin L, Hong JR, Yang JT, Tan XH, Li Q, Zhou Y, Guan CX. L-OPA1 deficiency aggravates necroptosis of alveolar epithelial cells through impairing mitochondrial function during ALI in mice. J Cell Physiol 2022; 237:3030-3043. [PMID: 35478455 DOI: 10.1002/jcp.30766] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/18/2022] [Accepted: 04/22/2022] [Indexed: 11/11/2022]
Abstract
Necroptosis, a recently described form of programmed cell death, is the main way of alveolar epithelial cells (AECs) death in acute lung injury (ALI). While the mechanism of how to trigger necroptosis in AECs during ALI has been rarely evaluated. Long optic atrophy protein 1 (L-OPA1) is a crucial mitochondrial inner membrane fusion protein, and its deficiency impairs mitochondrial function. This study aimed to investigate the role of L-OPA1 deficiency-mediated mitochondrial dysfunction in AECs necroptosis. We comprehensively investigated the detailed contribution and molecular mechanism of L-OPA1 deficiency in AECs necroptosis by inhibiting or activating L-OPA1. Firstly, our data showed that L-OPA1 expression was down-regulated in the lungs and AECs under the lipopolysaccharide (LPS) challenge. Furthermore, inhibition of L-OPA1 aggravated the pathological injury, inflammatory response, and necroptosis in the lungs of LPS-induced ALI mice. In vitro, inhibition of L-OPA1 induced necroptosis of AECs, while activation of L-OPA1 alleviated necroptosis of AECs under the LPS challenge. Mechanistically, inhibition of L-OPA1 aggravated necroptosis of AECs by inducing mitochondrial fragmentation and reducing mitochondrial membrane potential. While activation of L-OPA1 had the opposite effects. In summary, these findings indicate for the first time that L-OPA1 deficiency mediates mitochondrial fragmentation, induces necroptosis of AECs, and exacerbates ALI in mice. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Hui-Ling Jiang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Hui-Hui Yang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Yu-Biao Liu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Chen-Yu Zhang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Wen-Jing Zhong
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Xin-Xin Guan
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Ling Jin
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Jie-Ru Hong
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Jin-Tong Yang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Xiao-Hua Tan
- Experimental Center of Medical Morphology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Qing Li
- Department of Physiology, Hunan University of Medicine, Huaihua, Hunan, 418000, China
| | - Yong Zhou
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Cha-Xiang Guan
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| |
Collapse
|
26
|
Park J, Lee JJ, Hong Y, Seo H, Shin TS, Hong JY. Metagenomic Analysis of Plasma Microbial Extracellular Vesicles in Patients Receiving Mechanical Ventilation: A Pilot Study. J Pers Med 2022; 12:jpm12040564. [PMID: 35455680 PMCID: PMC9031263 DOI: 10.3390/jpm12040564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/29/2022] [Accepted: 03/30/2022] [Indexed: 02/05/2023] Open
Abstract
Background: Previous studies reported a significant association between pneumonia outcome and the respiratory microbiome. There is increasing interest in the roles of bacterial extracellular vesicles (EVs) in various diseases. We studied the composition and function of microbiota-derived EVs in the plasma of patients receiving mechanical ventilation to evaluate whether they can be used as a diagnostic marker and to predict clinical outcomes. Methods: Plasma samples (n = 111) from 59 mechanically ventilated patients (41 in the pneumonia group; 24 in the nursing home and hospital-associated infection [NHAI] group) were prospectively collected on days one and seven. After isolating the bacterial EVs from plasma samples, nucleic acid was extracted for 16S rRNA gene pyrosequencing. The samples were evaluated to determine the α and β diversity, bacterial composition, and predicted functions. Results: Principal coordinates analysis revealed significantly different clustering of microbial EVs between the pneumonia and non-pneumonia groups. The proportions of Lactobacillus, Cutibacterium, and Sphingomonas were significantly different between the pneumonia and non-pneumonia groups. In addition, the abundances of Lactobacillus and Bifidobacterium were significantly higher in the non-NHAI than the NHAI group. In the analysis of β diversity, the structure of microbial EVs differed significantly different between 28-day survivors and non-survivors (Bray-Curtis distance, p = 0.014). Functional profiling revealed significant differences between the pneumonia and non-pneumonia groups. The longitudinal change in predicted functions of microbial EV genes showed a significant difference between 28-day survivors and non-survivors. Conclusions: Bacterial microbiota–derived EVs in the plasma have potential as diagnostic and prognostic markers for patients receiving mechanical ventilation. Further large prospective studies are needed to determine the clinical utility of plasma microbiota-EVs in intubated patients.
Collapse
Affiliation(s)
- Jinkyeong Park
- Department of Pulmonary and Critical Care Medicine, Kyung Hee University Hospital at Gangdong, Kyung Hee University School of Medicine, Seoul 05278, Korea;
| | - Jae Jun Lee
- Institute of New Frontier Research Team, Hallym University College of Medicine, Chuncheon 24253, Korea;
| | - Yoonki Hong
- Department of Internal Medicine, School of Medicine, Kangwon National University, Kangwon National University Hospital, Chuncheon 24289, Korea;
| | - Hochan Seo
- MD Healthcare Inc., Seoul 03293, Korea; (H.S.); (T.-S.S.)
| | - Tae-Seop Shin
- MD Healthcare Inc., Seoul 03293, Korea; (H.S.); (T.-S.S.)
| | - Ji Young Hong
- Institute of New Frontier Research Team, Hallym University College of Medicine, Chuncheon 24253, Korea;
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Chuncheon Sacred Heart Hospital, Hallym University Medical Center, Chuncheon 24253, Korea
- Correspondence: ; Tel.: +82-33-240-8101; Fax: +033-255-6244
| |
Collapse
|
27
|
Yang S, Liu Q, Chen S, Zhang F, Li Y, Fan W, Mai L, He H, Huang F. Extracellular vesicles delivering nuclear factor I/C for hard tissue engineering: Treatment of apical periodontitis and dentin regeneration. J Tissue Eng 2022; 13:20417314221084095. [PMID: 35321254 PMCID: PMC8935403 DOI: 10.1177/20417314221084095] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 02/13/2022] [Indexed: 12/19/2022] Open
Abstract
Apical periodontitis (AP) causes arrest of tooth root development, which is associated with impaired odontoblastic differentiation of stem cells from apical papilla (SCAPs), but the underlying mechanism remains unclear. Here, we investigated roles of extracellular vesicle (EV) in AP and odontoblastic differentiation of SCAPs, moreover, a novel nuclear factor I/C (NFIC)-encapsulated EV was developed to promote dentin regeneration. We detected a higher expression of EV marker CD63 in inflamed apical papilla, and found that EVs from LPS-stimulated dental pulp cells suppressed odontoblastic differentiation of SCAPs through downregulating NFIC. Furthermore, we successfully constructed the NFIC-encapsulated EV by overexpressing NFIC in HEK293FT cells, which could upregulate cellular NFIC level in SCAPs, promoting the proliferation and migration of SCAPs, as well as dentinogenesis both in vitro and in vivo. Collectively, based on pathological roles of EV in AP, our study provides a novel strategy for dentin regeneration by exploiting EV to deliver NFIC.
Collapse
Affiliation(s)
- Shengyan Yang
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Department of Oral Anatomy and Physiology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Qing Liu
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Department of Oral Anatomy and Physiology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Shijing Chen
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Department of Oral Anatomy and Physiology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Fuping Zhang
- Department of Oral Anatomy and Physiology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Yaoyin Li
- Department of Oral Anatomy and Physiology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Department of Pediatric Dentistry, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Wenguo Fan
- Department of Oral Anatomy and Physiology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Lijia Mai
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Department of Oral Anatomy and Physiology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Hongwen He
- Department of Oral Anatomy and Physiology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Fang Huang
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Department of Oral Anatomy and Physiology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
28
|
STAT6/VDR Axis Mitigates Lung Inflammatory Injury by Promoting Nrf2 Signaling Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2485250. [PMID: 35047105 PMCID: PMC8763503 DOI: 10.1155/2022/2485250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 10/17/2021] [Accepted: 12/14/2021] [Indexed: 11/18/2022]
Abstract
Lung inflammatory injury is a global public health concern. It is characterized by infiltration of diverse inflammatory cells and thickening of pulmonary septum along with oxidative stress to airway epithelial cells. STAT6 is a nuclear transcription factor that plays a crucial role in orchestrating the immune response, but its function in tissue inflammatory injury has not been comprehensively studied. Here, we demonstrated that STAT6 activation can protect against particle-induced lung inflammatory injury by resisting oxidative stress. Specifically, genetic ablation of STAT6 was observed to worsen particle-induced lung injury mainly by disrupting the lungs' antioxidant capacity, as reflected by the downregulation of the Nrf2 signaling pathway, an increase in malondialdehyde levels, and a decrease in glutathione levels. Vitamin D receptor (VDR) has been previously proved to positively regulate Nrf2 signals. In this study, silencing VDR expression in human bronchial epithelial BEAS-2B cells consistently suppressed autophagy-mediated activation of the Nrf2 signaling pathway, thereby aggravating particle-induced cell damage. Mechanically, STAT6 activation promoted the nuclear translocation of VDR, which increased the transcription of autophagy-related genes and induced Nrf2 signals, and silencing VDR abolished these effects. Our research provides important insights into the role of STAT6 in oxidative damage and reveals its potential underlying mechanism. This information not only deepens the appreciation of STAT6 but also opens new avenues for the discovery of therapies for inflammatory respiratory system disorders.
