1
|
Gao L, Zheng F, Fu Z, Wang W. Dual-responsive nanoparticles targeting ACE-II senescence for therapeutic mitigation of acute lung injury. J Nanobiotechnology 2025; 23:339. [PMID: 40340852 PMCID: PMC12060536 DOI: 10.1186/s12951-025-03382-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 04/10/2025] [Indexed: 05/10/2025] Open
Abstract
Acute lung injury (ALI) is a life-threatening condition characterized by severe pulmonary dysfunction, with alveolar type II epithelial cell (ACE-II) senescence playing a pivotal role in its progression. In this study, we developed pH/reactive oxygen species (ROS) dual-responsive nanoparticles (GNPsanti-SP-C) for the targeted delivery of Growth Differentiation Factor 15 (GDF15) to counteract ACE-II senescence. These nanoparticles (NPs) effectively activate the AMP-activated protein kinase (AMPK)/Sirtuin 1 (SIRT1) signaling pathway, inducing the mitochondrial unfolded protein response (UPRmt) and reversing senescence-associated cellular dysfunction. GNPsanti-SP-C were systematically engineered and demonstrated robust pH/ROS sensitivity, efficient GDF15 release, and precise ACE-II targeting. In lipopolysaccharide (LPS)-induced ALI mouse model, GNPsanti-SP-C treatment significantly mitigated lung injury, reduced inflammatory responses, and enhanced pulmonary function, as evidenced by decreased inflammatory markers, lung edema, and improved histopathology. Single-cell transcriptomic and proteomic analyses revealed increased ACE-II cell populations, reduced expression of senescence markers, and upregulation of AMPK/SIRT1 signaling. In vitro studies further demonstrated that UPRmt activation is associated with the NPs' therapeutic effects, suggesting a potential role in their mechanism of action. These findings demonstrate the potential of GDF15-loaded dual-responsive NPs as an innovative strategy to address cellular senescence and alleviate ALI-associated pulmonary damage.
Collapse
Affiliation(s)
- Linlin Gao
- Department of Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Fushuang Zheng
- Department of Thoracic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Zhiling Fu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, Liaoning Province, China.
| | - Wei Wang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, Liaoning Province, China.
| |
Collapse
|
2
|
Luo R, Wang Z, Xu F, Xie K. Dexmedetomidine improve lung inflammation by regulating autophagy and apoptosis of CD4+ T cell via AMPK/mTOR signaling. Mol Immunol 2025; 183:1-11. [PMID: 40311186 DOI: 10.1016/j.molimm.2025.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 04/11/2025] [Accepted: 04/23/2025] [Indexed: 05/03/2025]
Abstract
OBJECTIVES To investigate the protective effect and potential mechanism of dexmedetomidine (Dex) in acute lung injury (ALI). MATERIALS AND METHODS C57BL/6 mice and EL-4 cells were used for in vivo and in vitro studies, respectively. Cecal ligation and puncture (CLP) method was used to prepare an acute lung injury model. After dexmedetomidine intervention, tissue and cell samples were collected to evaluate and measure the severity of lung damage, the proportion of Treg cells, the expression of autophagy-related protein levels and AMPK/mTOR pathways. RESULTS Dex reduced lung damage, and IL-17a, MPO positive cells in the lung, decreased the levels of pro-inflammatory cytokines, and restrain autophagy and apoptosis via the activation of the AMPK/mTOR pathway and increase of the proportion of Tregs. CONCLUSIONS Dex can inhibit the levels of autophagy and apoptosis, increase the proportion of Treg cells, and reduce CLP induced acute lung injury through regulating AKMP/MTOR pathway.
Collapse
Affiliation(s)
- Renjie Luo
- Department of Critical Care Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China; The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, Chongqing, China
| | - Zhao Wang
- Department of Critical Care Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fang Xu
- Department of Critical Care Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Sepsis Diagnosis and Treatment Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Ke Xie
- Department of Critical Care Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Sepsis Diagnosis and Treatment Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
3
|
Wang Y, Wei A, Su Z, Shi Y, Li X, He L. Characterization of lactylation-based phenotypes and molecular biomarkers in sepsis-associated acute respiratory distress syndrome. Sci Rep 2025; 15:13831. [PMID: 40263316 PMCID: PMC12015483 DOI: 10.1038/s41598-025-96969-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 04/01/2025] [Indexed: 04/24/2025] Open
Abstract
Sepsis-associated acute respiratory distress syndrome (ARDS) is a heterogeneous disease with high morbidity and mortality. Lactylation plays a crucial role in sepsis and sepsis-induced lung injury. This study aimed to identify distinct lactylation-based phenotypes in patients with sepsis-associated ARDS and determine relevant molecular biomarkers. We analyzed blood transcriptome and clinical data from patients with sepsis-associated ARDS and calculated the lactylation activity. KEGG pathway analysis, drug sensitivity prediction, and immune cell infiltration analysis were performed. Candidate molecular biomarkers were identified by intersecting the feature genes extracted from four machine learning models. Lactylation activity showed significant heterogeneity among patients with sepsis-associated ARDS, which enabled the classification into low- and high-lactylation activity phenotypes. Patients with high-lactylation experienced longer hospital stays and higher mortality rates, as well as distinct signaling pathways, drug responses, and circulating immune cell abundances. Six key markers (ALDOB, CCT5, EP300, PFKP, PPIA, and SIRT1) were identified to differentiate the two lactylation activity phenotypes, all significantly correlated with circulating immune cell populations. This study revealed significant heterogeneity in lactylation activity phenotypes among patients with sepsis-associated ARDS and identified potential biomarkers to facilitate the application of these phenotypes in clinical practice.
