1
|
Yang X, Xiao X, Zhou L, Shen Y, Wang L, Shen Q. Involvement of PRDX6 in the protective role of MANF in acute lung injury in rats. Exp Lung Res 2025; 51:1-10. [PMID: 39861940 DOI: 10.1080/01902148.2025.2454032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 01/07/2025] [Accepted: 01/09/2025] [Indexed: 01/27/2025]
Abstract
Aim/Purpose of the study: Acute lung injury (ALI) is a severe respiratory disease with high mortality, mainly due to overactivated oxidative stress and subsequent pyroptosis. Mesencephalic astrocyte-derived neurotrophic factor (MANF), an inducible secretory endoplasmic reticulum (ER) stress protein, inhibits lipopolysaccharide (LPS)-induced acute lung injury (ALI). However, the exact molecular mechanism remains unclear. Peroxiredoxin 6 (PRDX6), a peroxidase with a dual enzymatic function, is essential in regulating oxidative stress, which is closely associated with ALI. Furthermore, PRDX6 is an interacting protein of MANF. Therefore, this study aims to investigate the role of PRDX6 in the protective effect of MANF on ALI. Materials and Methods: In this study, we used LPS to establish the LPS-induced ALI model. Recombinant human MANF was administrated to wide-type (WT) and PRDX6 knockout (PRDX6-/-) rats. Results: In WT rats, MANF reversed the increases of PRDX6, ROS overgeneration, and pyroptosis-related protein-Gasdermin D (GSDMD) induced by LPS challenge. In PRDX6-/- rats, ROS generation, the protein level of GSDMD-N, and lung injury were not significantly decreased after human recombinant MANF administration in LPS-induced ALI. Conclusions: PRDX6 is involved in the protective role of MANF on ALI. It is a key target molecule for MANF to exert ALI inhibitory effects.
Collapse
Affiliation(s)
- Xiuli Yang
- Department of Anesthesiology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Xueying Xiao
- Department of Anesthesiology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Public Health Clinical Center, Hefei, China
| | - Leiying Zhou
- Department of Anesthesiology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Yujun Shen
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei, China
| | - Lixia Wang
- Department of Anesthesiology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Qiying Shen
- Department of Anesthesiology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| |
Collapse
|
2
|
Misar PL, Otari KV. Investigation of the protective effect of cilostazol on acute lung injury-mediated inflammation and in silico molecular modelling studies of inflammatory signalling pathway: a repurposing study. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-024-03734-z. [PMID: 39754679 DOI: 10.1007/s00210-024-03734-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 12/13/2024] [Indexed: 01/06/2025]
Abstract
Acute lung injury i.e. ALI and its serious form acute respiratory distress syndrome (ARDS) are incurable medical conditions associated with significant global mortality and morbidity. The objective of the present research was to repurpose cilostazol, an antiplatelet drug with anti-inflammatory, antioxidant and antiapoptotic effect, as a potential approach for treatment of ALI. Its multifaceted effects make it promising candidate but its mechanism against ALI remains elusive. Hence it is needed to elucidate its mechanism of action to revealed its therapeutic potential and improve its clinical outcomes. This study investigated the potential inflammatory therapeutic targets of cilostazol with its protective effect against lipopolysaccharide (LPS)-induced ALI. We have identified 10 inflammatory target proteins of cilostazol i.e. PDK1, RAC1, PTK6, KDR, EGFR, endothelin-I, caspase-3, TNF-α, NF-κB1/BTK, a TLR/IRAK4 by molecular docking and validated by in vivo evaluation to demonstrate its therapeutic efficacy. In vivo experiment was performed in two sets; first to determine cellular inflammation by analysing the biomarkers in both lung homogenate and bronchoalveolar fluid and second set to study lung edema with dexamethasone as a standard. Additionally, respiratory parameters, related mRNA expressions and histopathology was evaluated. Our results, molecular docking showed that cilostazol binds to identified inflammatory target proteins with the same binding affinity as that of experimental inhibitors. In vivo, downregulated oxidative stress, and inflammation i.e. attenuated the pulmonary edema and vascular leakage, release of inflammatory mediators i.e. IL-6, TNF-α, NO, C-reactive protein (CRP), lactate dehydrogenase (LDH) myeloperoxidase (MPO), Krebs von den Lungen 6 (KL-6), and the recruitment of inflammatory cells; downregulated the m-RNA gene expressions of tumour necrosis factor alpha (TNF-α), nuclear factor kappa B( NF-kB), Toll-like receptor 4 (TLR4), Janus kinase/signal transducer, and activator of transcription 3 (JAK and STAT3); and improved total lung capacity in LPS-challenged rats. These findings revealed the cilostazol's efficacy as promising therapeutic agent for ALI by inhibiting the NF-κB/TLR4/JAK-STAT3 signalling pathway.
Collapse
Affiliation(s)
- Pranaya L Misar
- Dr. Babasaheb Ambedkar Technological University, Lonere, Raigad, 402103, India.
- N.N. Sattha College of Pharmacy, Ahmednagar, Maharashtra, 414003, India.
| | - Kishor V Otari
- Dr. Babasaheb Ambedkar Technological University, Lonere, Raigad, 402103, India
- Navsahyadri College of Pharmacy, Nasrapur, Pune, Maharashtra, 412213, India
| |
Collapse
|
3
|
Xia S, Gu X, Wang G, Zhong Y, Ma F, Liu Q, Xie J. Regulated Cell Death of Alveolar Macrophages in Acute Lung Inflammation: Current Knowledge and Perspectives. J Inflamm Res 2024; 17:11419-11436. [PMID: 39722732 PMCID: PMC11669335 DOI: 10.2147/jir.s497775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 11/29/2024] [Indexed: 12/28/2024] Open
Abstract
Acute lung injury/acute respiratory distress syndrome (ALI/ARDS) is a common and serious clinical lung disease characterized by extensive alveolar damage and inflammation leading to impaired gas exchange. Alveolar macrophages (AMs) maintain homeostatic properties and immune defenses in lung tissues. Several studies have reported that AMs are involved in and regulate ALI/ARDS onset and progression via different regulated cell death (RCD) programs, such as pyroptosis, apoptosis, autophagic cell death, and necroptosis. Notably, the effects of RCD in AMs in disease are complex and variable depending on the environment and stimuli. In this review, we provide a comprehensive perspective on how regulated AMs death impacts on ALI/ARDS and assess its potential in new therapeutic development. Additionally, we describe the crosstalk between different RCD types in ALI, and provide new perspectives for the treatment of ALI/ARDS and other severe lung diseases.
Collapse
Affiliation(s)
- Siwei Xia
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Xiaoyan Gu
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Gaojian Wang
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Yizhi Zhong
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Fengjie Ma
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Qinxue Liu
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Junran Xie
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| |
Collapse
|
4
|
Zeng W, Deng Z, Li H, Gao S, Ju R. Purinergic P2X7 receptor mediates hyperoxia-induced injury in pulmonary microvascular endothelial cells via NLRP3-mediated pyroptotic pathway. Open Med (Wars) 2024; 19:20241097. [PMID: 39655049 PMCID: PMC11627065 DOI: 10.1515/med-2024-1097] [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: 04/19/2024] [Revised: 10/29/2024] [Accepted: 11/04/2024] [Indexed: 12/12/2024] Open
Abstract
Background Hyperoxia-induced injury is a well-recognized cause of bronchopulmonary dysplasia (BPD). Existing research studies have not well elucidated the exact mechanisms underlying hyperoxia-induced cellular damage. This study examines the involvement of the P2X7 receptor (P2X7R) in hyperoxia-induced damage to human pulmonary microvascular endothelial cells (HPMVECs) via the NOD-like receptor family, pyrin domain-containing protein 3 (NLRP3) pathway. Methods HPMVECs developing hyperoxia-induced injury were subjected to the treatment of either selective inhibitors or a P2X7R/NLRP3 agonist. Western blot analysis assisted in the quantification of the levels of P2X7R, NLRP3, caspase-1, and gasdermin D (GSDMD). Additionally, the release of TNF-α, IL-1β, and IL-18 was assessed by ELISA and qRT-PCR. Results Exposure to hyperoxia diminished cell viability and escalated the levels of P2X7R, caspase-1, NLRP3, GSDMD, and N-terminal-GSDMD. This exposure notably increased the release of TNF-α, IL-1β, and IL-18 in HPMVECs. Notably, the suppression of P2X7R using the inhibitor A438079 decreased pyroptosis and inflammatory responses. Conversely, stimulation of P2X7R by 3'-O-(4-benzoylbenzoyl) adenosine 5'-triphosphate (BzATP) triggered pyroptosis, while inhibition of NLRP3 with glibenclamide ameliorated the damage induced by BzATP. Conclusions The P2X7R/NLRP3 pathway crucially affects the hyperoxia-induced inflammation and pyroptosis in HPMVECs, hinting the potential of blocking P2X7R/NLRP3-mediated pyroptotic pathway as a valuable therapeutic strategy for BPD.