Collapse
|
29
|
Naito Y, Kato H, Zhou L, Sugita S, He H, Zheng J, Hao Q, Sawa T, Lee JW. Therapeutic Effects of Hyaluronic Acid Against Cytotoxic Extracellular Vesicles Released During Pseudomonas Aeruginosa Pneumonia. Shock 2022; 57:408-416. [PMID: 34387224 PMCID: PMC8840981 DOI: 10.1097/shk.0000000000001846] [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: 11/26/2022]
Abstract
ABSTRACT Extracellular vesicles (EVs) have now been recognized as important mediators of cellular communication during injury and repair. We previously found that plasma EVs isolated from ex vivo perfused human lungs injured with Escherichia coli bacterial pneumonia were inflammatory, and exogenous administration of high molecular weight (HMW) hyaluronic acid (HA) as therapy bound to these EVs, decreasing inflammation and injury. In the current study, we studied the role of EVs released during severe Pseudomonas aeruginosa (PA) pneumonia in mice and determined whether intravenous administration of exogenous HMW HA would have therapeutic effects against the bacterial pneumonia. EVs were collected from the bronchoalveolar lavage fluid (BALF) of mice infected with PA103 by ultracentrifugation and analyzed by NanoSight and flow cytometry. In a cytotoxicity assay, administration of EVs released from infected mice (I-EVs) decreased the viability of A549 cells compared to EV isolated from sham control mice (C-EVs). Either exogenous HMW HA or an anti-CD44 antibody, when co-incubated with I-EVs, significantly improved the viability of the A549 cells. In mice with PA103 pneumonia, administration of HMW HA improved pulmonary edema and bacterial count in the lungs and decreased TNF-α and caspase-3 levels in the supernatant of lung homogenates. In conclusion, EVs isolated from BALF of mice with P. aeruginosa pneumonia were cytotoxic and inflammatory, and intravenous HMW HA administration was protective against P. aeruginosa pneumonia.
Collapse
Affiliation(s)
- Yoshifumi Naito
- Department of Anesthesiology, Kyoto Prefectural University of Medicine, Japan
| | - Hideya Kato
- Department of Anesthesiology, Kyoto Prefectural University of Medicine, Japan
| | - Li Zhou
- Department of Anesthesiology, University of California San Francisco, San Francisco, California
| | - Shinji Sugita
- Department of Anesthesiology, University of California San Francisco, San Francisco, California
| | - Hongli He
- Department of Anesthesiology, University of California San Francisco, San Francisco, California
| | - Justin Zheng
- Department of Anesthesiology, University of California San Francisco, San Francisco, California
| | - Qi Hao
- Department of Anesthesiology, University of California San Francisco, San Francisco, California
| | - Teiji Sawa
- Department of Anesthesiology, Kyoto Prefectural University of Medicine, Japan
| | - Jae-Woo Lee
- Department of Anesthesiology, University of California San Francisco, San Francisco, California
| |
Collapse
|
30
|
Finicelli M, Digilio FA, Galderisi U, Peluso G. The Emerging Role of Macrophages in Chronic Obstructive Pulmonary Disease: The Potential Impact of Oxidative Stress and Extracellular Vesicle on Macrophage Polarization and Function. Antioxidants (Basel) 2022; 11:antiox11030464. [PMID: 35326114 PMCID: PMC8944669 DOI: 10.3390/antiox11030464] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 02/24/2022] [Accepted: 02/25/2022] [Indexed: 12/20/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is one of the most common airway diseases, and it is considered a major global health problem. Macrophages are the most representative immune cells in the respiratory tract, given their role in surveying airways, removing cellular debris, immune surveillance, and resolving inflammation. Macrophages exert their functions by adopting phenotypical changes based on the stimuli they receive from the surrounding tissue. This plasticity is described as M1/M2 macrophage polarization, which consists of a strictly coordinated process leading to a difference in the expression of surface markers, the production of specific factors, and the execution of biological activities. This review focuses on the role played by macrophages in COPD and their implication in inflammatory and oxidative stress processes. Particular attention is on macrophage polarization, given macrophage plasticity is a key feature in COPD. We also discuss the regulatory influence of extracellular vesicles (EVs) in cell-to-cell communications. EV composition and cargo may influence many COPD-related aspects, including inflammation, tissue remodeling, and macrophage dysfunctions. These findings could be useful for better addressing the role of macrophages in the complex pathogenesis and outcomes of COPD.
Collapse
Affiliation(s)
- Mauro Finicelli
- Research Institute on Terrestrial Ecosystems (IRET), National Research Council of Italy (CNR), Via Pietro Castellino 111, 80131 Naples, Italy;
- Correspondence: (M.F.); (G.P.); Tel.: +39-0816132553 (M.F.); +39-0816132280 (G.P.)
| | - Filomena Anna Digilio
- Research Institute on Terrestrial Ecosystems (IRET), National Research Council of Italy (CNR), Via Pietro Castellino 111, 80131 Naples, Italy;
| | - Umberto Galderisi
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Via Santa Maria di Costantinopoli 16, 80138 Naples, Italy;
| | - Gianfranco Peluso
- Research Institute on Terrestrial Ecosystems (IRET), National Research Council of Italy (CNR), Via Pietro Castellino 111, 80131 Naples, Italy;
- Faculty of Medicine and Surgery, Saint Camillus International University of Health Sciences, Via di Sant’Alessandro 8, 00131 Rome, Italy
- Correspondence: (M.F.); (G.P.); Tel.: +39-0816132553 (M.F.); +39-0816132280 (G.P.)
| |
Collapse
|
31
|
Liang R, Tong X, Dong Z, Qin W, Fan L, Bai Z, Zhang Z, Xiang T, Wang Z, Tan N. Suhuang antitussive capsule ameliorates post-infectious cough in mice through AhR-Nrf2 pathway. JOURNAL OF ETHNOPHARMACOLOGY 2022; 283:114664. [PMID: 34555451 DOI: 10.1016/j.jep.2021.114664] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 09/08/2021] [Accepted: 09/18/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Suhuang antitussive capsule (SH capsule), a typical traditional Chinese medicines (TCMs) compound, is widely used for the treatment of post-infectious cough (PIC) in the clinic. Our previous studies have demonstrated that SH capsule possesses significant ameliorative effects on cough variant asthma (CVA), sputum obstruction and airway remodeling. AIM OF THE STUDY This study is designed to investigate the ameliorative effects and potential mechanisms of SH capsule on PIC in mice. MATERIALS AND METHODS To establish the PIC model, ICR mice were induced by lipopolysaccharide (LPS) (3 mg/kg) once, followed by cigarettes smoke (CS) exposure for 30 min per day for 30 days. Mice were intragastrically (i.g.) administrated with SH capsule at the doses of 3.5, 7, 14 g/kg each day for 2 weeks since the 24th day. The number of coughs, coughs latencies, enzyme-linked immunosorbent assay (ELISA) and histological analysis were used to investigate the effects of SH capsule on PIC mice. Quantitative-polymerase chain reaction (Q-PCR) and western blotting were conducted to evaluate the levels of mRNA and proteins associated with Aryl hydrocarbon receptor (AhR)-NF-E2-related factor 2 (Nrf2) pathway. Superoxide dismutase (SOD), glutathione (GSH) and total antioxidant capacity (T-AOC) assays were performed to evaluate the oxidative stress. A549 cells were used to investigate the ameliorative effects of SH capsule in vitro. RESULTS SH capsule effectively diminished the number of coughs and extended coughs latencies in PIC mice. The airway inflammation was alleviated by decreasing the expression levels of inflammatory mediators including tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β) and interleukin-6 (IL-6). Moreover, SH capsule dose-dependently activated AhR-Nrf2 pathway and induced the nuclear translocation in vitro and in vivo. Besides, SH capsule significantly increased the levels of SOD, GSH and T-AOC in mice. CONCLUSION Our study demonstrates that SH capsule ameliorates airway inflammation-associated PIC in mice through activating AhR-Nrf2 pathway and consequently alleviating inflammatory responses and oxidative stress.