Collapse
Affiliation(s)
- Yiheng Wang
- Department of Anesthesiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, 69 Chuanshan Road, Shigu District, Hengyang, 421001, Hunan Province, China.
| | - An Wei
- Department of Anesthesiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, 69 Chuanshan Road, Shigu District, Hengyang, 421001, Hunan Province, China
| | - Zixuan Su
- Department of Anesthesiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, 69 Chuanshan Road, Shigu District, Hengyang, 421001, Hunan Province, China
| | - Yunyi Shi
- Department of Ophthalmology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan Province, China
| | - Xinqiu Li
- Department of Anesthesiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, 69 Chuanshan Road, Shigu District, Hengyang, 421001, Hunan Province, China
| | - Lixian He
- Department of Ophthalmology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan Province, China.
| |
Collapse
|
4
|
Ma W, Tang S, Yao P, Zhou T, Niu Q, Liu P, Tang S, Chen Y, Gan L, Cao Y. Advances in acute respiratory distress syndrome: focusing on heterogeneity, pathophysiology, and therapeutic strategies. Signal Transduct Target Ther 2025; 10:75. [PMID: 40050633 PMCID: PMC11885678 DOI: 10.1038/s41392-025-02127-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 12/27/2024] [Accepted: 12/27/2024] [Indexed: 03/09/2025] Open
Abstract
In recent years, the incidence of acute respiratory distress syndrome (ARDS) has been gradually increasing. Despite advances in supportive care, ARDS remains a significant cause of morbidity and mortality in critically ill patients. ARDS is characterized by acute hypoxaemic respiratory failure with diffuse pulmonary inflammation and bilateral edema due to excessive alveolocapillary permeability in patients with non-cardiogenic pulmonary diseases. Over the past seven decades, our understanding of the pathology and clinical characteristics of ARDS has evolved significantly, yet it remains an area of active research and discovery. ARDS is highly heterogeneous, including diverse pathological causes, clinical presentations, and treatment responses, presenting a significant challenge for clinicians and researchers. In this review, we comprehensively discuss the latest advancements in ARDS research, focusing on its heterogeneity, pathophysiological mechanisms, and emerging therapeutic approaches, such as cellular therapy, immunotherapy, and targeted therapy. Moreover, we also examine the pathological characteristics of COVID-19-related ARDS and discuss the corresponding therapeutic approaches. In the face of challenges posed by ARDS heterogeneity, recent advancements offer hope for improved patient outcomes. Further research is essential to translate these findings into effective clinical interventions and personalized treatment approaches for ARDS, ultimately leading to better outcomes for patients suffering from ARDS.
Collapse
Affiliation(s)
- Wen Ma
- Department of Emergency Medicine, Institute of Disaster Medicine and Institute of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, China
- Institute for Disaster Management and Reconstruction, Sichuan University-The Hong Kong Polytechnic University, Chengdu, China
| | - Songling Tang
- Department of Emergency Medicine, Institute of Disaster Medicine and Institute of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Peng Yao
- Department of Emergency Medicine, Institute of Disaster Medicine and Institute of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Tingyuan Zhou
- Department of Emergency Medicine, Institute of Disaster Medicine and Institute of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, China
- Institute for Disaster Management and Reconstruction, Sichuan University-The Hong Kong Polytechnic University, Chengdu, China
| | - Qingsheng Niu
- Department of Emergency Medicine, Institute of Disaster Medicine and Institute of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Peng Liu
- Department of Emergency Medicine, Institute of Disaster Medicine and Institute of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Shiyuan Tang
- Department of Emergency Medicine, Institute of Disaster Medicine and Institute of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Yao Chen
- Department of Emergency Medicine, Institute of Disaster Medicine and Institute of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Lu Gan
- Department of Emergency Medicine, Institute of Disaster Medicine and Institute of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, China.
| | - Yu Cao
- Department of Emergency Medicine, Institute of Disaster Medicine and Institute of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, China.
- Institute for Disaster Management and Reconstruction, Sichuan University-The Hong Kong Polytechnic University, Chengdu, China.