Collapse
Affiliation(s)
- Wen Zeng
- Department of Neonatology, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, Sichuan, China
| | - Zhuyu Deng
- Department of Neonatology, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, Sichuan, China
| | - Huaying Li
- Department of Neonatology, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, Sichuan, China
| | - Shuqiang Gao
- Department of Neonatology, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, Sichuan, China
| | - Rong Ju
- Department of Neonatology, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, Sichuan, China
| |
Collapse
|
5
|
Luo P, Gu Q, Wang J, Li X, Li N, Yang W, Meng X, Zhao M. SS31 alleviates LPS-induced acute lung injury by inhibiting inflammatory responses through the S100A8/NLRP3/GSDMD signaling pathway. Eur J Med Res 2024; 29:567. [PMID: 39609864 PMCID: PMC11605876 DOI: 10.1186/s40001-024-02169-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 11/21/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUND Acute lung injury/acute respiratory distress syndrome (ALI/ARDS) is an acute, diffuse, inflammatory lung injury caused by various endogenous or exogenous factors. It is currently widely recognized that an excessive inflammatory response resulting from immune imbalance constitutes a crucial pathogenic mechanism in ALI/ARDS. SS31 is a novel mitochondria-targeted antioxidant peptide. This article validates the role of SS31 in lipopolysaccharide (LPS)-induced ALI. METHODS The study applied transcriptome sequencing, immunofluorescence, PCR, immunofluorescence and other methods to explore the mechanism of SS31 in LPS induced ALI. RESULTS Transcriptome sequencing results indicate that LPS-induced ALI is closely associated with immune regulatory processes, the Toll-like receptor pathway, and the NF-κB signaling pathway. The role of SS31 in acute lung injury is closely related to biological processes, such as immune regulation and cell death. This study demonstrated that SS31 can inhibit the expression of inflammatory factors IL-6, IL-1β, IL-18, and TNF-α, and reduce the expression of pyroptosis-related proteins NLRP3, and GSDMD-N. Further analysis revealed that S100A8 may be a key gene in the effect of SS31. LPS stimulation leads to increased expression of S100A8, while SS31 decreases its expression. Recombinant protein S100A8 can attenuate the inhibitory effect of SS31 on IL-1β, IL-18, NLRP3, and GSDMD-N. CONCLUSIONS The research results indicate that SS31 may inhibit the activation of the NLRP3 inflammasome and suppress inflammatory responses by regulating S100A8, thereby alleviating LPS-induced ALI in mice; this process may be related to pyroptosis.
Collapse
Affiliation(s)
- Peiyao Luo
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, No. 2075, Qunli Seventh Avenue, Daoli District, Harbin, 150001, Heilongjiang Province, China
| | - Quankuan Gu
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, No. 2075, Qunli Seventh Avenue, Daoli District, Harbin, 150001, Heilongjiang Province, China
| | - Jianpeng Wang
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, No. 2075, Qunli Seventh Avenue, Daoli District, Harbin, 150001, Heilongjiang Province, China
| | - Xianyong Li
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, No. 2075, Qunli Seventh Avenue, Daoli District, Harbin, 150001, Heilongjiang Province, China
| | - Nana Li
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, No. 2075, Qunli Seventh Avenue, Daoli District, Harbin, 150001, Heilongjiang Province, China
| | - Wei Yang
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China.
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, No. 2075, Qunli Seventh Avenue, Daoli District, Harbin, 150001, Heilongjiang Province, China.
| | - Xianglin Meng
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China.
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, No. 2075, Qunli Seventh Avenue, Daoli District, Harbin, 150001, Heilongjiang Province, China.
| | - Mingyan Zhao
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China.
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, No. 2075, Qunli Seventh Avenue, Daoli District, Harbin, 150001, Heilongjiang Province, China.
| |
Collapse
|
6
|
Lu F, Wang R, Cheng Y, Li X. Preconditioning with β-hydroxybutyrate attenuates lung ischemia-reperfusion injury by suppressing alveolar macrophage pyroptosis through the SIRT1-FOXO3 signaling pathway. FASEB J 2024; 38:e70027. [PMID: 39221615 DOI: 10.1096/fj.202401188r] [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: 05/28/2024] [Revised: 07/23/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
The complex pathogenesis of lung ischemia-reperfusion injury (LIRI) was examined in a murine model, focusing on the role of pyroptosis and its exacerbation of lung injury. We specifically examined the levels and cellular localization of pyroptosis within the lung, which revealed alveolar macrophages as the primary site. The inhibition of pyroptosis by VX-765 reduced the severity of lung injury, underscoring its significant role in LIRI. Furthermore, the therapeutic potential of β-hydroxybutyrate (β-OHB) in ameliorating LIRI was examined. Modulation of β-OHB levels was evaluated by ketone ester supplementation and 3-hydroxybutyrate dehydrogenase 1 (BDH-1) gene knockout, along with the manipulation of the SIRT1-FOXO3 signaling pathway using EX-527 and pCMV-SIRT1 plasmid transfection. This revealed that β-OHB exerts lung-protective and anti-pyroptotic effects, which were mediated through the upregulation of SIRT1 and the enhancement of FOXO3 deacetylation, leading to decreased pyroptosis markers and lung injury. In addition, β-OHB treatment of MH-S cells in vitro showed a concentration-dependent improvement in pyroptosis, linking its therapeutic benefits to specific cell mechanisms. Overall, this study highlights the significance of alveolar macrophage pyroptosis in the exacerbation of LIRI and indicates the potential of β-OHB in mitigating injury by modulating the SIRT1-FOXO3 signaling pathway.
Collapse
Affiliation(s)
- Fan Lu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
- Department of Pain Management, West China Hospital, Sichuan University, Chengdu, China
| | - Rurong Wang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Yan Cheng
- Laboratory of Anesthesia & Critical Care Medicine, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - XueHan Li
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
7
|
Liu B, Li N, Liu Y, Zhang Y, Qu L, Cai H, Li Y, Wu X, Geng Q. BRD3308 suppresses macrophage oxidative stress and pyroptosis via upregulating acetylation of H3K27 in sepsis-induced acute lung injury. BURNS & TRAUMA 2024; 12:tkae033. [PMID: 39224841 PMCID: PMC11367671 DOI: 10.1093/burnst/tkae033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 04/04/2024] [Indexed: 09/04/2024]
Abstract
Background Sepsis-induced acute lung injury (ALI) leads to severe hypoxemia and respiratory failure, contributing to poor prognosis in septic patients. Endotoxin dissemination triggers oxidative stress and the release of inflammatory cytokines in macrophages, initiating diffuse alveolar damage. The role of epigenetic histone modifications in organ injury is increasingly recognized. The present study aimed to investigate the use of a histone modification inhibitor to alleviate sepsis-induced ALI, revealing a new strategy for improving sepsis patient survival. Methods In vivo models of ALI were established through the intraperitoneal injection of lipopolysaccharide and cecal ligation and puncture surgery. Furthermore, the disease process was simulated in vitro by stimulating Tamm-Horsfall protein-1 (THP-1) cells with lipopolysaccharide. Hematoxylin and eosin staining, blood gas analysis and pulmonary function tests were utilized to assess the extent of lung tissue damage. Western blot analysis, real-time polymerase chain reaction, enzyme-linked immunosorbent assay and immunofluorescence were used to measure the levels and distribution of the indicated indicators within cells and tissues. Reactive oxygen species and autophagic flux alterations were detected using specific probes. Results BRD3308, which is a inhibitor of histone deacetylase 3, improved lung tissue damage, inflammatory infiltration and edema in ALI by inhibiting Nod-like receptor protein3-mediated pyroptosis in macrophages. By upregulating autophagy, BRD3308 improved the disruption of redox balance in macrophages and reduced the accumulation of reactive oxygen species. Mechanistically, BRD3308 inhibited histone deacetylase 3 activity by binding to it and altering its conformation. Following histone deacetylase 3 inhibition, acetylation of H3K27 was significantly increased. Moreover, the increase in H3K27Ac led to the upregulation of autophagy-related gene 5, a key component of autophagosomes, thereby activating autophagy. Conclusions BRD3308 inhibits oxidative stress and pyroptosis in macrophages by modulating histone acetylation, thereby preventing sepsis-induced ALI. The present study provides a potential strategy and theoretical basis for the clinical treatment of sepsis-induced ALI.