Collapse
Affiliation(s)
- Rongyao Liang
- Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China.
| | - Xiyang Tong
- Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China.
| | - Zhikui Dong
- Beijing Haiyan Pharmaceutical Co., Ltd., Yangtze River Pharmaceutical Group, Beijing, 102206, PR China; Jiangsu Longfengtang Traditional Chinese Medicine Co., Ltd., Yangtze River Pharmaceutical Group, Taizhou, 225321, PR China.
| | - Weiwei Qin
- Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China.
| | - Lingling Fan
- Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China.
| | - Ziyu Bai
- Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China.
| | - Zhihao Zhang
- Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China.
| | - Ting Xiang
- Beijing Haiyan Pharmaceutical Co., Ltd., Yangtze River Pharmaceutical Group, Beijing, 102206, PR China.
| | - Zhen Wang
- Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China.
| | - Ninghua Tan
- Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China.
| |
Collapse
|
32
|
Quanzhenyiqitang Reverses LPS-Induced Inflammation via Inhibiting PYK2/p38MAPK/HDAC2/CK2 Signaling Pathway in Rat Alveolar Macrophage. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:7857022. [PMID: 35047050 PMCID: PMC8763520 DOI: 10.1155/2022/7857022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 11/20/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a common chronic pulmonary disease with multiple etiologies and pathological changes. PYK2 expression is significantly increased in lipopolysaccharide-induced lung injury, but it mediates chronic lung inflammation. The mechanism of its occurrence remains unclear. Quanzhenyiqitang is often used in clinical treatment of COPD, so this study explored the mechanism of its treatment of lipopolysaccharide-induced lung injury. In this study, transfection, flow cytometry, QRT-PCR, and Western blotting methods were used to study the mechanism of Quanzhenyiqitang lipopolysaccharide-induced lung injury. The results showed that the mechanism of occurrence remains unclear. Our novel observations imply that the PYK2/p38MAPK/HDAC2/CK2 pathway is one of the fundamental underlying mechanisms that mediate the pathogenic progression of COPD, and Quanzhenyiqitang may be the therapeutic drug to prevent chronic inflammation and delay the progression of COPD by inhibiting PYK2 signaling pathways.
Collapse
|
33
|
Esquivel-Ruiz S, González-Rodríguez P, Lorente JA, Pérez-Vizcaíno F, Herrero R, Moreno L. Extracellular Vesicles and Alveolar Epithelial-Capillary Barrier Disruption in Acute Respiratory Distress Syndrome: Pathophysiological Role and Therapeutic Potential. Front Physiol 2021; 12:752287. [PMID: 34887773 PMCID: PMC8650589 DOI: 10.3389/fphys.2021.752287] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 09/27/2021] [Indexed: 12/11/2022] Open
Abstract
Extracellular vesicles (EVs) mediate intercellular communication by transferring genetic material, proteins and organelles between different cells types in both health and disease. Recent evidence suggests that these vesicles, more than simply diagnostic markers, are key mediators of the pathophysiology of acute respiratory distress syndrome (ARDS) and other lung diseases. In this review, we will discuss the contribution of EVs released by pulmonary structural cells (alveolar epithelial and endothelial cells) and immune cells in these diseases, with particular attention to their ability to modulate inflammation and alveolar-capillary barrier disruption, a hallmark of ARDS. EVs also offer a unique opportunity to develop new therapeutics for the treatment of ARDS. Evidences supporting the ability of stem cell-derived EVs to attenuate the lung injury and ongoing strategies to improve their therapeutic potential are also discussed.
Collapse
Affiliation(s)
- Sergio Esquivel-Ruiz
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.,Ciber de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Paloma González-Rodríguez
- Ciber de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Department of Critical Care, Hospital Universitario de Getafe, Madrid, Spain
| | - José A Lorente
- Ciber de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Department of Critical Care, Hospital Universitario de Getafe, Madrid, Spain.,Clinical Section, School of Medicine, European University of Madrid, Madrid, Spain
| | - Francisco Pérez-Vizcaíno
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.,Ciber de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Raquel Herrero
- Ciber de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Department of Critical Care, Hospital Universitario de Getafe, Madrid, Spain
| | - Laura Moreno
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.,Ciber de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| |
Collapse
|
34
|
Choudhary I, Vo T, Paudel K, Wen X, Gupta R, Kesimer M, Patial S, Saini Y. Vesicular and extravesicular protein analyses from the airspaces of ozone-exposed mice revealed signatures associated with mucoinflammatory lung disease. Sci Rep 2021; 11:23203. [PMID: 34853335 PMCID: PMC8636509 DOI: 10.1038/s41598-021-02256-5] [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: 12/20/2020] [Accepted: 11/08/2021] [Indexed: 12/16/2022] Open
Abstract
Lung epithelial lining fluid (ELF) harbors a variety of proteins that influence homeostatic and stress responses in the airspaces. Exosomes, nano-sized extracellular vesicles, contain many proteins that vary in abundance and composition based on the prevailing conditions. Ozone causes inflammatory responses in the airspaces of experimental animals and humans. However, the exosomal protein signatures contained within the ELF from ozone-exposed lung airspaces remain poorly characterized. To explore this, we hypothesized that ozone triggers the release of exosome-bound inflammatory proteins from various cells that reflect mucoobstructive lung disease. Accordingly, we repetitively exposed adult male and female C57BL/6 mice to HEPA-filtered air (air) or 0.8 ppm ozone (4 h per day) for 14 days (five consecutive days of exposure, 2 days of rest, five consecutive days of exposure, 2 days of rest, four consecutive days of exposure). Exosome-bound proteomic signatures, as well as the levels of soluble inflammatory mediators in the bronchoalveolar lavage fluid (BALF), were determined 12-16 h after the last exposure. Principal component analyses of the exosome-bound proteome revealed a clear distinction between air-exposed and ozone-exposed mice, as well as between ozone-exposed males and ozone-exposed females. In addition to 575 proteins that were enriched in both sexes upon ozone exposure, 243 and 326 proteins were enriched uniquely in ozone-exposed males and females, respectively. Ingenuity pathway analyses on enriched proteins between ozone- and air-exposed mice revealed enrichment of pro-inflammatory pathways. More specifically, macrophage activation-related proteins were enriched in exosomes from ozone-exposed mice. Cytokine analyses on the BALF revealed elevated levels of G-CSF, KC, IP-10, IL-6, and IL-5 in ozone-exposed mice. Finally, the histopathological assessment revealed significantly enhanced intracellular localization of mucoinflammatory proteins including MUC5B and FIZZ1 in ozone-exposed mice in a cell-specific manner indicating the cellular sources of the proteins that are ferried in the exosomes upon ozone-induced lung injury. Collectively, this study identified exosomal, secretory, and cell-specific proteins and biological pathways following repetitive exposure of mice to ozone.
Collapse
Affiliation(s)
- Ishita Choudhary
- grid.64337.350000 0001 0662 7451Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, 1909 Skip Bertman Drive, Baton Rouge, LA 70803 USA
| | - Thao Vo
- grid.64337.350000 0001 0662 7451Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, 1909 Skip Bertman Drive, Baton Rouge, LA 70803 USA
| | - Kshitiz Paudel
- grid.64337.350000 0001 0662 7451Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, 1909 Skip Bertman Drive, Baton Rouge, LA 70803 USA
| | - Xue Wen
- grid.64337.350000 0001 0662 7451Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803 USA
| | - Richa Gupta
- grid.10698.360000000122483208Department of Pathology and Laboratory Medicine, UNC School of Medicine, Chapel Hill, NC 27510 USA
| | - Mehmet Kesimer
- grid.10698.360000000122483208Department of Pathology and Laboratory Medicine, UNC School of Medicine, Chapel Hill, NC 27510 USA
| | - Sonika Patial
- grid.64337.350000 0001 0662 7451Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, 1909 Skip Bertman Drive, Baton Rouge, LA 70803 USA
| | - Yogesh Saini
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, 1909 Skip Bertman Drive, Baton Rouge, LA, 70803, USA.
| |
Collapse
|
35
|
Kanda K, Asawa Y, Inaki R, Fujihara Y, Hoshi K, Hikita A. Requirement of direct contact between chondrocytes and macrophages for the maturation of regenerative cartilage. Sci Rep 2021; 11:22476. [PMID: 34795319 PMCID: PMC8602279 DOI: 10.1038/s41598-021-01437-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 10/27/2021] [Indexed: 11/09/2022] Open
Abstract
Regenerative cartilage prepared from cultured chondrocytes is generally immature in vitro and matures after transplantation. Although many factors, including host cells and humoral factors, have been shown to affect cartilage maturation in vivo, the requirement of direct cell-cell contact between host and donor cells remains to be verified. In this study, we examined the host cells that promote cartilage maturation via cell-cell contact. Based on analysis of the transplanted chondrocytes, we examined the contribution of endothelial cells and macrophages. Using a semiclosed device that is permeable to tissue fluids while blocking host cells, we selectively transplanted chondrocytes and HUVECs or untreated/M1-polarized/M2-polarized RAW264.7 cells. As a result, untreated RAW264.7 cells induced cartilage regeneration. Furthermore, an in vitro coculture assay indicated communication between chondrocytes and RAW264.7 cells mediated by RNA, suggesting the involvement of extracellular vesicles in this process. These findings provide insights for establishing a method of in vitro cartilage regeneration.