| |
Collapse
|
5
|
Byun JE, Lee JW, Choi EJ, Lee J, Yun SH, Park CH, Kim H, Kim MS, Yoon SR, Kim TD, Noh JY, Min SH, Seong HA, Ahn KS, Choi I, Jung H. Therapeutic Effects of TN13 Peptide on Acute Respiratory Distress Syndrome and Sepsis Models In Vivo. J Clin Med 2025; 14:1804. [PMID: 40142612 PMCID: PMC11942723 DOI: 10.3390/jcm14061804] [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: 02/10/2025] [Revised: 03/05/2025] [Accepted: 03/06/2025] [Indexed: 03/28/2025] Open
Abstract
Background/Objectives: Regulation of acute inflammatory responses is crucial for host mortality and morbidity induced by pathogens. The pathogenesis of acute respiratory distress syndrome (ARDS) and sepsis are associated with systemic inflammation. p38 MAPK is a crucial regulator of inflammatory responses and is a potential target for acute inflammatory diseases, including ARDS and sepsis. We investigated the therapeutic effects of the TAT-TN13 peptide (TN13) on severe inflammatory diseases, including ARDS and sepsis, in vivo. Methods: To establish the ARDS model, C57BL/6 mice were intranasally (i.n.) administered lipopolysaccharide (LPS; 5 mg/kg, 40 µL) to induce lung inflammation. As a positive control, dexamethasone (DEX; 0.2 mg/kg) was administered intraperitoneally (i.n.) 1 h post-LPS exposure. In the experimental groups, TN13 was administered intranasally (i.n.) at doses of 2.5 mg or 5 mg/kg at the same time point. In the LPS-induced sepsis model, mice received an intraperitoneal injection of LPS (20 mg/kg) to induce systemic inflammation. TN13 (25 mg/kg, i.p.) was administered 1 h after LPS treatment. Control mice received phosphate-buffered saline (PBS). Lung histopathology, inflammatory cell infiltration, cytokine levels, and survival rates were assessed to evaluate TN13 efficacy. Results: TN13 significantly reduced inflammatory cell recruitment and cytokine production in the lungs, thereby mitigating LPS-induced ARDS. In the sepsis model, TN13 treatment improved survival rates by suppressing inflammatory responses. Mechanistically, TN13 exerted its effects by inhibiting the p38 MAPK/NF-κB signaling pathway. Conclusions: These results collectively suggested that TN13 could be an effective treatment option for severe inflammatory diseases.
Collapse
Affiliation(s)
- Jae-Eun Byun
- Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Yuseong-gu, Daejeon 34141, Republic of Korea; (J.-E.B.)
- Department of Biochemistry, School of Life Sciences, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Jae-Won Lee
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea; (J.-W.L.)
- Department of Biotechnology, University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Eun Ji Choi
- Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Yuseong-gu, Daejeon 34141, Republic of Korea; (J.-E.B.)
- Department of Functional Genomics, Korea University of Science and Technology (UST), Yuseong-gu, Daejeon 34113, Republic of Korea
| | - Juhyun Lee
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea; (J.-W.L.)
| | - Seok Han Yun
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea; (J.-W.L.)
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - Chan Ho Park
- Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Yuseong-gu, Daejeon 34141, Republic of Korea; (J.-E.B.)
- Department of Functional Genomics, Korea University of Science and Technology (UST), Yuseong-gu, Daejeon 34113, Republic of Korea
| | - Hanna Kim
- Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Yuseong-gu, Daejeon 34141, Republic of Korea; (J.-E.B.)
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Mi Sun Kim
- Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Yuseong-gu, Daejeon 34141, Republic of Korea; (J.-E.B.)
| | - Suk Ran Yoon
- Department of Functional Genomics, Korea University of Science and Technology (UST), Yuseong-gu, Daejeon 34113, Republic of Korea
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Tae-Don Kim
- Department of Functional Genomics, Korea University of Science and Technology (UST), Yuseong-gu, Daejeon 34113, Republic of Korea
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Ji-Yoon Noh
- Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Yuseong-gu, Daejeon 34141, Republic of Korea; (J.-E.B.)
- Department of Functional Genomics, Korea University of Science and Technology (UST), Yuseong-gu, Daejeon 34113, Republic of Korea
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Sang-Hyun Min
- Department of Innovative Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Hyun-A. Seong
- Department of Biochemistry, School of Life Sciences, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Kyung-Seop Ahn
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea; (J.-W.L.)
| | - Inpyo Choi
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Yuseong-gu, Daejeon 34141, Republic of Korea
- Ingenium Therapeutics, 1662 Yuseong daero, Daejeon 34054, Republic of Korea
| | - Haiyoung Jung
- Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Yuseong-gu, Daejeon 34141, Republic of Korea; (J.-E.B.)
- Department of Functional Genomics, Korea University of Science and Technology (UST), Yuseong-gu, Daejeon 34113, Republic of Korea
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Yuseong-gu, Daejeon 34141, Republic of Korea
| |
Collapse
|
6
|
Ding X, Shi D, Cai H, Yan Z, Shen G. Intranasal Atomized Dexmedetomidine in Combination With Intranasal Atomized Butorphanol for Dressing Change Sedation and Analgesia in Adult Burn Patients: A Randomized Clinical Trial. J Burn Care Res 2025; 46:341-348. [PMID: 39126665 DOI: 10.1093/jbcr/irae158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Indexed: 08/12/2024]
Abstract
We aimed to evaluate the efficacy of the intranasal atomized dexmedetomidine (IAD) + intranasal atomized butorphanol (IAB) combination therapy on adult patients with burns undergoing dressing changes. Herein, 46 adult patients with burns were enrolled and randomly divided into 2 groups: dexmedetomidine-butorphanol (DB) and saline-butorphanol, treated with atomized dexmedetomidine + butorphanol and saline + butorphanol, respectively. The primary outcomes were the Ramsay Sedation Scale (RSS) and the Visual Analog Scale (VAS) scores. The secondary outcomes were mean blood pressure (MBP), heart rate, respiratory rate (RR), peripheral blood oxygen saturation (SpO2), total butorphanol consumption, and adverse effects. The 2 groups were comparable in age, sex, weight, and total burn surface area. During dressing changes, the DB group exhibited significantly lower RSS levels (P < .05). Besides, the 2 groups showed no significant differences in VAS scores across all measurement time points. Notably, the DB group exhibited decreased MBP at the beginning of the operation (P < .0001), 10 min after (P < .0001), and 20 min after (P = .0205). Heart rate decreased significantly at the beginning (P = .0005) and 10 min after (P = .0088) in the DB group. Furthermore, the 2 groups showed no significant differences in RR and SpO2 levels. In addition, the rescue butorphanol dose was lower in the DB group (P < .001). Finally, dizziness and nausea incidences were significantly lower in the DB group (P < .05). In conclusion, besides its hemodynamic adverse reactions, the IAD + IAB combination therapy exerted a better sedation effect in adult patients with burns than IAB treatment alone.