Collapse
Affiliation(s)
- Bohao Liu
- Department of Thoracic Surgery, The First Hospital of Jilin University, 71 Xinmin Street, Chaoyang District, Changchun, Jilin, 130021, China
| | - Ning Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, Hubei, 430060, China
| | - Yi Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, Hubei, 430060, China
| | - Yan Zhang
- Department of Thoracic Surgery, The First Hospital of Jilin University, 71 Xinmin Street, Chaoyang District, Changchun, Jilin, 130021, China
| | - Limei Qu
- Department of Pathology, The First Hospital of Jilin University, 71 Xinmin Street, Chaoyang District, Changchun, Jilin, 130021, China
| | - Hongfei Cai
- Department of Thoracic Surgery, The First Hospital of Jilin University, 71 Xinmin Street, Chaoyang District, Changchun, Jilin, 130021, China
| | - Yang Li
- Department of Thoracic Surgery, The First Hospital of Jilin University, 71 Xinmin Street, Chaoyang District, Changchun, Jilin, 130021, China
- Organ Transplantation Center, The First Hospital of Jilin University, 71 Xinmin Street, Chaoyang District, Changchun, Jilin, 130021, China
| | - Xiaojing Wu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, Hubei, 430060, China
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, Hubei, 430060, China
| |
Collapse
|
8
|
Chen Y, Luo X, Xu B, Bao X, Jia H, Yu B. Oxidative Stress-Mediated Programmed Cell Death: a Potential Therapy Target for Atherosclerosis. Cardiovasc Drugs Ther 2024; 38:819-832. [PMID: 36522550 DOI: 10.1007/s10557-022-07414-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/04/2022] [Indexed: 12/23/2022]
Abstract
Nowadays, as a type of orderly and active death determined by genes, programmed cell death (PCD), including apoptosis, pyroptosis, ferroptosis, and necroptosis, has attracted much attention owing to its participation in numerous chronic cardiovascular diseases, especially atherosclerosis (AS), a canonical chronic inflammatory disease featured by lipid metabolism disturbance. Abundant researches have reported that PCD under distinct internal conditions fulfills different roles of atherosclerotic pathological processes, including lipid core expansion, leukocyte adhesion, and infiltration. Noteworthy, emerging evidence recently has also suggested that oxidative stress (OS), an imbalance of antioxidants and oxygen free radicals, has the potential to mediate PCD occurrence via multiple ways, including oxidization and deubiquitination. Interestingly, more recently, several studies have proposed that the mediating mechanisms could effect on the atherosclerotic initiation and progression significantly from variable aspects, so it is of great clinical importance to clarify how OS-mediated PCD and AS interact. Herein, with the aim of summarizing potential and sufficient atherosclerotic therapy targets, we seek to provide extensive analysis of the specific regulatory mechanisms of PCD mediated by OS and their multifaceted effects on the entire pathological atherosclerotic progression.
Collapse
Affiliation(s)
- Yuwu Chen
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, People's Republic of China
| | - Xing Luo
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, People's Republic of China
| | - Biyi Xu
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, People's Republic of China
| | - Xiaoyi Bao
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, People's Republic of China
| | - Haibo Jia
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China.
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, People's Republic of China.
| | - Bo Yu
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, People's Republic of China
| |
Collapse
|
9
|
Xu Z, Hu H, Wang K, Zhou Z, He X, Huang X, Hu Y, Huang J, Luo Z. Sinensetin, a polymethoxyflavone from citrus fruits, ameliorates LPS-induced acute lung injury by suppressing Txnip/NLRP3/Caspase-1/GSDMD signaling-mediated inflammatory responses and pyroptosis. Food Funct 2024; 15:7592-7604. [PMID: 38938065 DOI: 10.1039/d4fo01704h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/29/2024]
Abstract
Sinensetin (SIN), a polymethoxylated flavonoid, exists widely in citrus fruits with abundant biological activities, such as antioxidant and anti-inflammatory properties, delaying the progression of lung fibers and ameliorating inflammatory lung injury. Herein, an in vivo model of LPS-induced acute lung injury (ALI) in mice and an in vitro model of LPS + IFN-γ-induced M1 polarization in RAW264.7 cells were established to assess the effects and molecular mechanisms of SIN in ameliorating ALI. In the present study, the results showed that SIN significantly reduced BALF IL1β, IL6, and TNF-α levels and neutrophil infiltration, inhibited lung tissue COX2 and iNOS expression, reduced serum and lung tissue inflammatory factor levels, and attenuated lung tissue inflammatory infiltration and ROS levels in animal experiments. RNA sequencing analysis showed that SIN markedly inhibited the expression of inflammation-related pathway genes such as NOD-like receptor signaling. Further mechanistic studies confirmed that SIN significantly inhibited the dissociation of Txnip and Trx-1 and decreased the expression of NLRP3, ASC, pro-Caspase-1, cleavage Caspase-1 p10, NEK7, Caspase-8, IL1β, IL18, and GSDMD. Meanwhile, SIN docked to NLRP3 with strong affinity and bound stably in the hydrophobic docking pocket. Similarly, the same results were observed in in vitro macrophage M1 polarization experiments. In conclusion, the results revealed that SIN ameliorated the onset and progression of ALI by inhibiting Txnip/NLRP3/Caspase-1/GSDMD signaling-mediated inflammatory responses and pyroptosis. These findings emphasize the significant role of SIN in ameliorating ALI and provide insights into the strategy for exploring the functional effects of foods.
Collapse
Affiliation(s)
- Zaibin Xu
- Research Center for Drug Safety Evaluation of Hainan Province, Hainan Medical University, Haikou 571199, China.
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China.
| | - Huiyu Hu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China.
| | - Kongyan Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China.
| | - Ziyi Zhou
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510422, China
| | - Xinqian He
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510422, China
| | - Xinan Huang
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510422, China
| | - Yingjie Hu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China.
| | - Jiawen Huang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China.
| | - Zhuohui Luo
- Research Center for Drug Safety Evaluation of Hainan Province, Hainan Medical University, Haikou 571199, China.
- Hainan Pharmaceutical Research and Development Science Park, Hainan Medical University, Haikou 571199, China
| |
Collapse
|
10
|
Peng Z, Xiao H, Tan Y, Zhang X. Spotlight on macrophage pyroptosis: A bibliometric and visual analysis from 2001 to 2023. Heliyon 2024; 10:e31819. [PMID: 38845992 PMCID: PMC11154638 DOI: 10.1016/j.heliyon.2024.e31819] [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: 12/01/2023] [Revised: 05/22/2024] [Accepted: 05/22/2024] [Indexed: 06/09/2024] Open
Abstract
Macrophage pyroptosis plays a significant role in the pathogenesis of various diseases, especially acute lung injury, atherosclerosis, and sepsis. Despite its importance, analysis of the existing literature has been limited. Therefore, we conducted a bibliometric analysis to provide a comprehensive overview of research on macrophage pyroptosis and identify the current research foci and trends in this field. We collected articles related to macrophage pyroptosis published between 2001 and 2022 from the Web of Science Core Collection and PubMed. Citespace, VOSviewer, bibliometrix R package, and Microsoft Excel 2019 were used to analyze co-occurrence relationships and the contribution of countries/regions, institutions, journals, authors, references, and keywords. In total, 1321 papers were included. China and the United States of America published the most articles in this field. TD Kanneganti had the most publications; BT Cookson was the most cited. Although China contributed the most publications, it had a relatively low ratio of multiple-country collaborations (0.132). Among journals, Frontiers in Immunology and Cell Death Disease published the most papers; Nature and the Journal of Immunology were frequently co-cited. Frequently occurring keywords included "inflammation," "NLRP3 inflammasome," "apoptosis," "caspase-1," and "cell death." Moreover, with the advancement of gene editing technology and the integration of clinical applications, novel molecules ("caspases," "GSDMD," "ASC"), programmed cell death topics ("pyroptosis," "ferroptosis," "necrosis"), and clinical applications ("alveolar macrophage," "atherosclerosis," "prognosis") emerged as frontiers. The macrophage pyroptosis field is rapidly evolving and holds promise as a potential target for treating macrophage pyroptosis-related diseases.
Collapse
Affiliation(s)
- Zhimei Peng
- Department of Nephrology, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, China
- Shenzhen Key Laboratory of Kidney Diseases, Shenzhen People's Hospital, Shenzhen, China
- The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Hua Xiao
- Department of Nephrology, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, China
| | - Yao Tan
- Department of Ophthalmology, The Third Xiangya Hospital, Central South University, No. 138 Tongzipo Road, Yuelu District, Changsha, 410000, China
| | - Xinzhou Zhang
- Department of Nephrology, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, China
- Shenzhen Key Laboratory of Kidney Diseases, Shenzhen People's Hospital, Shenzhen, China
- The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, China
| |
Collapse
|
11
|
Abdel-Hamid GR, Mostafa DM, Fathy RM, Lotfy DM, Osman S. Cytokine storm modulation using cholecalciferol and low dose gamma radiation in Escherichia coli infected mice. Cell Biochem Funct 2024; 42:e4026. [PMID: 38693631 DOI: 10.1002/cbf.4026] [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: 02/20/2024] [Revised: 03/31/2024] [Accepted: 04/21/2024] [Indexed: 05/03/2024]
Abstract
This work investigates the efficiency of cholecalciferol and low dose gamma radiation in modulating cytokine storm through their impact on inflammatory and anti-inflammatory cytokine and protecting against lung and liver injuries. Male Swiss albino mice were exposed to 0.2 Gy gamma radiation/week for four consecutive weeks then injected intraperitoneally (i.p) with a single dose of 8.3 × 106 CFU Escherichia coli/g b.w. then injected i.p. with 1.0 mg/kg cholecalciferol (Vit D3) for 7 days starting 4 h after E. coli injection. The results revealed that Cholecalciferol and low dose gamma radiation caused significant depletion in the severity of E. coli infection (colony forming unit per milliliter), log10 of E. coli, Tumor necrosis factor alpha, Interleukin 6, VEGF, alanine aminotransferase, and aspartate aminotransferase levels and significant elevation in IL-10, IL-4, and HO-1. Immunohistochemical analysis of caspase-3 expression in lung tissue section showed low caspase-3 expression in cholecalciferol and low dose gamma radiation treated group. Histopathological examinations were performed in both lung and liver tissues which also emphasis the biochemical findings. Our results exhibit the importance of cholecalciferol and low dose gamma radiation in improving liver function and providing anti-inflammatory response in diseases causing cytokine storm.