Collapse
Affiliation(s)
- Kengo Kanda
- Department of Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yukiyo Asawa
- Department of Tissue Engineering, The University of Tokyo Hospital, Tokyo, Japan
| | - Ryoko Inaki
- Department of Tissue Stem Cell and Dental Life Science, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yuko Fujihara
- Department of Oral-Maxillofacial Surgery, Dentistry and Orthodontics, The University of Tokyo Hospital, Tokyo, Japan
| | - Kazuto Hoshi
- Department of Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Tissue Engineering, The University of Tokyo Hospital, Tokyo, Japan
- Department of Oral-Maxillofacial Surgery, Dentistry and Orthodontics, The University of Tokyo Hospital, Tokyo, Japan
| | - Atsuhiko Hikita
- Department of Tissue Engineering, The University of Tokyo Hospital, Tokyo, Japan.
| |
Collapse
|
36
|
Xia Z, Qing B, Wang W, Gu L, Chen H, Yuan Y. Formation, contents, functions of exosomes and their potential in lung cancer diagnostics and therapeutics. Thorac Cancer 2021; 12:3088-3100. [PMID: 34734680 PMCID: PMC8636224 DOI: 10.1111/1759-7714.14217] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/18/2021] [Accepted: 10/19/2021] [Indexed: 02/06/2023] Open
Abstract
Lung cancer is the leading cause of cancer-related death worldwide due to diagnosis in the advanced stage and drug resistance in the subsequent treatments. Development of novel diagnostic and therapeutic methods is urged to improve the disease outcome. Exosomes are nano-sized vehicles which transport different types of biomolecules intercellularly, including DNA, RNA and proteins, and are implicated in cross-talk between cells and their surrounding microenvironment. Tumor-derived exosomes (TEXs) have been revealed to strongly influence the tumor microenvironment, antitumor immunoregulatory activities, tumor progression and metastasis. Potential of TEXs as biomarkers for lung cancer diagnosis, prognosis and treatment prediction is supported by numerous studies. Moreover, exosomes have been proposed to be promising drug carriers. Here, we review the mechanisms of exosomal formation and uptake, the functions of exosomes in carcinogenesis, and potential clinical utility of exosomes as biomarkers, tumor vaccine and drug delivery vehicles in the diagnosis and therapeutics of lung cancer.
Collapse
Affiliation(s)
- Zhenkun Xia
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Bei Qing
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Wei Wang
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Linguo Gu
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Hongzuo Chen
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yunchang Yuan
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
37
|
Emodin attenuates severe acute pancreatitis-associated acute lung injury by suppressing pancreatic exosome-mediated alveolar macrophage activation. Acta Pharm Sin B 2021; 12:3986-4003. [PMID: 36213542 PMCID: PMC9532455 DOI: 10.1016/j.apsb.2021.10.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 09/30/2021] [Accepted: 10/03/2021] [Indexed: 12/23/2022] Open
Abstract
Severe acute pancreatitis-associated acute lung injury (SAP-ALI) is a serious disease associated with high mortality. Emodin has been applied to alleviate SAP-ALI; however, the mechanism remains unclear. We report that the therapeutic role of emodin in attenuating SAP-ALI is partly dependent on an exosomal mechanism. SAP rats had increased levels of plasma exosomes with altered protein contents compared to the sham rats. These infused plasma exosomes tended to accumulate in the lungs and promoted the hyper-activation of alveolar macrophages and inflammatory damage. Conversely, emodin treatment decreased the plasma/pancreatic exosome levels in the SAP rats. Emodin-primed exosomes showed less pro-inflammatory effects in alveolar macrophages and lung tissues than SAP exosomes. In detail, emodin-primed exosomes suppressed the NF-κB pathway to reduce the activation of alveolar macrophage and ameliorate lung inflammation by regulating PPARγ pathway, while these effects were amplified/abolished by PPARγ agonist/antagonist. Blockage of pancreatic acinar cell exosome biogenesis also exhibited suppression of alveolar macrophage activation and reduction of lung inflammation. This study suggests a vital role of exosomes in participating inflammation-associated organ-injury, and indicates emodin can attenuate SAP-ALI by reducing the pancreatic exosome-mediated alveolar macrophage activation.
Collapse
|
38
|
Yang Y, Yuan L, Du X, Zhou K, Qin L, Wang L, Yang M, Wu M, Zheng Z, Xiang Y, Qu X, Liu H, Qin X, Liu C. Involvement of epithelia-derived exosomes in chronic respiratory diseases. Biomed Pharmacother 2021; 143:112189. [PMID: 34560534 DOI: 10.1016/j.biopha.2021.112189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 09/07/2021] [Accepted: 09/10/2021] [Indexed: 12/20/2022] Open
Abstract
Exosomes are tiny membrane lipid bilayer vesicles (φ40-100 nm) formed by the fusion of multivesicular bodies with plasma membrane, which are released extracellular by exocytosis. As natural nanocarriers, exosomes contain a variety of signal substances of the mother cell: nucleic acids, proteins and lipids, etc., which always play a vital role in the transmission of signal molecules between different cells. Epithelial cells are the first-line defense system against various inhaled allergens causing chronic respiratory diseases (CRD), such as asthma and chronic obstructive pulmonary disease (COPD). It's noted that increasing literature shows the exosomes derived from epithelial cells are involved in the pathogenesis of CRD. Moreover, the correlations between exosome cargo and the disease phenotypes show a high potential of using exosomes as biomarkers of CRD. In this review, we mainly focus on the physiological functions of epithelial-derived exosomes and illustrate the involved mechanism of epithelial-derived exosomes in common CRD.
Collapse
Affiliation(s)
- Yu Yang
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China; Basic and Clinical Research Laboratory of Major Respiratory Diseases, Central South University, Changsha, Hunan, China
| | - Lin Yuan
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China
| | - Xizi Du
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China
| | - Kai Zhou
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China
| | - Ling Qin
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, China; Basic and Clinical Research Laboratory of Major Respiratory Diseases, Central South University, Changsha, Hunan, China
| | - Leyuan Wang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China
| | - Ming Yang
- Centre for Asthma and Respiratory Disease, School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle and Hunter Medical Research Institute, Callaghan, New South Wales, Australia
| | - Mengping Wu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China
| | - Zhiyuan Zheng
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, China; Basic and Clinical Research Laboratory of Major Respiratory Diseases, Central South University, Changsha, Hunan, China
| | - Yang Xiang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China
| | - Xiangping Qu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China
| | - Huijun Liu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China
| | - Xiaoqun Qin
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China
| | - Chi Liu
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China; Research Center of China-Africa Infectious Diseases, Xiangya School of Medicine Central South University, Changsha, Hunan, China.
| |
Collapse
|
39
|
The mechanism of nicotinamide on reducing acute lung injury by inhibiting MAPK and NF-κB signal pathway. Mol Med 2021; 27:115. [PMID: 34544355 PMCID: PMC8451170 DOI: 10.1186/s10020-021-00376-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 09/08/2021] [Indexed: 01/17/2023] Open
Abstract
Background Acute lung injury is an important factor that leads to the death of patients with pneumonia. Previous studies have shown that nicotinamide (NAM) plays a role in reducing cell damage, so this study explored the mechanism by which NAM functions in acute lung injury. Methods We explored the mechanism by which NAM affects acute lung injury in vivo and in vitro by qRT-PCR, western blotting and ELISA. Results The results showed that NAM could significantly reduce lung injury and proinflammatory mediator accumulation. Further mechanistic studies showed that NAM could significantly inhibit the MAPK and AKT/NF-κB signaling pathways. Conclusion These results suggested that NAM may reduce the release of proinflammatory mediators by inhibiting the MAPK and AKT/NF-κB signaling pathways and ultimately alleviate lung injury. Supplementary Information The online version contains supplementary material available at 10.1186/s10020-021-00376-2.