Collapse
Affiliation(s)
- Xianchao Ding
- Department of Burn and Plastic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, PR China
- Department of Burn and Plastic Surgery, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212001, PR China
| | - Daoming Shi
- Department of Burn and Plastic Surgery, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212001, PR China
| | - Honghua Cai
- Department of Burn and Plastic Surgery, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212001, PR China
| | - Zhixin Yan
- Department of Burn and Plastic Surgery, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212001, PR China
| | - Guoliang Shen
- Department of Burn and Plastic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, PR China
| |
Collapse
|
7
|
Chen Y, Peng S, Liang J, Wei K. SIRT1 in acute lung injury: unraveling its pleiotropic functions and therapeutic development prospects. Mol Cell Biochem 2025; 480:1449-1464. [PMID: 39269678 DOI: 10.1007/s11010-024-05111-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024]
Abstract
Acute lung injury (ALI) is a continuum of lung changes caused by multiple lung injuries, often associated with severe complications and even death. In ALI, macrophages, alveolar epithelial cells and vascular endothelial cells in the lung are damaged to varying degrees and their function is impaired. Research in recent years has focused on the use of SIRT1 for the treatment of ALI. In this paper, we reviewed the role of SIRT1 in ALI in terms of its cellular and molecular mechanism, targeting of SIRT1 by non-coding RNAs and drug components, as well as pointing out the value of SIRT1 for clinical diagnosis and prognosis. Based on the current literature, SIRT1 exhibits diverse functionalities and possesses significant therapeutic potential. Targeting SIRT1 may provide new therapeutic ideas for the treatment of ALI.
Collapse
Affiliation(s)
- Yina Chen
- Medical School, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Shuangyan Peng
- Medical School, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Junjie Liang
- Medical School, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Ke Wei
- Medical School, Hunan University of Chinese Medicine, Changsha, 410208, China.
- Hunan Province Key Laboratory of Integrative Pathogen Biology, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China.
| |
Collapse
|
8
|
Zheng Z, Qiao X, Yin J, Kong J, Han W, Qin J, Meng F, Tian G, Feng X. Advancements in omics technologies: Molecular mechanisms of acute lung injury and acute respiratory distress syndrome (Review). Int J Mol Med 2025; 55:38. [PMID: 39749711 PMCID: PMC11722059 DOI: 10.3892/ijmm.2024.5479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 12/09/2024] [Indexed: 01/04/2025] Open
Abstract
Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) is an inflammatory response arising from lung and systemic injury with diverse causes and associated with high rates of morbidity and mortality. To date, no fully effective pharmacological therapies have been established and the relevant underlying mechanisms warrant elucidation, which may be facilitated by multi‑omics technology. The present review summarizes the application of multi‑omics technology in identifying novel diagnostic markers and therapeutic strategies of ALI/ARDS as well as its pathogenesis.
Collapse
Affiliation(s)
- Zhihuan Zheng
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Xinyu Qiao
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Junhao Yin
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Junjie Kong
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Wanqing Han
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Jing Qin
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Fanda Meng
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Ge Tian
- School of Life Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong 271000, P.R. China
| | - Xiujing Feng
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| |
Collapse
|
9
|
Xiong M, Luo R, Zhang Z, Liu P, Peng Q, Xu F, Guo M. IL-27 regulates macrophage ferroptosis by inhibiting the Nrf2/HO1 signaling pathway in sepsis-induced ARDS. Inflamm Res 2025; 74:39. [PMID: 39945893 DOI: 10.1007/s00011-024-01986-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 10/09/2024] [Accepted: 10/16/2024] [Indexed: 05/09/2025] Open
Abstract
OBJECTIVES Acute respiratory distress syndrome (ARDS) is a clinical syndrome characterized by high morbidity and mortality rates. Sepsis-induced ARDS involves excessive inflammatory responses, which are modulated by macrophages. This study aimed to elucidate the effect of Recombinant Mouse IL-27 Protein on macrophage ferroptosis and polarization, as well as its impact on sepsis-induced ARDS. METHODS A cecal ligation and puncture (CLP)-induced sepsis model was established using wild-type (WT) or IL27R-/- mice. Then, the mice were randomly divided into 4 groups: a control group, a CLP group, an IL-27 + CLP combination group, and an IL-27, CLP, and Oltipraz combination group. RAW 264.7 cells and BMDMs were used to further determine the role and mechanism of IL-27 in vitro. RESULTS In vitro, IL-27 alone did not alter the expression of proteins linked to the ferroptosis pathway or macrophage polarization. Contrastingly, the combination of IL-27 with LPS further amplified LPS-induced alterations in the ferroptosis pathway, thereby promoting macrophage M1 polarization and inhibiting M2 polarization. Additionally, IL-27 + LPS increased ROS levels in macrophages. A sepsis-induced ARDS mouse model was then established via CLP. In vivo, IL-27 exacerbated CLP-induced lung injury in WT mice. Additionally, it decreased the expression levels of ferroptosis-related proteins (Nrf2, HO-1, GPX4) and increased those of Ptgs2 in the lung tissue of septic mice. Besides, GSH and SOD levels in lung tissue were also reduced. Moreover, IL-27 also promoted M1 polarization and inhibited M2 polarization in macrophages. In IL27R-/- mice, the effects of IL-27 were abrogated. Oltipraz inhibited IL-27-induced changes by up-regulating Nrf2 expression. Overall, this present study demonstrated that the combination of IL-27 and LPS-induced macrophage ferroptosis, promoted macrophage M1 polarization, and inhibited M2 polarization by inhibiting the Nrf2/HO-1 pathway. CONCLUSION Oltipraz may alleviate ARDS-related lung injury by up-regulating Nrf2 expression and concurrently inhibiting macrophage ferroptosis.