Collapse
Affiliation(s)
- Gehan R Abdel-Hamid
- Radiation Biology, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Dalia M Mostafa
- Radiation Biology, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Rasha M Fathy
- Drug Radiation Research, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Dina M Lotfy
- Drug Radiation Research, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Soheir Osman
- Radiation Biology, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| |
Collapse
|
12
|
Chen M, Zhang J, Huang H, Wang Z, Gao Y, Liu J. miRNA-206-3p alleviates LPS-induced acute lung injury via inhibiting inflammation and pyroptosis through modulating TLR4/NF-κB/NLRP3 pathway. Sci Rep 2024; 14:11860. [PMID: 38789583 PMCID: PMC11126654 DOI: 10.1038/s41598-024-62733-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 05/21/2024] [Indexed: 05/26/2024] Open
Abstract
Acute lung injury (ALI) is life-threatening. MicroRNAs (miRNAs) are often abnormally expressed in inflammatory diseases and are closely associated with ALI. This study investigates whether miRNA-206-3p attenuates pyroptosis in ALI and elucidates the underlying molecular mechanisms. ALI mouse and cell models were established through lipopolysaccharide (LPS) treatment for 24 h. Subsequently, the models were evaluated based on ultrasonography, the lung tissue wet/dry (W/D) ratio, pathological section assessment, electron microscopy, and western blotting. Pyroptosis in RAW264.7 cells was then assessed via electron microscopy, immunofluorescence, and western blotting. Additionally, the regulatory relationship between miRNA-206-3p and the Toll-like receptor (TLR)4/nuclear factor (NF)-κB/Nod-like receptor protein-3 (NLRP3) pathway was verified. Finally, luciferase reporter gene and RNA pull-down assays were used to verify the targeting relationship between miRNA-206-3p and TLR4. miRNA206-3p levels are significantly decreased in the LPS-induced ALI model. Overexpression of miRNA-206-3p improves ALI, manifested as improved lung ultrasound, improved pathological changes of lung tissue, reduced W/D ratio of lung tissue, release of inflammatory factors in lung tissue, and reduced pyroptosis. Furthermore, overexpression of miRNA-206-3p contributed to reversing the ALI-promoting effect of LPS by hindering TLR4, myeloid differentiation primary response 88 (MyD88), NF-κB, and NLRP3 expression. In fact, miRNA-206-3p binds directly to TLR4. In conclusion, miRNA-206-3p alleviates LPS-induced ALI by inhibiting inflammation and pyroptosis via TLR4/NF-κB/NLRP3 pathway modulation.
Collapse
Affiliation(s)
- Mengchi Chen
- The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530000, Guangxi, China
| | - Jingfeng Zhang
- Health Management Center of The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, 528200, Guangdong, China
| | - Hongyuan Huang
- The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530000, Guangxi, China
| | - Zichen Wang
- The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530000, Guangxi, China
| | - Yong Gao
- The First Affiliated Hospital of Guangxi Medical University, Nanning, 530000, Guangxi, China
| | - Jianghua Liu
- The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530000, Guangxi, China.
- School of Nursing, Guangxi Medical University, Nanning, 530000, Guangxi, China.
| |
Collapse
|
13
|
Zhou T, Long K, Chen J, Zhi L, Zhou X, Gao P. Global research progress of endothelial cells and ALI/ARDS: a bibliometric analysis. Front Physiol 2024; 15:1326392. [PMID: 38774649 PMCID: PMC11107300 DOI: 10.3389/fphys.2024.1326392] [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: 11/21/2023] [Accepted: 04/18/2024] [Indexed: 05/24/2024] Open
Abstract
Background Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are severe respiratory conditions with complex pathogenesis, in which endothelial cells (ECs) play a key role. Despite numerous studies on ALI/ARDS and ECs, a bibliometric analysis focusing on the field is lacking. This study aims to fill this gap by employing bibliometric techniques, offering an overarching perspective on the current research landscape, major contributors, and emerging trends within the field of ALI/ARDS and ECs. Methods Leveraging the Web of Science Core Collection (WoSCC) database, we conducted a comprehensive search for literature relevant to ALI/ARDS and ECs. Utilizing Python, VOSviewer, and CiteSpace, we performed a bibliometric analysis on the corpus of publications within this field. Results This study analyzed 972 articles from 978 research institutions across 40 countries or regions, with a total of 5,277 authors contributing. These papers have been published in 323 different journals, spanning 62 distinct research areas. The first articles in this field were published in 2011, and there has been a general upward trend in annual publications since. The United States, Germany, and China are the principal contributors, with Joe G. N. Garcia from the University of Arizona identified as the leading authority in this field. American Journal of Physiology-Lung Cellular and Molecular Physiology has the highest publication count, while Frontiers in Immunology has been increasingly focusing on this field in recent years. "Cell Biology" stands as the most prolific research area within the field. Finally, this study identifies endothelial glycocalyx, oxidative stress, pyroptosis, TLRs, NF-κB, and NLRP3 as key terms representing research hotspots and emerging frontiers in this field. Conclusion This bibliometric analysis provides a comprehensive overview of the research landscape surrounding ALI/ARDS and ECs. It reveals an increasing academic focus on ALI/ARDS and ECs, particularly in the United States, Germany, and China. Our analysis also identifies several emerging trends and research hotspots, such as endothelial glycocalyx, oxidative stress, and pyroptosis, indicating directions for future research. The findings can guide scholars, clinicians, and policymakers in targeting research gaps and setting priorities to advance the field.
Collapse
Affiliation(s)
- Tong Zhou
- Department of Critical Care Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Kunlan Long
- Department of Critical Care Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jun Chen
- Department of Critical Care Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lijia Zhi
- Department of Critical Care Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiujuan Zhou
- Department of Critical Care Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Peiyang Gao
- Department of Critical Care Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
14
|
Yu PR, Tseng CY, Hsu CC, Chen JH, Lin HH. In vitro and in vivo protective potential of quercetin-3-glucuronide against lipopolysaccharide-induced pulmonary injury through dual activation of nuclear factor-erythroid 2 related factor 2 and autophagy. Arch Toxicol 2024; 98:1415-1436. [PMID: 38436694 DOI: 10.1007/s00204-024-03691-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 01/23/2024] [Indexed: 03/05/2024]
Abstract
In vitro and in vivo models of lipopolysaccharide (LPS)-induced pulmonary injury, quercetin-3-glucuronide (Q3G) has been previously revealed the lung-protective potential via downregulation of inflammation, pyroptotic, and apoptotic cell death. However, the upstream signals mediating anti-pulmonary injury of Q3G have not yet been clarified. It has been reported that concerted dual activation of nuclear factor-erythroid 2 related factor 2 (Nrf2) and autophagy may prove to be a better treatment strategy in pulmonary injury. In this study, the effect of Q3G on antioxidant and autophagy were further investigated. Noncytotoxic doses of Q3G abolished the LPS-caused cell injury, and reactive oxygen species (ROS) generation with inductions in Nrf2-antioxidant signaling. Moreover, Q3G treatment repressed Nrf2 ubiquitination, and enhanced the association of Keap1 and p62 in the LPS-treated cells. Q3G also showed potential in inducing autophagy, as demonstrated by formation of acidic vesicular organelles (AVOs) and upregulation of autophagy factors. Next, the autolysosomes formation and cell survival were decreased by Q3G under pre-treatment with a lysosome inhibitor, chloroquine (CQ). Furthermore, mechanistic assays indicated that anti-pulmonary injury effects of Q3G might be mediated via Nrf2 signaling, as confirmed by the transfection of Nrf2 siRNA. Finally, Q3G significantly alleviated the development of pulmonary injury in vivo, which may result from inhibiting the LPS-induced lung dysfunction and edema. These findings emphasize a toxicological perspective, providing new insights into the mechanisms of Q3G's protective effects on LPS-induced pulmonary injury and highlighting its role in dual activating Nrf2 and autophagy pathways.
Collapse
Affiliation(s)
- Pei-Rong Yu
- Department of Nutrition, Chung Shan Medical University, Taichung City, 40201, Taiwan
| | - Chiao-Yun Tseng
- Department of Nutrition, Chung Shan Medical University, Taichung City, 40201, Taiwan
| | - Cheng-Chin Hsu
- Department of Nutrition, Chung Shan Medical University, Taichung City, 40201, Taiwan
| | - Jing-Hsien Chen
- Department of Nutrition, Chung Shan Medical University, Taichung City, 40201, Taiwan
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung City, 40201, Taiwan
| | - Hui-Hsuan Lin
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung City, 40201, Taiwan.
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, No. 110, Sec. 1, Jianguo N. Road, Taichung City, 40201, Taiwan.
| |
Collapse
|
15
|
Kappelhoff S, Margheritis EG, Cosentino K. New insights into Gasdermin D pore formation. Biochem Soc Trans 2024; 52:681-692. [PMID: 38497302 DOI: 10.1042/bst20230549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 03/01/2024] [Accepted: 03/04/2024] [Indexed: 03/19/2024]
Abstract
Gasdermin D (GSDMD) is a pore-forming protein that perforates the plasma membrane (PM) during pyroptosis, a pro-inflammatory form of cell death, to induce the unconventional secretion of inflammatory cytokines and, ultimately, cell lysis. GSDMD is activated by protease-mediated cleavage of its active N-terminal domain from the autoinhibitory C-terminal domain. Inflammatory caspase-1, -4/5 are the main activators of GSDMD via either the canonical or non-canonical pathways of inflammasome activation, but under certain stimuli, caspase-8 and other proteases can also activate GSDMD. Activated GSDMD can oligomerize and assemble into various nanostructures of different sizes and shapes that perforate cellular membranes, suggesting plasticity in pore formation. Although the exact mechanism of pore formation has not yet been deciphered, cysteine residues are emerging as crucial modulators of the oligomerization process. GSDMD pores and thus the outcome of pyroptosis can be modulated by various regulatory mechanisms. These include availability of activated GSDMD at the PM, control of the number of GSDMD pores by PM repair mechanisms, modulation of the lipid environment and post-translational modifications. Here, we review the latest findings on the mechanisms that induce GSDMD to form membrane pores and how they can be tightly regulated for cell content release and cell fate modulation.