Collapse
|
40
|
Sui X, Liu W, Liu Z. Exosomes derived from LPS-induced MHs cells prompted an inflammatory response in sepsis-induced acute lung injury. Respir Physiol Neurobiol 2021; 292:103711. [PMID: 34091074 DOI: 10.1016/j.resp.2021.103711] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/30/2021] [Accepted: 06/02/2021] [Indexed: 01/04/2023]
Abstract
Exosome is a novel tool with an essential role in cell communication. However, its role in the pathogenesis of sepsis-induced acute lung injury is currently unknown. Here, we first found that lipopolysaccharide (LPS) could up-regulate the expression of pro-inflammatory cytokines and promote exosomes release in the murine alveolar macrophage cell line (MHs cells). Moreover, we found MHs cells derived exosomes also maintain the pro-inflammatory effect after LPS stimulation. Treating with hydrochloride hydrate (GW4869) could dose-dependently downregulated the release of exosomes and inhibited the upregulation of inflammatory cytokines in MHs cells with LPS treatment. Also, we further identified GW4869 administration induced the remission of histopathologic changes, the reduction of pro-inflammatory cytokines in lung tissue, and inhibit serum exosomes release. These results indicate that the downregulation of exosome release by GW4869 might protect lung tissue from LPS induced injury through the suppression of excessive inflammatory responses, suggesting its potential therapeutic effects on sepsis-induced acute lung injury.
Collapse
Affiliation(s)
- Xintong Sui
- Emergency Department, First Hospital of China Medical University, Shenyang City, 110001, Liaoning Province, China.
| | - Wei Liu
- Emergency Department, First Hospital of China Medical University, Shenyang City, 110001, Liaoning Province, China.
| | - Zhi Liu
- Emergency Department, First Hospital of China Medical University, Shenyang City, 110001, Liaoning Province, China.
| |
Collapse
|
41
|
Hu Q, Lyon CJ, Fletcher JK, Tang W, Wan M, Hu TY. Extracellular vesicle activities regulating macrophage- and tissue-mediated injury and repair responses. Acta Pharm Sin B 2021; 11:1493-1512. [PMID: 34221864 PMCID: PMC8245807 DOI: 10.1016/j.apsb.2020.12.014] [Citation(s) in RCA: 122] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 12/04/2020] [Accepted: 12/09/2020] [Indexed: 02/08/2023] Open
Abstract
Macrophages are typically identified as classically activated (M1) macrophages and alternatively activated (M2) macrophages, which respectively exhibit pro- and anti-inflammatory phenotypes, and the balance between these two subtypes plays a critical role in the regulation of tissue inflammation, injury, and repair processes. Recent studies indicate that tissue cells and macrophages interact via the release of small extracellular vesicles (EVs) in processes where EVs released by stressed tissue cells can promote the activation and polarization of adjacent macrophages which can in turn release EVs and factors that can promote cell stress and tissue inflammation and injury, and vice versa. This review discusses the roles of such EVs in regulating such interactions to influence tissue inflammation and injury in a number of acute and chronic inflammatory disease conditions, and the potential applications, advantage and concerns for using EV-based therapeutic approaches to treat such conditions, including their potential role of drug carriers for the treatment of infectious diseases.
Collapse
Key Words
- ADSCs, adipose-derived stem cells
- AKI, acute kidney injury
- ALI, acute lung injury
- AMs, alveolar macrophages
- BMSCs, bone marrow stromal cells
- CLP, cecal ligation and puncture
- DSS, dextran sodium sulphate
- EVs, extracellular vesicles
- Extracellular vesicles
- HSPA12B, heat shock protein A12B
- HUCMSCs, human umbilical cord mesenchymal stem cells
- IBD, inflammatory bowel disease
- ICAM-1, intercellular adhesion molecule 1
- IL-1β, interleukin-1β
- Inflammatory disease
- Interaction loop
- KCs, Kupffer cells
- KLF4, krüppel-like factor 4
- LPS, lipopolysaccharides
- MHC, major histocompatibility complex
- MSCs, mesenchymal stromal cells
- MVs, microvesicles
- Macrophage
- PEG, polyethylene glycol
- PMFA, 5,7,30,40,50-pentamethoxyflavanone
- PPARγ, peroxisome proliferator-activated receptor γ
- SIRPα, signal regulatory protein α
- Sepsis
- Stem cell
- TECs, tubular epithelial cells
- TNF, tumor necrosis factor
- TRAIL, tumor necrosis factor-related apoptosis-inducing ligand
- Targeted therapy
- Tissue injury
- iNOS, inducible nitrogen oxide synthase
Collapse
|
42
|
Mukai A, Otsuki Y, Ito E, Fujita T, Ueno M, Maeda T, Kinoshita S, Sotozono C, Hamuro J. Mitochondrial miRNA494-3p in extracellular vesicles participates in cellular interplay of iPS-Derived human retinal pigment epithelium with macrophages. Exp Eye Res 2021; 208:108621. [PMID: 34000275 DOI: 10.1016/j.exer.2021.108621] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 04/22/2021] [Accepted: 05/09/2021] [Indexed: 12/14/2022]
Abstract
To explore new molecular targets for therapy in human model systems by discerning the role of extracellular vesicle (EV) microRNAs (miRs) secreted by human retinal pigment epithelium (hRPE) cells and their cellular interplay with macrophages (Mps). Human Mps differentiated from THP-1 cells stimulated by phorbol myristate acetate were co-cultured with induced pluripotent stem cell-derived differentiated hRPE (iPS-hRPE) cells in Transwell® system separated by 0.40 μm or 0.03 μm filters. EV-associated CD63+ proteins (CD63+ EV) were detected by western blotting, and secreted EVs were analyzed by Nanosight tracking. The miR profiles of the secreted EVs were determined using 3D-gene human microRNA chips (Toray Industries, Inc.). Levels of CD63+ EV were increased in co-cultures concomitantly with the increased production of EV particles (50-150 nm). The increased production of EVs was associated with higher production of MCP-1, IL-6, IL-8 from hRPE cells, and VEGF and repressed production of TNF-α from Mps and pigment epithelium-derived factor (PEDF) from RPE cells. Ultracentrifugation of semi-purified EVs increased the secretion of these pro-inflammatory cytokines and EV particles from hRPE cells, but this effect was eliminated in transwells equipped with 0.03 μm filters, whereas no repression of PEDF and TNF-α secretion occurred. 3D-gene miR analysis revealed a selective increase in secretion of miR494-3p in EVs from iPS-hRPE cells during the interplay with Mps. The miRs in EVs secreted by hRPE cells may have a critical role in the vicious inflammatory cycle, whereas repression of TNF-α and PEDF require cell-to-cell contact that is independent of EVs or exosomes. MiR494-3p may be a candidate molecular target of diagnosis and therapy for age-related macular degeneration.
Collapse
Affiliation(s)
- Atsushi Mukai
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Hirokoji-agaru, Kawaramachi-dori, Kamigyo-ku, Kyoto 602-0841, Japan
| | - Yohei Otsuki
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Hirokoji-agaru, Kawaramachi-dori, Kamigyo-ku, Kyoto 602-0841, Japan
| | - Eiko Ito
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Hirokoji-agaru, Kawaramachi-dori, Kamigyo-ku, Kyoto 602-0841, Japan
| | - Tomoko Fujita
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Hirokoji-agaru, Kawaramachi-dori, Kamigyo-ku, Kyoto 602-0841, Japan
| | - Morio Ueno
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Hirokoji-agaru, Kawaramachi-dori, Kamigyo-ku, Kyoto 602-0841, Japan
| | - Tadao Maeda
- Kobe Eye Center Hospital, 2-1-8 Minatojima-minami-cho, Chuo-ku, Kobe, 650-0047, Japan
| | - Shigeru Kinoshita
- Department of Frontier Medical Science and Technology for Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Chie Sotozono
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Hirokoji-agaru, Kawaramachi-dori, Kamigyo-ku, Kyoto 602-0841, Japan
| | - Junji Hamuro
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Hirokoji-agaru, Kawaramachi-dori, Kamigyo-ku, Kyoto 602-0841, Japan.