Collapse
Affiliation(s)
- Meng Xiong
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, Chongqing, 400016, China
| | - Renjie Luo
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Zhijiao Zhang
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Panting Liu
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Qiaozhi Peng
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Fang Xu
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Minkang Guo
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, Chongqing, 400016, China.
| |
Collapse
|
10
|
Cui Y, Tao H, Hu S, Zhang Y, Li H, Wang J, Wu M, Guo J. Effect of Multidimensional Integrated Lung Protection Measures in Elderly Patients With Fragile Lungs or Combined Lung Dysfunction by Regulating AMPK/SIRT1 Pathway. J Cell Mol Med 2025; 29:e70408. [PMID: 39988974 PMCID: PMC11847988 DOI: 10.1111/jcmm.70408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 10/15/2024] [Accepted: 01/24/2025] [Indexed: 02/25/2025] Open
Abstract
Fragile lungs or lung dysfunction can significantly impact a patient's quality of life. Currently, no specific treatment exists to prevent lung dysfunction in elderly patients. The detailed mechanism of fragile lungs or lung dysfunction in elderly patients remains elusive, and this study aimed to clarify it. General data and blood specimens were obtained from patients with fragile lungs or lung dysfunction. The mice were exposed to cigarette smoke using a smoking apparatus to induce fragile lungs or lung dysfunction mice model. Blood samples and lung tissues were collected from all groups for further testing. haematoxylin-eosin (HE) staining, immunofluorescence, Western blot, flow cytometry and quantitative reverse transcriptase PCR (qRT-PCR) were used to elucidate the molecular mechanisms of multidimensional integrated lung protection measures (MILPM) in fragile lungs or lung dysfunction mice by targeting the AMP-activated protein kinase (AMPK)/Sirtuin 1 (SIRT1) pathway. The results indicated that upregulation of the AMPK/SIRT1 signalling pathway accelerates the fragile lungs or lung dysfunction process, whereas downregulation of the AMPK/SIRT1 signalling pathway can prevent it. Similarly, the change of forced vital capacity (FVC), total lung capacity (TLC) levels is associated with the fragile lungs or lung dysfunction process, whereas reducing their levels can serve as a preventative method against fragile lungs or lung dysfunction development. Upregulation of the AMPK/SIRT1 pathway can accelerate the process of fragile lungs or lung dysfunction.
Collapse
Affiliation(s)
- Yinghui Cui
- Department of AnesthesiologyGongli Hospital of Shanghai Pudong New AreaShanghaiChina
| | - Haiyong Tao
- Department of AnesthesiologyGongli Hospital of Shanghai Pudong New AreaShanghaiChina
| | - Shejun Hu
- Department of AnesthesiologyGongli Hospital of Shanghai Pudong New AreaShanghaiChina
| | - Yan Zhang
- Department of AnesthesiologyGongli Hospital of Shanghai Pudong New AreaShanghaiChina
| | - Hao Li
- Department of AnesthesiologyGongli Hospital of Shanghai Pudong New AreaShanghaiChina
| | - Jinhuo Wang
- Department of AnesthesiologyGongli Hospital of Shanghai Pudong New AreaShanghaiChina
| | - Mandi Wu
- Department of AnesthesiologyGongli Hospital of Shanghai Pudong New AreaShanghaiChina
| | - Jianrong Guo
- Department of AnesthesiologyGongli Hospital of Shanghai Pudong New AreaShanghaiChina
| |
Collapse
|
11
|
Zhao YS, Shi YK, Li KF, Ma B, Lin SH, Xing Y, Xu F. Dexmedetomidine Regulates Macrophage Phenotype Remodeling Through AMPK/SIRT1 to Alleviate Inflammatory Mediators and Lung Injury. J Biochem Mol Toxicol 2025; 39:e70108. [PMID: 39692360 DOI: 10.1002/jbt.70108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 11/22/2024] [Accepted: 12/07/2024] [Indexed: 12/19/2024]
Abstract
Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) is associated with high morbidity and mortality in the intensive care unit (ICU) and can cause excessive inflammation. Dexmedetomidine (DEX) is a drug that exerts anti-inflammatory effects. Identifying the anti-inflammatory mechanism of DEX in the context of ALI/ARDS possesses potential significance for the prevention and treatment of ARDS. In this study, DEX was used to treat mouse models of cecal ligation and puncture (CLP) and lipopolysaccharide (LPS)-stimulated cells. Immunofluorescence, western blot analysis, and flow cytometry were used to detect macrophage phenotypic markers in mice, and western blot analysis, real-time qPCR (RT-qPCR), ELISA, and immunofluorescence were used to detect macrophage phenotype markers in RAW264.7 cells. Flow cytometry was used to detect phenotypic markers of bone marrow-derived macrophages (BMDM). Culture medium collected from macrophages was used to cultivate human non-small cell adenocarcinoma epithelial cells (A549) to detect their aquaporins 1 (AQP1) expression and apoptosis status. Western blot analysis was used to detect the activation of the AMP-activated protein kinase (AMPK)/sirtuin 1(SIRT1) signaling pathway both in vivo and in vitro. The regulatory effect of DEX on macrophage phenotype remodeling was detected by knocking down AMPK expression in cells using AMPK shRNA. The results showed that in both in vivo and in vitro experiments, DEX downregulated the expression of M1 markers (tumor necrosis factor-α [TNF-α], nitric oxide synthase [iNOS], and cluster of differentiation [CD]-86) and upregulated the expression of M2 markers (arginase-1 [ARG-1], interleukin [IL]-10, and CD206) in macrophages. The culture medium of macrophages treated with DEX alleviated the edema and apoptosis of A549 cells. DEX activates the AMPK/SIRT1 signaling pathway in macrophages. After AMPK knockdown, the ability of DEX to regulate macrophage phenotype remodeling decreased. Together, this study suggests that DEX regulates macrophage phenotype remodeling by activating the AMPK/SIRT1 pathway, thereby reducing ALI/ARDS.