Collapse
Affiliation(s)
- Shirin Kappelhoff
- Department of Biology/Chemistry and Center for Cellular Nanoanalytics (CellNanOs), University of Osnabrück, Osnabrück, Germany
| | - Eleonora G Margheritis
- Department of Biology/Chemistry and Center for Cellular Nanoanalytics (CellNanOs), University of Osnabrück, Osnabrück, Germany
| | - Katia Cosentino
- Department of Biology/Chemistry and Center for Cellular Nanoanalytics (CellNanOs), University of Osnabrück, Osnabrück, Germany
| |
Collapse
|
16
|
Osorio-Valencia S, Zhou B. Roles of Macrophages and Endothelial Cells and Their Crosstalk in Acute Lung Injury. Biomedicines 2024; 12:632. [PMID: 38540245 PMCID: PMC10968255 DOI: 10.3390/biomedicines12030632] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/08/2024] [Accepted: 03/10/2024] [Indexed: 11/11/2024] Open
Abstract
Acute lung injury (ALI) and its severe form, acute respiratory distress syndrome (ARDS), present life-threatening conditions characterized by inflammation and endothelial injury, leading to increased vascular permeability and lung edema. Key players in the pathogenesis and resolution of ALI are macrophages (Mφs) and endothelial cells (ECs). The crosstalk between these two cell types has emerged as a significant focus for potential therapeutic interventions in ALI. This review provides a brief overview of the roles of Mφs and ECs and their interplay in ALI/ARDS. Moreover, it highlights the significance of investigating perivascular macrophages (PVMs) and immunomodulatory endothelial cells (IMECs) as crucial participants in the Mφ-EC crosstalk. This sheds light on the pathogenesis of ALI and paves the way for innovative treatment approaches.
Collapse
Affiliation(s)
| | - Bisheng Zhou
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL 60612, USA;
| |
Collapse
|
17
|
Chai Y, Wang Z, Li Y, Wang Y, Wan Y, Chen X, Xu Y, Ge L, Li H. Glycyrrhizin alleviates radiation-induced lung injury by regulating the NLRP3 inflammasome through endoplasmic reticulum stress. Toxicol Res (Camb) 2024; 13:tfae009. [PMID: 38283822 PMCID: PMC10811523 DOI: 10.1093/toxres/tfae009] [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: 11/13/2023] [Revised: 01/02/2024] [Accepted: 01/09/2024] [Indexed: 01/30/2024] Open
Abstract
Objective Radiation pneumonitis (RP) is the major adverse response of radiation therapy for thoracic malignant tumors, and there is a lack of effective interventions. The aim of this study was to investigate the radioprotective effect of Glycyrrhizin (GL) on RP and its potential mechanism. Method The body weight and lung weight of mice were monitored. HE staining was used to observe lung injury, and the expression of endoplasmic reticulum (ER) stress biomarkers and the activation of NLRP3 inflammasome were determined by Western blotting and immunohistochemistry. Flow cytometry was performed to check MLE-12 apoptosis. ER stress activator, Tunicamycin (Tuni), was used to verify the potential mechanism of GL. A systemic pharmacology explored the potential targets and pathways of GL. Results In this study, the lungs of irradiated mice showed significant pneumonic changes. In vivo and in vitro assay, NLRP3 inflammasome was significantly activated, the expression of ER stress biomarkers was elevated, flow cytometry confirms increased apoptosis in irradiated MLE-12 cells. GL inhibits the activation of NLRP3 inflammasome and ER stress pathways. Furthermore, systemic pharmacology revealed that the radioprotective effect of GL may be related to the MAPK signaling pathway. Conclusion In the present study, the results indicated that GL may regulate NLRP3 inflammasome through ER stress, thus exerting irradiation-protective effects on RP, and the ER stress pathway may be a potential target for RP treatment.
Collapse
Affiliation(s)
- Yuqing Chai
- Department of Oncology, The Third Affiliated Hospital of Anhui Medical University, No. 3200, Changsha Road, Baohe District, Hefei, Anhui 230000, China
| | - Ziming Wang
- Department of Oncology, The Third Affiliated Hospital of Anhui Medical University, No. 3200, Changsha Road, Baohe District, Hefei, Anhui 230000, China
| | - Yun Li
- Kindstar Global Precision Medicine Institute, Gaoxin 2nd Road, Jiangxia District, Wuhan, Hubei 43000, China
- Department of Scientific Research Project, Wuhan Kindstar Medical Laboratory Co., Ltd., Guanggu Biological City, No. 666 Gaoxin Avenue, Hongshan District, Wuhan, Hubei 43000, China
| | - Yi Wang
- Department of Oncology, The Third Affiliated Hospital of Anhui Medical University, No. 3200, Changsha Road, Baohe District, Hefei, Anhui 230000, China
| | - Yu Wan
- Department of Oncology, The Third Affiliated Hospital of Anhui Medical University, No. 3200, Changsha Road, Baohe District, Hefei, Anhui 230000, China
| | - Xue Chen
- Department of Oncology, The Third Affiliated Hospital of Anhui Medical University, No. 3200, Changsha Road, Baohe District, Hefei, Anhui 230000, China
| | - Yang Xu
- Department of Oncology, The Third Affiliated Hospital of Anhui Medical University, No. 3200, Changsha Road, Baohe District, Hefei, Anhui 230000, China
| | - Lei Ge
- Department of Oncology, The Third Affiliated Hospital of Anhui Medical University, No. 3200, Changsha Road, Baohe District, Hefei, Anhui 230000, China
| | - Hongxia Li
- Department of Oncology, The Third Affiliated Hospital of Anhui Medical University, No. 3200, Changsha Road, Baohe District, Hefei, Anhui 230000, China
| |
Collapse
|
18
|
Fan C, Zhang Z, Lai Z, Yang Y, Li J, Liu L, Chen S, Hu X, Zhao H, Cui S. Chemical Evolution and Biological Evaluation of Natural Products for Efficient Therapy of Acute Lung Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305432. [PMID: 38126681 PMCID: PMC10870070 DOI: 10.1002/advs.202305432] [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: 08/08/2023] [Revised: 11/01/2023] [Indexed: 12/23/2023]
Abstract
Acute lung injury (ALI) is one of the most common complications in COVID-19 and also a syndrome of acute respiratory failure with high mortality rates, but lacks effective therapeutic drugs. Natural products provide inspiration and have proven to be the most valuable source for bioactive molecule discovery. In this study, the chemical evolution of the natural product Tanshinone IIA (Tan-IIA) to achieve a piperidine-fused scaffold through a synthetic route of pre-activation, multi-component reaction, and post-modification is presented. Through biological evaluation, it is pinpointed that compound 8b is a standout candidate with remarkable anti-inflammation and anti-oxidative stress properties, coupled with low toxicity. The mechanistic study unveils a multifaceted biological profile of 8b and shows that 8b is highly efficient in vivo for the treatment of ALI. Therefore, this work not only provides an effective strategy for the treatment of ALI, but also offers a distinctive natural product-inspired drug discovery.