| |
Collapse
|
43
|
Teng Y, Xu F, Zhang X, Mu J, Sayed M, Hu X, Lei C, Sriwastva M, Kumar A, Sundaram K, Zhang L, Park JW, Chen SY, Zhang S, Yan J, Merchant ML, Zhang X, McClain CJ, Wolfe JK, Adcock RS, Chung D, Palmer KE, Zhang HG. Plant-derived exosomal microRNAs inhibit lung inflammation induced by exosomes SARS-CoV-2 Nsp12. Mol Ther 2021; 29:2424-2440. [PMID: 33984520 PMCID: PMC8110335 DOI: 10.1016/j.ymthe.2021.05.005] [Citation(s) in RCA: 117] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 03/28/2021] [Accepted: 05/05/2021] [Indexed: 12/19/2022] Open
Abstract
Lung inflammation is a hallmark of coronavirus disease 2019 (COVID-19). In this study, we show that mice develop inflamed lung tissue after being administered exosomes released from the lung epithelial cells exposed to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Nsp12 and Nsp13 (exosomesNsp12Nsp13). Mechanistically, we show that exosomesNsp12Nsp13 are taken up by lung macrophages, leading to activation of nuclear factor κB (NF-κB) and the subsequent induction of an array of inflammatory cytokines. Induction of tumor necrosis factor (TNF)-α, interleukin (IL)-6, and IL-1β from exosomesNsp12Nsp13-activated lung macrophages contributes to inducing apoptosis in lung epithelial cells. Induction of exosomesNsp12Nsp13-mediated lung inflammation was abolished with ginger exosome-like nanoparticle (GELN) microRNA (miRNA aly-miR396a-5p. The role of GELNs in inhibition of the SARS-CoV-2-induced cytopathic effect (CPE) was further demonstrated via GELN aly-miR396a-5p- and rlcv-miR-rL1-28-3p-mediated inhibition of expression of Nsp12 and spike genes, respectively. Taken together, our results reveal exosomesNsp12Nsp13 as potentially important contributors to the development of lung inflammation, and GELNs are a potential therapeutic agent to treat COVID-19.
Collapse
Affiliation(s)
- Yun Teng
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA.
| | - Fangyi Xu
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA
| | - Xiangcheng Zhang
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA; Department of ICU, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu 223300, China
| | - Jingyao Mu
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA
| | - Mohammed Sayed
- Department of Computer Engineering and Computer Science, University of Louisville, Louisville, KY 40202, USA
| | - Xin Hu
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Chao Lei
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA
| | - Mukesh Sriwastva
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA
| | - Anil Kumar
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA
| | - Kumaran Sundaram
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA
| | - Lifeng Zhang
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA
| | - Juw Won Park
- Department of Computer Engineering and Computer Science, University of Louisville, Louisville, KY 40202, USA; KBRIN Bioinformatics Core, University of Louisville, Louisville, KY 40202, USA
| | - Shao-Yu Chen
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Shuangqin Zhang
- Peeples Cancer Institute at Hamilton Medical Center, Dalton, GA 30720, USA
| | - Jun Yan
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA
| | - Michael L Merchant
- Kidney Disease Program and Clinical Proteomics Center, University of Louisville, Louisville, KY 40202, USA
| | - Xiang Zhang
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Craig J McClain
- Robley Rex Veterans Affairs Medical Center, Louisville, KY 40206, USA; Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Jennifer K Wolfe
- Center for Predictive Medicine for Emerging Infectious Diseases, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Robert S Adcock
- Center for Predictive Medicine for Emerging Infectious Diseases, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Donghoon Chung
- Department of Microbiology & Immunology, University of Louisville, Louisville, KY 40202, USA; Center for Predictive Medicine for Emerging Infectious Diseases, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Kenneth E Palmer
- Center for Predictive Medicine for Emerging Infectious Diseases, School of Medicine, University of Louisville, Louisville, KY 40202, USA; Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Huang-Ge Zhang
- Robley Rex Veterans Affairs Medical Center, Louisville, KY 40206, USA; Department of Microbiology & Immunology, University of Louisville, Louisville, KY 40202, USA; James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA.
| |
Collapse
|
44
|
Balbi C, Burrello J, Bolis S, Lazzarini E, Biemmi V, Pianezzi E, Burrello A, Caporali E, Grazioli LG, Martinetti G, Fusi-Schmidhauser T, Vassalli G, Melli G, Barile L. Circulating extracellular vesicles are endowed with enhanced procoagulant activity in SARS-CoV-2 infection. EBioMedicine 2021; 67:103369. [PMID: 33971404 PMCID: PMC8104913 DOI: 10.1016/j.ebiom.2021.103369] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/26/2021] [Accepted: 04/14/2021] [Indexed: 12/29/2022] Open
Abstract
Background Coronavirus-2 (SARS-CoV-2) infection causes an acute respiratory syndrome accompanied by multi-organ damage that implicates a prothrombotic state leading to widespread microvascular clots. The causes of such coagulation abnormalities are unknown. The receptor tissue factor, also known as CD142, is often associated with cell-released extracellular vesicles (EV). In this study, we aimed to characterize surface antigens profile of circulating EV in COVID-19 patients and their potential implication as procoagulant agents. Methods We analyzed serum-derived EV from 67 participants who underwent nasopharyngeal swabs molecular test for suspected SARS-CoV-2 infection (34 positives and 33 negatives) and from 16 healthy controls (HC), as referral. A sub-analysis was performed on subjects who developed pneumonia (n = 28). Serum-derived EV were characterized for their surface antigen profile and tested for their procoagulant activity. A validation experiment was performed pre-treating EV with anti-CD142 antibody or with recombinant FVIIa. Serum TNF-α levels were measured by ELISA. Findings Profiling of EV antigens revealed a surface marker signature that defines circulating EV in COVID-19. A combination of seven surface molecules (CD49e, CD209, CD86, CD133/1, CD69, CD142, and CD20) clustered COVID (+) versus COVID (-) patients and HC. CD142 showed the highest discriminating performance at both multivariate models and ROC curve analysis. Noteworthy, we found that CD142 exposed onto surface of EV was biologically active. CD142 activity was higher in COVID (+) patients and correlated with TNF-α serum levels. Interpretation In SARS-CoV-2 infection the systemic inflammatory response results in cell-release of substantial amounts of procoagulant EV that may act as clotting initiation agents, contributing to disease severity. Funding Cardiocentro Ticino Institute, Ente ospedaliero Cantonale, Lugano-Switzerland.
Collapse
Affiliation(s)
- Carolina Balbi
- Laboratory of Cellular and Molecular Cardiology, Istituto Cardiocentro Ticino, Ente Ospedaliero Cantonale, Lugano, Switzerland; Center for Molecular Cardiology, Zurich, Switzerland
| | - Jacopo Burrello
- Laboratory for Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Ente Ospedaliero Cantonale Lugano, Switzerland
| | - Sara Bolis
- Laboratory of Cellular and Molecular Cardiology, Istituto Cardiocentro Ticino, Ente Ospedaliero Cantonale, Lugano, Switzerland; Laboratory for Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Ente Ospedaliero Cantonale Lugano, Switzerland
| | - Edoardo Lazzarini
- Laboratory for Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Ente Ospedaliero Cantonale Lugano, Switzerland
| | - Vanessa Biemmi
- Laboratory for Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Ente Ospedaliero Cantonale Lugano, Switzerland
| | - Enea Pianezzi
- Laboratory of Microbiology, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Alessio Burrello
- Department of Electrical, Electronic and Information Engineering (DEI), University of Bologna, Bologna, Italy
| | - Elena Caporali
- Cardiology Department, Istituto Cardiocentro Ticino, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Lorenzo Gauthier Grazioli
- Internal Medicine Department, Ospedale Regionale di Lugano, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Gladys Martinetti
- Laboratory of Microbiology, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Tanja Fusi-Schmidhauser
- Internal Medicine Department, Ospedale Regionale di Lugano, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Giuseppe Vassalli
- Laboratory of Cellular and Molecular Cardiology, Istituto Cardiocentro Ticino, Ente Ospedaliero Cantonale, Lugano, Switzerland; Center for Molecular Cardiology, Zurich, Switzerland; Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland
| | - Giorgia Melli
- Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland; Laboratory for Biomedical Neurosciences, Neurocenter of Southern Switzerland, Lugano, Switzerland
| | - Lucio Barile
- Laboratory for Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Ente Ospedaliero Cantonale Lugano, Switzerland; Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland; Institute of Life Science, Scuola Superiore Sant'Anna, Pisa, Italy.