Collapse
Affiliation(s)
- Yi-Si Zhao
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ya-Kang Shi
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Forensic Medicine and Biomedical Informatics Research Room, Chongqing Medical University, Chongqing, China
| | - Ke-Feng Li
- Department of Critical Care Medicine, Fengjie Hospital, A Branch of The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Bei Ma
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Critical Care Medicine, The First People's Hospital of Chongqing Liang Jiang New Area, Chongqing, China
| | - Shi-Hui Lin
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yu Xing
- Forensic Medicine and Biomedical Informatics Research Room, Chongqing Medical University, Chongqing, China
| | - Fang Xu
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
12
|
Zhu X, Meng L, Xu L, Hua Y, Feng J. Novel Therapeutic Target for ALI/ARDS: Forkhead Box Transcription Factors. Lung 2024; 202:513-522. [PMID: 39259274 DOI: 10.1007/s00408-024-00740-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 08/17/2024] [Indexed: 09/12/2024]
Abstract
ALI/ARDS can be a pulmonary manifestation of a systemic inflammatory response or a result of overexpression of the body's normal inflammatory response involving various effector cells, cytokines, and inflammatory mediators, which regulate the body's immune response through different signalling pathways. Forkhead box transcription factors are evolutionarily conserved transcription factors that play a crucial role in various cellular processes, such as cell cycle progression, proliferation, differentiation, migration, metabolism, and DNA damage response. Transcription factors control protein synthesis by regulating gene transcription levels, resulting in diverse biological outcomes. The Fox family plays a role in activating or inhibiting the expression of various molecules related to ALI/ARDS through phosphorylation, acetylation/deacetylation, and control of multiple signalling pathways. An in-depth analysis of the integrated Fox family's role in ALI/ARDS can aid in the development of potential diagnostic and therapeutic targets for the condition.
Collapse
Affiliation(s)
- Xi Zhu
- Department of Respiratory and Critical Care Medicine, Respiratory Disease Key Laboratory of Nantong, Affiliated Hospital of Nantong University, 20 Xi-Si Road, Nantong, 226001, Jiangsu, China
| | - Leyuan Meng
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital and Medical School of Nantong University, Nantong, 226001, Jiangsu, China
| | - Liqin Xu
- Department of Respiratory and Critical Care Medicine, Respiratory Disease Key Laboratory of Nantong, Affiliated Hospital of Nantong University, 20 Xi-Si Road, Nantong, 226001, Jiangsu, China
| | - Yun Hua
- Department of Respiratory and Critical Care Medicine, Respiratory Disease Key Laboratory of Nantong, Affiliated Hospital of Nantong University, 20 Xi-Si Road, Nantong, 226001, Jiangsu, China
| | - Jian Feng
- Department of Respiratory and Critical Care Medicine, Respiratory Disease Key Laboratory of Nantong, Affiliated Hospital of Nantong University, 20 Xi-Si Road, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
13
|
Huang C. Toll-like receptor 4 (TLR4) deficiency impedes Toxoplasma gondii excreted-secreted antigens (ESA)-induced abortion. Placenta 2024; 154:1-8. [PMID: 38824786 DOI: 10.1016/j.placenta.2024.05.137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/14/2024] [Accepted: 05/28/2024] [Indexed: 06/04/2024]
Abstract
INTRODUCTION Toxoplasma gondii is an opportunistic intracellular parasite that is a major pathogenic factor in miscarriage, especially when it occurs early in pregnancy. We have previously demonstrated that the regulation of forkhead box transcription factor (Foxp3) is associated with abortion in early pregnancy caused by excretory-secretory antigen (ESA) of strain China 1. We aimed to reveal the underlying mechanism of miscarriage caused by ESA. METHODS A TLR4-/- pregnant mouse model was successfully constructed. Pregnant mice at gestational day 5 (G5) were injected with ESA. All animals were sacrificed on G13, pregnancy outcomes were observed, and abortion rates were calculated. Placental status observed by Hematoxylin-eosin staining; gene expression was measured by IHC; flow cytometry analysis was used to determine the number and function of regulatory T cells. In EL4 cells, real-time PCR and Western blot were used to evaluate gene expression and cytokines assay. RESULTS In vivo studies revealed that ESA injection caused 83% abortion in pregnant mice but only 35% abortion in TLR4-/- pregnant mice. In addition, ESA attenuated the number and function of regulatory T cells, further suppressed Foxp3, FOXO1 levels, and upregulated CD127 expression. TLR4-/- mice partially reversed this inhibitory effect on regulatory T cells. Furthermore, in vitro studies revealed that ESA inhibited TLR4/NF-κB signaling pathway expression and that TLR4 agonists significantly restored the ESA-induced decrease in Foxp3. DISCUSSION These findings suggest that ESA suppresses Foxp3 expression by blocking TLR4/NF-κB signaling, resulting in miscarriage. More importantly, the results indicated that miscarriage caused by ESA is TLR4 dependent.