Collapse
Affiliation(s)
- Chengcheng Fan
- Institute of Drug Discovery and DesignCollege of Pharmaceutical SciencesNational Key Laboratory of Advanced Drug Delivery and Release SystemsZhejiang University866 Yuhangtang RoadHangzhou310058China
| | - Zeyi Zhang
- College of Pharmaceutical SciencesZhejiang Chinese Medical UniversityHangzhou311402China
| | - Zhencheng Lai
- Institute of Drug Discovery and DesignCollege of Pharmaceutical SciencesNational Key Laboratory of Advanced Drug Delivery and Release SystemsZhejiang University866 Yuhangtang RoadHangzhou310058China
| | - Yanzi Yang
- College of Pharmaceutical SciencesZhejiang Chinese Medical UniversityHangzhou311402China
| | - Jiaming Li
- Institute of Drug Discovery and DesignCollege of Pharmaceutical SciencesNational Key Laboratory of Advanced Drug Delivery and Release SystemsZhejiang University866 Yuhangtang RoadHangzhou310058China
| | - Lei Liu
- Institute of Drug Discovery and DesignCollege of Pharmaceutical SciencesNational Key Laboratory of Advanced Drug Delivery and Release SystemsZhejiang University866 Yuhangtang RoadHangzhou310058China
| | - Siyu Chen
- Institute of Drug Discovery and DesignCollege of Pharmaceutical SciencesNational Key Laboratory of Advanced Drug Delivery and Release SystemsZhejiang University866 Yuhangtang RoadHangzhou310058China
| | - Xueping Hu
- Institute of Molecular Sciences and EngineeringInstitute of Frontier and Interdisciplinary ScienceShandong UniversityQingdao266237China
| | - Huajun Zhao
- College of Pharmaceutical SciencesZhejiang Chinese Medical UniversityHangzhou311402China
| | - Sunliang Cui
- Institute of Drug Discovery and DesignCollege of Pharmaceutical SciencesNational Key Laboratory of Advanced Drug Delivery and Release SystemsZhejiang University866 Yuhangtang RoadHangzhou310058China
- Jinhua Institute of Zhejiang UniversityJinhuaZhejiang321299China
| |
Collapse
|
19
|
Xu C, Song C, Wang W, Liu B, Li G, Fu T, Hao B, Li N, Geng Q. Comprehensive analysis of m6A modification in lipopolysaccharide-induced acute lung injury in mice. Mol Med 2024; 30:14. [PMID: 38254010 PMCID: PMC10804706 DOI: 10.1186/s10020-024-00782-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND N6-Methyladenosine (m6A) methylation is the most prevalent post-transcriptional modification in mRNA, and plays significant roles in various diseases. Nevertheless, the precise functions of m6A modification in the formation of ALI remain unclear. In this study we explore the transcriptome distribution of m6A methylation and its probable roles of in ALI. METHODS Lipopolysaccharide (LPS) was utilized to establish an ALI mouse model. Real-time qPCR, Western blotting and m6A dot blot were utilized to assess m6A methylation level and the expression of m6A methylation enzymes. MeRIP-Seq and RNA-seq were utilized to explore differential m6A modifications and differentially expressed genes in ALI mice. The hub genes and enriched pathways were assessed by Real-time qPCR and Western blotting. RESULTS Our findings showed that overall m6A methylation level was increased in ALI mice lung tissues, accompanied by lower levels of METTL3 and FTO. Notably, the protein expression of these methylases were different in various cells. There were 772 differently expressed m6A peaks in ALI as compared to the control group, with 316 being hypermethylated and 456 being hypomethylated. GO and KEGG analyses demonstrated these differentially methylated genes were associated with the calcium signaling pathway and cAMP signaling pathway. Furthermore, we identified 50 genes with distinct m6A peaks and mRNA expressions by combined analysis of MeRIP-Seq and RNA-Seq. KEGG analysis also demonstrated that these overlapped genes were closely associated with the calcium signaling pathway, cGMP-PKG signaling pathway, etc. Besides, Western blotting results demonstrated that the protein expression of Fibronectin leucine-rich transmembrane protein 3 (Flrt3) as well as the calcium signaling pathway and cGMP-PKG signaling pathway, increased significantly after ALI. CONCLUSIONS m6A modification was paramount in the pathogenesis of ALI, and provided a foundation for the further investigation in the prevention and treatment of ALI.
Collapse
Affiliation(s)
- Chenzhen Xu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Congkuan Song
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Wenjie Wang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Bohao Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, 130021, China
| | - Guorui Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Tinglv Fu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Bo Hao
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ning Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
20
|
Huang Q, Le Y, Li S, Bian Y. Signaling pathways and potential therapeutic targets in acute respiratory distress syndrome (ARDS). Respir Res 2024; 25:30. [PMID: 38218783 PMCID: PMC10788036 DOI: 10.1186/s12931-024-02678-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 01/03/2024] [Indexed: 01/15/2024] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a common condition associated with critically ill patients, characterized by bilateral chest radiographical opacities with refractory hypoxemia due to noncardiogenic pulmonary edema. Despite significant advances, the mortality of ARDS remains unacceptably high, and there are still no effective targeted pharmacotherapeutic agents. With the outbreak of coronavirus disease 19 worldwide, the mortality of ARDS has increased correspondingly. Comprehending the pathophysiology and the underlying molecular mechanisms of ARDS may thus be essential to developing effective therapeutic strategies and reducing mortality. To facilitate further understanding of its pathogenesis and exploring novel therapeutics, this review provides comprehensive information of ARDS from pathophysiology to molecular mechanisms and presents targeted therapeutics. We first describe the pathogenesis and pathophysiology of ARDS that involve dysregulated inflammation, alveolar-capillary barrier dysfunction, impaired alveolar fluid clearance and oxidative stress. Next, we summarize the molecular mechanisms and signaling pathways related to the above four aspects of ARDS pathophysiology, along with the latest research progress. Finally, we discuss the emerging therapeutic strategies that show exciting promise in ARDS, including several pharmacologic therapies, microRNA-based therapies and mesenchymal stromal cell therapies, highlighting the pathophysiological basis and the influences on signal transduction pathways for their use.
Collapse
Affiliation(s)
- Qianrui Huang
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095, Jie Fang Avenue, Wuhan, 430030, China
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jie Fang Avenue, Wuhan, 430030, China
| | - Yue Le
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjia Bridge, Hunan Road, Gu Lou District, Nanjing, 210009, China
| | - Shusheng Li
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095, Jie Fang Avenue, Wuhan, 430030, China.
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jie Fang Avenue, Wuhan, 430030, China.
| | - Yi Bian
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095, Jie Fang Avenue, Wuhan, 430030, China.
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jie Fang Avenue, Wuhan, 430030, China.
| |
Collapse
|
21
|
Alzamami A, Radwan EM, Abo-Elabass E, Behery ME, Alshwyeh HA, Al-Olayan E, Altamimi AS, Attallah NGM, Altwaijry N, Jaremko M, Saied EM. Novel 8-Methoxycoumarin-3-Carboxamides with potent anticancer activity against liver cancer via targeting caspase-3/7 and β-tubulin polymerization. BMC Chem 2023; 17:174. [PMID: 38041156 PMCID: PMC10693084 DOI: 10.1186/s13065-023-01063-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 10/23/2023] [Indexed: 12/03/2023] Open
Abstract
In the present study, we explored the potential of coumarin-based compounds, known for their potent anticancer properties, by designing and synthesizing a novel category of 8-methoxycoumarin-3-carboxamides. Our aim was to investigate their antiproliferative activity against liver cancer cells. Toward this, we developed a versatile synthetic approach to produce a series of 8-methoxycoumarin-3-carboxamide analogues with meticulous structural features. Assessment of their antiproliferative activity demonstrated their significant inhibitory effects on the growth of HepG2 cells, a widely studied liver cancer cell line. Among screened compounds, compound 5 exhibited the most potent antiproliferative activity among the screened compounds (IC50 = 0.9 µM), outperforming the anticancer drug staurosporine (IC50 = 8.4 µM), while showing minimal impact on normal cells. The flow cytometric analysis revealed that compound 5 induces cell cycle arrest during the G1/S phase and triggers apoptosis in HepG2 cells by increasing the percentage of cells arrested in the G2/M and pre-G1 phases. Annexin V-FITC/PI screening further supported the induction of apoptosis without significant necrosis. Further, compound 5 exhibited the ability to activate caspase3/7 protein and substantially inhibited β-tubulin polymerization activity in HepG2 cells. Finally, molecular modelling analysis further affirmed the high binding affinity of compound 5 toward the active cavity of β-tubulin protein, suggesting its mechanistic involvement. Collectively, our findings highlight the therapeutic potential of the presented class of coumarin analogues, especially compound 5, as promising candidates for the development of effective anti-hepatocellular carcinoma agents.
Collapse
Affiliation(s)
- Ahmad Alzamami
- Clinical Laboratory Science Department, College of Applied Medical Science, Shaqra University, AlQuwayiyah 11961, Sahqra, Saudi Arabia
| | - Eman M Radwan
- Chemistry Department (The Division of Organic Chemistry), Faculty of Science, Port-Said University, Port-Said, Egypt
| | - Eman Abo-Elabass
- Chemistry Department (The Division of Biochemistry), Faculty of Science, Port-Said University, Port-Said, Egypt
| | - Mohammed El Behery
- Chemistry Department (The Division of Biochemistry), Faculty of Science, Port-Said University, Port-Said, Egypt
| | - Hussah Abdullah Alshwyeh
- Department of Biology, College of Science, Imam Abdulrahman Bin Faisal University, 31441, Dammam, Saudi Arabia
- Basic & Applied Scientific Research Centre, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, 31441, Dammam, Saudi Arabia
| | - Ebtesam Al-Olayan
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Abdulmalik S Altamimi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, PO Box 173, 11942, Alkharj, Saudi Arabia
| | | | - Najla Altwaijry
- Department of Pharmaceutical Sciences, Princess Nourah bint Abdulrahman University, P.O. Box 84428, 11671, Riyadh, Saudi Arabia
| | - Mariusz Jaremko
- Division of Biological and Environmental Sciences and Engineering, Smart-Health Initiative and Red Sea Research Center, King Abdullah University of Science and Technology, P.O. Box 4700, 23955-6900, Thuwal, Saudi Arabia.
| | - Essa M Saied
- Chemistry Department, Faculty of Science, Suez Canal University, Ismailia, 41522, Egypt.