| |
Collapse
|
45
|
Kendall RT, Renaud L, Baatz JE, Malaab M, Nguyen XX, Feghali-Bostwick CA. Systemic sclerosis biomarkers detection in the secretome of TGFβ1-activated primary human lung fibroblasts. J Proteomics 2021; 242:104243. [PMID: 33930553 DOI: 10.1016/j.jprot.2021.104243] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 02/16/2021] [Accepted: 04/20/2021] [Indexed: 12/17/2022]
Abstract
TGFβ1 is a profibrotic mediator that contributes to a broad spectrum of pathologies, including systemic sclerosis-associated pulmonary fibrosis (SSc-PF). However, the secretome of TGFβ1-stimulated primary human normal lung (NL) fibroblasts has not been well characterized. Using fluorescent 2-dimensional gel electrophoresis (2D-PAGE) and differential gel electrophoresis (DIGE) followed by Mass Spectrometry, we identified 37 differentially secreted proteins in the conditioned media of TGFβ1-activated NL fibroblasts and generated a protein-protein association network of the TGFβ1 secretome using STRING. Functional enrichment revealed that several biological processes and pathways characteristic of PF were enriched. Additionally, by comparing the TGFβ1 secretome of NL fibroblasts to proteomic biomarkers from biological fluids of systemic sclerosis (SSc) patients, we identified 11 overlapping proteins. Together our data validate the TGFβ1-induced secretome of NL fibroblasts as a valid in vitro model that reflects SSc biomarkers and identify potential therapeutic targets for SSc-PF. SIGNIFICANCE: All proteins secreted by fibroblasts into the extracellular space, representing the secretome, promote cell-to-cell communication as well as tissue homeostasis, immune mechanisms, developmental regulation, proteolysis, development of the extracellular matrix (ECM) and cell adhesion. Therefore, it is crucial to understand how TGFβ1, a well-known profibrotic cytokine, modulates the secretome of pulmonary fibroblasts, and how the TGFβ1-induced secretome resembles biomarkers in SSc. Using functional enrichment analysis, key pathways and hub proteins can be identified and studied as potential therapeutic targets for pulmonary fibrosis.
Collapse
Affiliation(s)
- Ryan T Kendall
- Department of Medicine, Rheumatology & Immunology, MUSC, Charleston, SC, United States of America
| | - Ludivine Renaud
- Department of Medicine, Rheumatology & Immunology, MUSC, Charleston, SC, United States of America.
| | - John E Baatz
- Department of Pediatrics, MUSC, Charleston, SC, United States of America.
| | - Maya Malaab
- Department of Medicine, Rheumatology & Immunology, MUSC, Charleston, SC, United States of America.
| | - Xinh-Xinh Nguyen
- Department of Medicine, Rheumatology & Immunology, MUSC, Charleston, SC, United States of America.
| | - Carol A Feghali-Bostwick
- Department of Medicine, Rheumatology & Immunology, MUSC, Charleston, SC, United States of America.
| |
Collapse
|
46
|
Zhang Y, Li Y, Liu P, Gong D, Zhou H, Li W, Zhang H, Zheng W, Xu J, Cheng H, Zhang X, Ke Y. Phosphatase Shp2 regulates biogenesis of small extracellular vesicles by dephosphorylating Syntenin. J Extracell Vesicles 2021; 10:e12078. [PMID: 33732417 PMCID: PMC7944561 DOI: 10.1002/jev2.12078] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 01/22/2021] [Accepted: 02/22/2021] [Indexed: 12/12/2022] Open
Abstract
As novel mediators of cell‐to‐cell signalling, small extracellular vesicles (sEVs) play a critical role in physiological and pathophysiological processes. To date, the molecular mechanisms that support sEV generation are incompletely understood. Many kinases are reported for their roles in sEV generation or composition, whereas the involvement of phosphatases remains largely unexplored. Here we reveal that pharmacological inhibition and shRNA‐mediated down‐regulation of tyrosine phosphatase Shp2 significantly increases the formation of sEVs. By Co‐immunoprecipitation (Co‐IP) and in vitro dephosphorylation assays, we identified that Shp2 negatively controlled sEV biogenesis by directly dephosphorylating tyrosine 46 of Syntenin, which has been reported as a molecular switch in sEV biogenesis. More importantly, Shp2 dysfunction led to enhanced epithelial sEV generation in vitro and in vivo. The increase of epithelial sEVs caused by shRNA‐mediated down‐regulation of Shp2 promoted macrophage activation, resulting in strengthened inflammation. Our findings highlight the role of Shp2 in regulating sEV‐mediated epithelial‐macrophage crosstalk by controlling sEV biogenesis through dephosphorylation of Syntenin Y46. The present study determines the strengthened inflammatory characteristics of alveolar macrophages elicited by epithelial sEVs transferred intercellularly. These findings provide a basis for understanding the mechanism of sEV formation and relevant function in epithelial‐macrophage crosstalk.
Collapse
Affiliation(s)
- Yuefei Zhang
- Department of Pathology and Pathophysiology and Department of Respiratory Medicine at Sir Run Run Shaw Hospital Zhejiang University School of Medicine Hangzhou Zhejiang 310058 China.,Zhejiang Laboratory for Systems and Precision Medicine Zhejiang University Medical Center Hangzhou 311121 China
| | - Yiqing Li
- Department of Pathology and Pathophysiology and Department of Respiratory Medicine at Sir Run Run Shaw Hospital Zhejiang University School of Medicine Hangzhou Zhejiang 310058 China
| | - Pan Liu
- Department of Pathology and Pathophysiology and Department of Respiratory Medicine at Sir Run Run Shaw Hospital Zhejiang University School of Medicine Hangzhou Zhejiang 310058 China
| | - Dacheng Gong
- Department of Pathology and Pathophysiology and Department of Respiratory Medicine at Sir Run Run Shaw Hospital Zhejiang University School of Medicine Hangzhou Zhejiang 310058 China
| | - Hui Zhou
- Department of Pathology and Pathophysiology and Department of Respiratory Medicine at Sir Run Run Shaw Hospital Zhejiang University School of Medicine Hangzhou Zhejiang 310058 China
| | - Wenjuan Li
- Department of Obstetrics and Gynecology Women's hospital Zhejiang University School of Medicine Hangzhou Zhejiang 310058 China
| | - Huilun Zhang
- Department of Pathology and Pathophysiology and Department of Respiratory Medicine at Sir Run Run Shaw Hospital Zhejiang University School of Medicine Hangzhou Zhejiang 310058 China
| | - Wenfang Zheng
- Department of Gastroenterology Sir Run Run Shaw Hospital Zhejiang University School of Medicine Hangzhou Zhejiang 310058 China
| | - Jiaqi Xu
- Department of Pathology Sir Run Run Shaw Hospital Zhejiang University School of Medicine Hangzhou Zhejiang 310058 China
| | - Hongqiang Cheng
- Department of Pathology and Pathophysiology and Department of Cardiology at Sir Run Run Shaw Hospital Zhejiang University School of Medicine Hangzhou Zhejiang 310058 China
| | - Xue Zhang
- Department of Pathology and Pathophysiology and Department of Respiratory Medicine at Sir Run Run Shaw Hospital Zhejiang University School of Medicine Hangzhou Zhejiang 310058 China.,Zhejiang Laboratory for Systems and Precision Medicine Zhejiang University Medical Center Hangzhou 311121 China
| | - Yuehai Ke
- Department of Pathology and Pathophysiology and Department of Respiratory Medicine at Sir Run Run Shaw Hospital Zhejiang University School of Medicine Hangzhou Zhejiang 310058 China.,Zhejiang Laboratory for Systems and Precision Medicine Zhejiang University Medical Center Hangzhou 311121 China
| |
Collapse
|
47
|
Zhu Y, Wang Y, Xing S, Xiong J. Blocking SNHG14 Antagonizes Lipopolysaccharides-Induced Acute Lung Injury via SNHG14/miR-124-3p Axis. J Surg Res 2021; 263:140-150. [PMID: 33652176 DOI: 10.1016/j.jss.2020.10.034] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 09/15/2020] [Accepted: 10/31/2020] [Indexed: 12/30/2022]
Abstract
BACKGROUND Emerging evidence show that long noncoding RNAs (lncRNAs) are crucial regulators in pathophysiology of acute lung injury (ALI). Small nucleolar RNA host gene 14 (SNHG14) is a novel oncogenic lncRNA, and has been associated with inflammation-related cell injuries. Thus, we wondered the role and mechanism of SNHG14 in lipopolysaccharides (LPS)-induced ALI cell model. METHODS Expression of SNHG14, miRNA (miR)-124-3p, and transforming growth factor β type 2 receptor (TGFBR2) was detected by RT-qPCR and western blotting. Cell apoptosis was determined by methyl thiazolyl tetrazolium assay, flow cytometry, western blotting, and lactate dehydrogenase activity kit. Inflammation was measured by enzyme-linked immunosorbent assay. The interaction among SNHG14, miR-124-3p, and TGFBR2 was validated by dual-luciferase reporter assay and RNA immunoprecipitation. RESULTS LPS administration attenuated human lung epithelial cell viability and B-cell lymphoma-2 expression, but augmented apoptosis rate, cleaved-caspase-3 expression, lactate dehydrogenase activity, and secretions of tumor necrosis factor-α, interleukin-1β, and IL-6 in A549 cells. Thus, LPS induced A549 cells apoptosis and inflammation, wherein SNHG14 was upregulated and miR-124-3p was downregulated. However, silencing SNHG14 could suppress LPS-induced apoptosis and inflammation depending on upregulating miR-124-3p via target binding. Similarly, overexpressing miR-124-3p attenuated LPS-induced A549 cells injury through inhibiting its downstream target TGFBR2. Furthermore, SNHG14 knockdown could also affect TGFBR2 expression via miR-124-3p. CONCLUSIONS SNHG14 knockdown prevents A549 cells from LPS-induced apoptosis and inflammation through regulating miR-124-3p and TGFBR2, suggesting a novel SNHG14/miR-124-3p/TGFBR2 circuit in alveolar epithelial cells on the set of ALI.