Collapse
Affiliation(s)
- Caiqun Huang
- Departments of Central Laboratory, Jinhua Municipal Central Hospital, Jinhua, Zhejiang, 321000, PR China.
| |
Collapse
|
14
|
Chen Z, Zuo Z, Song X, Zuo Y, Zhang L, Ye Y, Ma Y, Pan L, Zhao X, Jin Y. Mapping Theme Trends and Research Frontiers in Dexmedetomidine Over Past Decade: A Bibliometric Analysis. Drug Des Devel Ther 2024; 18:3043-3061. [PMID: 39050803 PMCID: PMC11268573 DOI: 10.2147/dddt.s459431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Accepted: 07/04/2024] [Indexed: 07/27/2024] Open
Abstract
Background Dexmedetomidine, an α2-adrenergic receptor (α2-AR) agonist, is extensively used in clinical and animal studies owing to its sedative, analgesic, and anxiolytic effects. The diverse range of research domains associated with dexmedetomidine poses challenges in defining pivotal research directions. Therefore, this study aimed to conduct a qualitative and quantitative bibliometric study in the field of dexmedetomidine over the past decade to establish current research trends and emerging frontiers. Methods Relevant publications in the field of dexmedetomidine between 2014 and 2023 were extracted from the Web of Science Core Collection database. The bibliometric analysis, incorporating statistical and visual analyses, was conducted using CiteSpace (6.1.R6) and R (4.3.1). Results The present study encompassed a total of 5,482 publications, exhibiting a consistent upward trend over the past decade. The United States and its institutions had the highest centrality. Ji, Fuhai, and Ebert, Thomas J. were identified as the most productive author and the most cited author, respectively. As anticipated, the most cited journal was Anesthesiology. Moreover, cluster analysis of cited references and co-occurrence of keywords revealed that recent studies were primarily focused on sedation, delirium, and opioid-free anesthesia. Finally, a timeline view of keywords clusters and keywords burst demonstrated that primary research frontiers were stress response, neuroinflammation, delirium, opioid-free anesthesia, peripheral nerve block, and complications. Conclusion Current research trends and directions are focused on sedation, delirium, and opioid-free anesthesia, as evidenced by our results. The frontier of future research is anticipated to encompass basic investigations into dexmedetomidine, including stress response and neuroinflammation, as well as clinical studies focusing on delirium, opioid-free anesthesia, peripheral nerve block, and associated complications.
Collapse
Affiliation(s)
- Zheping Chen
- Department of Anesthesiology, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250033, People’s Republic of China
| | - Zhenxiang Zuo
- Department of Gastroenterology, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250033, People’s Republic of China
| | - Xinyu Song
- Department of Anesthesiology, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250033, People’s Republic of China
| | - Yaqun Zuo
- Department of Anesthesiology, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250033, People’s Republic of China
| | - Le Zhang
- Department of Anesthesiology, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250033, People’s Republic of China
| | - Yuyang Ye
- Department of Anesthesiology, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250033, People’s Republic of China
| | - Yufeng Ma
- Department of Anesthesiology, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250033, People’s Republic of China
| | - Lili Pan
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People’s Republic of China
| | - Xin Zhao
- Department of Anesthesiology, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250033, People’s Republic of China
| | - Yanwu Jin
- Department of Anesthesiology, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250033, People’s Republic of China
| |
Collapse
|
15
|
Giri BR, Li S, Cheng G. Exogenous modification of EL-4 T cell extracellular vesicles with miR-155 induce macrophage into M1-type polarization. Drug Deliv Transl Res 2024; 14:934-944. [PMID: 37817019 DOI: 10.1007/s13346-023-01442-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2023] [Indexed: 10/12/2023]
Abstract
Extracellular vesicles (EVs) show promising potential to be used as therapeutics, disease biomarkers, and drug delivery vehicles. We aimed to modify EVs with miR-155 to modulate macrophage immune response that can be potentially used against infectious diseases. Primarily, we characterized T cells (EL-4) EVs by several standardized techniques and confirmed that the EVs could be used for experimental approaches. The bioactivities of the isolated EVs were confirmed by the uptake assessment, and the results showed that target cells can successfully uptake EVs. To standardize the loading protocol by electroporation for effective biological functionality, we chose fluorescently labelled miR-155 mimics because of its important roles in the immune regulations to upload them into EVs. The loading procedure showed that the dosage of 1 µg of miRNA mimics can be efficiently loaded to the EVs at 100 V, further confirmed by flow cytometry. The functional assay by incubating these modified EVs (mEVs) with in vitro cultured cells led to an increased abundance of miR-155 and decreased the expressions of its target genes such as TSHZ3, Jarid2, ZFP652, and WWC1. Further evaluation indicated that these mEVs induced M1-type macrophage polarization with increased TNF-α, IL-6, IL-1β, and iNOS expression. The bioavailability analysis revealed that mEVs could be detected in tissues of the livers. Overall, our study demonstrated that EVs can be engineered with miR-155 of interest to modulate the immune response that may have implications against infectious diseases.