- Institute for Chemistry, Humboldt-Universität zu Berlin, Brook-Taylor-Str. 2, 12489, Berlin, Germany.
| |
Collapse
|
22
|
Wang L, Jiang S, Li X, Lin T, Qin T. Astringin protects LPS-induced toxicity by suppressing oxidative stress and inflammation via suppression of PI3K/AKT/NF-κB pathway for pediatric acute lung injury. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:2369-2377. [PMID: 37193771 DOI: 10.1007/s00210-023-02439-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 02/20/2023] [Indexed: 05/18/2023]
Abstract
Acute lung injury (ALI) is a major pathophysiological problem defined by severe inflammation and acute disease with substantial morbidity and death. It is known that lipopolysaccharide (LPS) induces ALI by causing oxidative stress and inflammation. The goal of this study was to investigate the protective effect of astringin on LPS-induced ALI and probable pathways. Astringin is a stilbenoid, the 3-β-D-glucoside of piceatannol, mainly found in the bark of Picea sitchensis. The findings showed that astringin prevented LPS-induced cellular damage by reducing the generation of oxidative stress in LPS-stimulated A549 lung epithelial cells. Furthermore, astringin extensively decreased the production of inflammatory factors such as TNF-α, IL-1β, and IL-6. In addition, the western blot results revealed that the ability of astringin to reduce oxidative stress and the generation of inflammatory cytokines by inhibiting the ROS-mediated PI3K/AKT/NF-κB pathway could be the reason for its protective effect against LPS-induced ALI. Overall, the results suggest that astringin could be a possible inhibitor of ALI triggered by LPS for pediatric lung injury.
Collapse
Affiliation(s)
- Ling Wang
- Department of Neonatology, The First People's Hospital of Wenling, No.333, Chuanan South Road, Chengxi Street, Wenling, 317500, Zhejiang, China
| | - Shanshan Jiang
- Department of Neonatology, The First People's Hospital of Wenling, No.333, Chuanan South Road, Chengxi Street, Wenling, 317500, Zhejiang, China
| | - Xiaoxiao Li
- Department of Neonatology, The First People's Hospital of Wenling, No.333, Chuanan South Road, Chengxi Street, Wenling, 317500, Zhejiang, China
| | - Tingting Lin
- Department of Neonatology, The First People's Hospital of Wenling, No.333, Chuanan South Road, Chengxi Street, Wenling, 317500, Zhejiang, China.
| | - Tao Qin
- Department of Neonatology, The First People's Hospital of Wenling, No.333, Chuanan South Road, Chengxi Street, Wenling, 317500, Zhejiang, China.
| |
Collapse
|
23
|
Mokra D, Mokry J, Barosova R, Hanusrichterova J. Advances in the Use of N-Acetylcysteine in Chronic Respiratory Diseases. Antioxidants (Basel) 2023; 12:1713. [PMID: 37760016 PMCID: PMC10526097 DOI: 10.3390/antiox12091713] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 08/23/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
N-acetylcysteine (NAC) is widely used because of its mucolytic effects, taking part in the therapeutic protocols of cystic fibrosis. NAC is also administered as an antidote in acetaminophen (paracetamol) overdosing. Thanks to its wide antioxidative and anti-inflammatory effects, NAC may also be of benefit in other chronic inflammatory and fibrotizing respiratory diseases, such as chronic obstructive pulmonary disease, bronchial asthma, idiopathic lung fibrosis, or lung silicosis. In addition, NAC exerts low toxicity and rare adverse effects even in combination with other treatments, and it is cheap and easily accessible. This article brings a review of information on the mechanisms of inflammation and oxidative stress in selected chronic respiratory diseases and discusses the use of NAC in these disorders.
Collapse
Affiliation(s)
- Daniela Mokra
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, SK-03601 Martin, Slovakia; (R.B.); (J.H.)
| | - Juraj Mokry
- Department of Pharmacology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, SK-03601 Martin, Slovakia;
| | - Romana Barosova
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, SK-03601 Martin, Slovakia; (R.B.); (J.H.)
| | - Juliana Hanusrichterova
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, SK-03601 Martin, Slovakia; (R.B.); (J.H.)
| |
Collapse
|
24
|
Yan J, Li Y, Ding L, Hou R, Xing C, Jiang C, Miao Z, Zhuang C. Fragment-Based Discovery of Azocyclic Alkyl Naphthalenesulfonamides as Keap1-Nrf2 Inhibitors for Acute Lung Injury Treatment. J Med Chem 2023. [PMID: 37257073 DOI: 10.1021/acs.jmedchem.3c00686] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Blocking the Kelch-like epichlorohydrin-related protein 1 (Keap1)-nuclear factor-erythroid 2 related factor 2 (Nrf2) pathway is a promising strategy to alleviate acute lung injury (ALI). A naphthalensulfonamide NXPZ-2, targeting Keap1-Nrf2 interaction to release Nrf2, was confirmed to exhibit significant anti-inflammatory activities, however, accompanying nonideal solubility and PK profiles. To further improve the properties, twenty-nine novel naphthalenesulfonamide derivatives were designed by a fragment-based strategy. Among them, compound 10u with a (R)-azetidine group displayed the highest PPI inhibitory activity (KD2 = 0.22 μM). The hydrochloric acid form of 10u exhibited a 9-fold improvement on water solubility (S = 484 μg/mL, pH = 7.0) compared to NXPZ-2 (S = 55 μg/mL, pH = 7.0). It could significantly reduce LPS-induced lung oxidative damages and inflammations in vitro and in vivo. Furthermore, a satisfactory pharmacokinetic property was revealed. In conclusion, the novel azetidine-containing naphthalenesulfonamide represents a promising drug candidate for Keap1-targeting ALI treatment.
Collapse
Affiliation(s)
- Jianyu Yan
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Yue Li
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Li Ding
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Ruilin Hou
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
| | - Chengguo Xing
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Chengshi Jiang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Zhenyuan Miao
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Chunlin Zhuang
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
| |
Collapse
|
25
|
Guo J, Luo Y, Zuo J, Teng J, Shen B, Liu X. Echinacea Polyphenols Inhibit NLRP3-Dependent Pyroptosis, Apoptosis, and Necroptosis via Suppressing NO Production during Lipopolysaccharide-Induced Acute Lung Injury. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:7289-7298. [PMID: 37154470 DOI: 10.1021/acs.jafc.2c08382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
PANoptosis is an intricate programmed death pathway that involves the interaction between pyroptosis, apoptosis, and necroptosis. We systematically explored the protective effect of Echinacea polyphenols (EPP) against the lipopolysaccharide (LPS)-induced acute lung injury (ALI) and the underlying mechanisms both in vitro and in vivo. We noted that EPP pretreatment could significantly alleviate LPS-induced lung tissue injury and pulmonary edema. EPP inhibited the PANoptosis by regulating the expression of nucleotide-binding oligomerization domain-like receptor protein 3 inflammasome, gasdermin D, caspase-8, caspase-3, and mixed lineage kinase domain-like protein. Meanwhile, a comparative study of EPP and inducible nitric oxide synthase inhibitor S-methylisothiourea sulfate indicated that EPP may play a preprotective role in inhibiting PANoptosis via reducing the activity of inducible nitric oxide synthase and the production of nitric oxide (NO) during ALI. Our results clearly indicated that PANoptosis existed in LPS-induced ALI, and EPP pretreatment could provide obvious protective effects to LPS-induced ALI by inhibiting PANoptosis, which may be related to NO production.
Collapse
Affiliation(s)
- Jingjing Guo
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Ying Luo
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Jingru Zuo
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Jiang Teng
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Bingyu Shen
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Xiaoqiang Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| |
Collapse
|
26
|
Fan XY, Ma ZX, Tang LB, Shen HZ, Qi F, Xia JW. lncRNA NEAT1 mediates LPS-induced pyroptosis of BEAS-2B cells via targeting miR-26a-5p/ROCK1 axis. Kaohsiung J Med Sci 2023. [PMID: 37052185 DOI: 10.1002/kjm2.12681] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 02/13/2023] [Accepted: 03/14/2023] [Indexed: 04/14/2023] Open
Abstract
Acute lung injury (ALI) is an adverse disease of the respiratory system, and one of its prevalent causes is sepsis induction. Cell pyroptosis facilitates the progression of ALI and lncRNAs play critical roles in ALI. Thus, this research seeks to investigate the specific mechanism of NEAT1 in sepsis-ALI.BEAS-2B cells were exposed to lipopolysaccharide (LPS) to construct a cell model of sepsis-induced ALI. The gene and protein expression were assessed using qRT-PCR and western blot. Cell viability was identified by CCK-8. Cell death was discovered using PI staining. The secretion of IL-1β and IL-18 was examined using ELISA. The interconnections among NEAT1, miR-26a-5p, and ROCK1 were confirmed using starbase, luciferase assay, and RIP.LPS treatment augmented NEAT1 and ROCK1 levels while mitigating miR-26a-5p level in BEAS-2B cells. Additionally, LPS treatment facilitated cell death and cell pyroptosis, whereas NEAT1 silencing could reverse these effects in BEAS-2B cells. Mechanistically, NEAT1 positively mediated ROCK1 expression by targeting miR-26a-5p. Furthermore, miR-26a-5p inhibitor offset NEAT1 depletion-mediated suppressive effects on cell death and cell pyroptosis. ROCK1 upregulation decreased the inhibitory impacts produced by miR-26a-5p overexpression on cell death and cell pyroptosis. Our outcomes demonstrated NEAT1 could reinforce LPS-induced cell death and cell pyroptosis by repressing the miR-26a-5p/ROCK1 axis, thereby worsening ALI caused by sepsis. Our data indicated NEAT1, miR-26a-5p, and ROCK1 might be biomarkers and target genes for relieving sepsis-induced ALI.