Collapse
Affiliation(s)
- Yuanbin Zhu
- Department of Respiratory, Linyi Central Hospital, Linyi, Shandong, China
| | - Yingying Wang
- Department of Respiratory, Linyi Central Hospital, Linyi, Shandong, China
| | - Shigang Xing
- Department of Respiratory, Linyi Central Hospital, Linyi, Shandong, China
| | - Jie Xiong
- Department of Respiratory, Linyi Central Hospital, Linyi, Shandong, China.
| |
Collapse
|
48
|
Zhang S, Hong Y, Liu H, Wang Q, Xu J, Zhang Y, Zhao X, Yao Y, Zhou K, Ding X. miR-584 and miR-146 are candidate biomarkers for acute respiratory distress syndrome. Exp Ther Med 2021; 21:445. [PMID: 33747181 DOI: 10.3892/etm.2021.9873] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Accepted: 10/06/2020] [Indexed: 12/11/2022] Open
Abstract
MicroRNAs (miRNAs/miRs) have important roles in inflammation and infections, which are common manifestations of acute respiratory distress syndrome (ARDS). The present study aimed to assess whether serum miRNAs are potential diagnostic biomarkers for human ARDS. For this, two sets of serum samples from healthy individuals and patients with ARDS were analysed by high-throughput sequencing to identify differentially expressed genes in ARDS. A total of 679 valid sequences were identified as differentially expressed (P<0.05). Of these, five differentially expressed miRNAs were subjected to reverse transcription-quantitative PCR validation. Finally, two miRNAs (miR-584 and miR-146a) were successfully verified. These two miRNAs were significantly downregulated in the serum of patients with ARDS. Gene Ontology annotations and Kyoto Encyclopedia of Genes and Genomes pathway analysis revealed that their target transcripts were implicated in a broad range of biological processes and various metabolic pathways, including involvement in the regulation of various inflammatory factors. The present study provided a framework for understanding the molecular mechanisms of ARDS and suggested that miR-584 and miR-146a are associated with ARDS and may be potential therapeutic targets.
Collapse
Affiliation(s)
- Siquan Zhang
- Intensive Care Unit, XiXi Hospital of Hangzhou, Hangzhou, Zhejiang 310023, P.R. China
| | - Yinuo Hong
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, P.R. China
| | - Huafeng Liu
- Intensive Care Unit, XiXi Hospital of Hangzhou, Hangzhou, Zhejiang 310023, P.R. China
| | - Qianpeng Wang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, P.R. China
| | - Juan Xu
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, P.R. China
| | - Yujuan Zhang
- Intensive Care Unit, XiXi Hospital of Hangzhou, Hangzhou, Zhejiang 310023, P.R. China
| | - Xi Zhao
- Intensive Care Unit, XiXi Hospital of Hangzhou, Hangzhou, Zhejiang 310023, P.R. China
| | - Yan Yao
- Intensive Care Unit, XiXi Hospital of Hangzhou, Hangzhou, Zhejiang 310023, P.R. China
| | - Kexing Zhou
- Intensive Care Unit, XiXi Hospital of Hangzhou, Hangzhou, Zhejiang 310023, P.R. China
| | - Xianfeng Ding
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, P.R. China
| |
Collapse
|
49
|
Otsuki Y, Ito E, Mukai A, Ueno M, Yamawaki T, Sotozono C, Kinoshita S, Hamuro J. CD63 + extracellular vesicles from retinal pigment epithelial cells participate in crosstalk with macrophages in the innate inflammatory axis. Exp Eye Res 2021; 205:108496. [PMID: 33610602 DOI: 10.1016/j.exer.2021.108496] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 01/27/2021] [Accepted: 02/08/2021] [Indexed: 12/24/2022]
Abstract
The aim of the study is to clarify the participation of extracellular vesicles (EV) secreted by murine primary retinal pigment epithelial (mpRPE) cells in the cell to cell communication with macrophages (Mps), firstly described by the authors in 2016. In ocular inflammation, Mps act as sources of tumor necrosis factor-α (TNF-α), an activator of RPE cells. TNF-α stimulates the production of monocyte chemotactic protein (MCP-1) by RPE cells, thereby causing greater recruitment of Mps to the sub-RPE space. Murine RAW 264.7 Mps cells were co-cultured with C57BL/6 mouse mpRPE cells, either together or separated in transwells, vertically or horizontally connectable, with 0.40 or 0.03 μm membrane filters. The association of EV with mpRPE or RAW 264.7 was quantified by fluorescence cell sorting (FACS) using Qdot655 streptavidin-conjugated biotinylated EV. Increased levels of CD63+ EV were detected in co-cultures by western blotting or FACS analysis, in accordance with the increased production of nanoparticles (50-150 nm) detected by Nanosight tracking analysis. The gene expressions of cytokines, MCP-1, IL-6, IL-8, and VEGF in mpRPE cells and the corresponding proteins were increased in co-cultures even in transwells, vertically connected with 0.40 μm membrane filters, while the repressed TNF-α protein production was not affected. Most of the CD63+ EVs produced by mpRPE cells in co-cultures were associated with Raw264.7, but not with mpRPE cells. Semi-purified CD63+ EV secreted from mpRPE cells, increased the secretion of MCP-1, IL-6, and VEGF in co-cultures with RAW 264.7. Culture chamber separation horizontally connected with 0.03 μm membrane filters reduced this increased secretion. Collectively, mpRPE derived CD63+ EV partly participate in the sub-retinal innate inflammation. To evaluate the functional damage of RPE cells upon chronic exposure to here defined EVs will be the critical issue to uncover their roles in chronic retinal degenerative diseases.
Collapse
Affiliation(s)
- Yohei Otsuki
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Hirokoji-agaru, Kawaramachi-dori, Kamigyo-ku, Kyoto, 602-0841, Japan
| | - Eiko Ito
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Hirokoji-agaru, Kawaramachi-dori, Kamigyo-ku, Kyoto, 602-0841, Japan
| | - Atsushi Mukai
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Hirokoji-agaru, Kawaramachi-dori, Kamigyo-ku, Kyoto, 602-0841, Japan
| | - Morio Ueno
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Hirokoji-agaru, Kawaramachi-dori, Kamigyo-ku, Kyoto, 602-0841, Japan
| | - Takahiro Yamawaki
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Hirokoji-agaru, Kawaramachi-dori, Kamigyo-ku, Kyoto, 602-0841, Japan
| | - Chie Sotozono
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Hirokoji-agaru, Kawaramachi-dori, Kamigyo-ku, Kyoto, 602-0841, Japan
| | - Shigeru Kinoshita
- Department of Frontier Medical Science and Technology for Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Junji Hamuro
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Hirokoji-agaru, Kawaramachi-dori, Kamigyo-ku, Kyoto, 602-0841, Japan.
| |
Collapse
|
50
|
Abstract
ABSTRACT Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are caused by an exaggerated inflammatory response arising from a wide variety of pulmonary and systemic insults. Lung tissue is composed of a variety of cell populations, including parenchymal and immune cells. Emerging evidence has revealed that multiple cell populations in the lung work in concert to regulate lung inflammation in response to both direct and indirect stimulations. To date, the question of how different types of pulmonary cells communicate with each other and subsequently regulate or modulate inflammatory cascades remains to be fully addressed. In this review, we provide an overview of current advancements in understanding the role of cell-cell interaction in the development of ALI and depict molecular mechanisms by which cell-cell interactions regulate lung inflammation, focusing on inter-cellular activities and signaling pathways that point to possible therapeutic opportunities for ALI/ARDS.
Collapse
Affiliation(s)
- Huiting Zhou
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, China
| | - Erica K. Fan
- Department of Epidemiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania
| | - Jie Fan
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Research and Development, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, Pennsylvania
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|