Collapse
Affiliation(s)
- Bikash R Giri
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, 500 Zhen-Nan Road, Shanghai, 200311, China
- Department of Zoology, Utkal University, Bhubaneswar, 751004, India
| | - Shun Li
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai, 200241, China
- School of Life Sciences and Engineering, Foshan University, Foshan, Guangdong, 528225, China
| | - Guofeng Cheng
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, 500 Zhen-Nan Road, Shanghai, 200311, China.
| |
Collapse
|
16
|
Chen Y, Guo M, Xie K, Lei M, Chai Y, Zhang Z, Deng Z, Peng Q, Cao J, Lin S, Xu F. Progranulin promotes regulatory T cells plasticity by mitochondrial metabolism through AMPK/PGC-1α pathway in ARDS. Clin Immunol 2024; 261:109940. [PMID: 38365048 DOI: 10.1016/j.clim.2024.109940] [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: 11/01/2023] [Revised: 02/01/2024] [Accepted: 02/10/2024] [Indexed: 02/18/2024]
Abstract
As the aging population increases, the focus on elderly patients with acute respiratory distress syndrome (ARDS) is also increasing. In this article, we found progranulin (PGRN) differential expression in ARDS patients and healthy controls, even in young and old ARDS patients. Its expression strongly correlates with several cytokines in both young and elderly ARDS patients. PGRN has comparable therapeutic effects in young and elderly mice with lipopolysaccharide-induced acute lung injury, manifesting as lung injury, apoptosis, inflammation, and regulatory T cells (Tregs) differentiation. Considering that Tregs differentiation relies on metabolic reprogramming, we discovered that Tregs differentiation was mediated by mitochondrial function, especially in the aged population. Furthermore, we demonstrated that PGRN alleviated the mitochondrial damage during Tregs differentiation through the AMPK/PGC-1α pathway in T cells. Collectively, PGRN may regulate mitochondria function to promote Tregs differentiation through the AMPK/PGC-1α pathway to improve ARDS.
Collapse
Affiliation(s)
- Yanqing Chen
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Minkang Guo
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ke Xie
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ming Lei
- Department of Critical Care Medicine, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yusen Chai
- Department of Anaesthesiology and Intensive Care Medicine, Pulmonary Engineering Group, University Hospital Carl Gustav Carus Dresden at Technische Universität Dresden, Dresden, Germany
| | - Zhengtao Zhang
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhenhua Deng
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qiaozhi Peng
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ju Cao
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shihui Lin
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Fang Xu
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
17
|
Ning L, Shishi Z, Bo W, Huiqing L. Targeting immunometabolism against acute lung injury. Clin Immunol 2023; 249:109289. [PMID: 36918041 PMCID: PMC10008193 DOI: 10.1016/j.clim.2023.109289] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 03/06/2023] [Accepted: 03/08/2023] [Indexed: 03/14/2023]
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are life-threatening conditions triggered by multiple intra- and extra-pulmonary injury factors, characterized by complicated molecular mechanisms and high mortality. Great strides have been made in the field of immunometabolism to clarify the interplay between intracellular metabolism and immune function in the past few years. Emerging evidence unveils the crucial roles of immunometabolism in inflammatory response and ALI. During ALI, both macrophages and lymphocytes undergo robust metabolic reprogramming and discrete epigenetic changes after activated. Apart from providing ATP and biosynthetic precursors, these metabolic cellular reactions and processes in lung also regulate inflammation and immunity.In fact, metabolic reprogramming involving glucose metabolism and fatty acidoxidation (FAO) acts as a double-edged sword in inflammatory response, which not only drives inflammasome activation but also elicits anti-inflammatory response. Additionally, the features and roles of metabolic reprogramming in different immune cells are not exactly the same. Here, we outline the evidence implicating how adverse factors shape immunometabolism in differentiation types of immune cells during ALI and summarize key proteins associated with energy expenditure and metabolic reprogramming. Finally, novel therapeutic targets in metabolic intermediates and enzymes together with current challenges in immunometabolism against ALI were discussed.
Collapse
Affiliation(s)
- Li Ning
- Department of Thoracic Surgery, Renmin Hospital, Wuhan University, Wuhan, Hubei Province, China
| | - Zou Shishi
- Department of Thoracic Surgery, Renmin Hospital, Wuhan University, Wuhan, Hubei Province, China
| | - Wang Bo
- Department of Thoracic Surgery, Renmin Hospital, Wuhan University, Wuhan, Hubei Province, China.
| | - Lin Huiqing
- Department of Thoracic Surgery, Renmin Hospital, Wuhan University, Wuhan, Hubei Province, China.
| |
Collapse
|