Collapse
Affiliation(s)
- Xiu-Ying Fan
- Department of Critical care, The Third People's Hospital of Kunming, Kunming, Yunnan Province, People's Republic of China
- The Third People's Hospital of Kunming, (Yunnan Infectious Disease Clinical Medical Center), Kunming, Yunnan Province, People's Republic of China
| | - Zhong-Xu Ma
- Department of General Medicine, The Third People's Hospital of Kunming, Kunming, Yunnan Province, People's Republic of China
| | - Li-Bin Tang
- Department of Drug resistance and severe tuberculosis, The Third People's Hospital of Kunming, Kunming, Yunnan Province, People's Republic of China
| | - Han-Zhang Shen
- Department of Critical care, The Third People's Hospital of Kunming, Kunming, Yunnan Province, People's Republic of China
| | - Fei Qi
- Department of Critical care, The Third People's Hospital of Kunming, Kunming, Yunnan Province, People's Republic of China
| | - Jia-Wei Xia
- Department of Critical care, The Third People's Hospital of Kunming, Kunming, Yunnan Province, People's Republic of China
| |
Collapse
|
27
|
NLRX1 knockdown attenuates pro-apoptotic signaling and cell death in pulmonary hyperoxic acute injury. Sci Rep 2023; 13:3441. [PMID: 36859435 PMCID: PMC9975446 DOI: 10.1038/s41598-023-28206-x] [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: 10/19/2022] [Accepted: 01/13/2023] [Indexed: 03/03/2023] Open
Abstract
Hyperoxia is frequently used for treating acute respiratory failure, but it can cause acute lung injury. Nucleotide-binding domain and leucine-rich-repeat-containing family member X1 (NLRX1) is localized in mitochondria and involved in production of reactive oxygen species, inflammation, and apoptosis, which are the features of hyperoxic acute lung injury (HALI). The contribution of NLRX1 to HALI has not previously been addressed. Thus, to investigate the role of NLRX1 in hyperoxia, we generated a murine model of HALI in wild-type (WT) and NLRX1-/- mice by exposure to > 95% oxygen for 72 h. As a result, NLRX1 expression was elevated in mice exposed to hyperoxia. In acute lung injury, levels of inflammatory cells, protein leakage, cell cytotoxicity, and pro-inflammatory cytokines were diminished in NLRX1-/- mice compared to WT mice. In a survival test, NLRX1-/- mice showed reduced mortality under hyperoxic conditions, and apoptotic cell death and caspase expression and activity were also lower in NLRX1-/- mice. Furthermore, levels of the MAPK signaling proteins ERK 1/2, JNK, and p38 were decreased in NLRX1-deficient mice than in WT mice exposed to hyperoxia. The study shows that a genetic deficit in NLRX1 can suppress hyperoxia-induced apoptosis, suggesting that NLRX1 acts as a pivotal regulator of HALI.
Collapse
|
28
|
Mitophagy-promoting miR-138-5p promoter demethylation inhibits pyroptosis in sepsis-associated acute lung injury. Inflamm Res 2023; 72:329-346. [PMID: 36538076 DOI: 10.1007/s00011-022-01675-y] [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: 07/25/2022] [Revised: 07/25/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The present study was designed to explore the potential regulatory mechanism between mitophagy and pyroptosis during sepsis-associated acute lung injury (ALI). METHODS In vitro or in vivo models of sepsis-associated ALI were established by administering lipopolysaccharide (LPS) or performing caecal ligation and puncture (CLP) surgery. Pyroptosis levels were detected by electron microscopy, immunofluorescence, flow cytometry, western blotting and immunohistochemistry. Dual-luciferase reporter gene assay was applied to verify the targeting relationship between miR-138-5p and NLRP3. Methylation-specific PCR and chromatin immunoprecipitation assays were used to determine methylation of the miR-138-5p promoter. Mitophagy levels were examined by transmission electron microscopy and western blotting. RESULTS NLRP3 inflammasome silencing alleviated alveolar macrophage (AM) pyroptosis and septic lung injury. In addition, we confirmed the direct targeting relationship between miR-138-5p and NLRP3. Overexpressed miR-138-5p alleviated AM pyroptosis and the pulmonary inflammatory response. Moreover, the decreased expression of miR-138-5p was confirmed to depend on promoter methylation, while inhibition of miR-138-5p promoter methylation attenuated AM pyroptosis and pulmonary inflammation. Here, we discovered that an increased cytoplasmic mtDNA content in sepsis-induced ALI models induced the methylation of the miR-138-5p promoter, thereby decreasing miR-138-5p expression, which may activate the NLRP3 inflammasome and trigger AM pyroptosis. Mitophagy, a form of selective autophagy that clears damaged mitochondria, reduced cytoplasmic mtDNA levels. Furthermore, enhanced mitophagy might suppress miR-138-5p promoter methylation and relieve the pulmonary inflammatory response, changes that were reversed by treatment with isolated mtDNA. CONCLUSIONS In summary, our study indicated that mitophagy induced the demethylation of the miR-138-5p promoter, which may subsequently inhibit NLRP3 inflammasome, AM pyroptosis and inflammation in sepsis-induced lung injury. These findings may provide a promising therapeutic target for sepsis-associated ALI.
Collapse
|
29
|
Liu J, Dai Y, Lu Y, Liu X, Deng J, Lu W, Liu Q. Identification and validation of a new pyroptosis-associated lncRNA signature to predict survival outcomes, immunological responses and drug sensitivity in patients with gastric cancer. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2023; 20:1856-1881. [PMID: 36899512 DOI: 10.3934/mbe.2023085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
BACKGROUND Gastric cancer (GC) ranks fifth in prevalence among carcinomas worldwide. Both pyroptosis and long noncoding RNAs (lncRNAs) play crucial roles in the occurrence and development of gastric cancer. Therefore, we aimed to construct a pyroptosis-associated lncRNA model to predict the outcomes of patients with gastric cancer. METHODS Pyroptosis-associated lncRNAs were identified through co-expression analysis. Univariate and multivariate Cox regression analyses were performed using the least absolute shrinkage and selection operator (LASSO). Prognostic values were tested through principal component analysis, a predictive nomogram, functional analysis and Kaplan‒Meier analysis. Finally, immunotherapy and drug susceptibility predictions and hub lncRNA validation were performed. RESULTS Using the risk model, GC individuals were classified into two groups: low-risk and high-risk groups. The prognostic signature could distinguish the different risk groups based on principal component analysis. The area under the curve and the conformance index suggested that this risk model was capable of correctly predicting GC patient outcomes. The predicted incidences of the one-, three-, and five-year overall survivals exhibited perfect conformance. Distinct changes in immunological markers were noted between the two risk groups. Finally, greater levels of appropriate chemotherapies were required in the high-risk group. AC005332.1, AC009812.4 and AP000695.1 levels were significantly increased in gastric tumor tissue compared with normal tissue. CONCLUSIONS We created a predictive model based on 10 pyroptosis-associated lncRNAs that could accurately predict the outcomes of GC patients and provide a promising treatment option in the future.
Collapse
Affiliation(s)
- Jinsong Liu
- Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Changzhou 213017, China
- Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Changzhou 213017, China
| | - Yuyang Dai
- Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Changzhou 213017, China
- Department of Radiology, Wujin Hospital Affiliated with Jiangsu University, Changzhou 213017, China
| | - Yueyao Lu
- Department of Oncology, The Changzhou Clinical School of Nanjing Medical University, Changzhou 213017, China
- Department of Oncology, The Wujin Clinical College of Xuzhou Medical University, Changzhou 213017, China
| | - Xiuling Liu
- Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Changzhou 213017, China
- Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Changzhou 213017, China
| | - Jianzhong Deng
- Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Changzhou 213017, China
- Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Changzhou 213017, China
| | - Wenbin Lu
- Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Changzhou 213017, China
- Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Changzhou 213017, China
- Department of Oncology, The Changzhou Clinical School of Nanjing Medical University, Changzhou 213017, China
- Department of Oncology, The Wujin Clinical College of Xuzhou Medical University, Changzhou 213017, China
| | - Qian Liu
- Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Changzhou 213017, China
- Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Changzhou 213017, China
- Department of Oncology, The Wujin Clinical College of Xuzhou Medical University, Changzhou 213017, China
| |
Collapse
|
30
|
Astaxanthin Prevents Tuberculosis-Associated Inflammatory Injury by Inhibiting the Caspase 4/11-Gasdermin-Pyroptosis Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:4778976. [DOI: 10.1155/2022/4778976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 09/08/2022] [Accepted: 09/29/2022] [Indexed: 12/12/2022]
Abstract
Pyroptosis is a programmed cell death caused by inflammation. Multiple studies have suggested that Mycobacterium tuberculosis infection causes tissue pyroptosis. However, there are currently no protective drugs against the inflammatory damage caused by pyroptosis. In this study, anti-pyroptotic effects of the natural compound astaxanthin (ASTA) were explored in a simulated pulmonary tuberculosis-associated inflammatory environment. The results showed that ASTA maintained the stability of MLE-12 lung epithelial cell numbers in the inflammatory environment established by lipopolysaccharide. The reason is not to promote cell proliferation but to inhibit lipopolysaccharide-induced pyroptosis. The results showed that ASTA significantly inhibited the expression of key proteins in the caspase 4/11-gasdermin D pathway and the release of pyroptosis-related inflammatory mediators. Therefore, ASTA inhibits inflammation-induced pyroptosis by inhibiting the caspase 4/11-gasdermin D pathway and has the potential to protect lung tissue from tuberculosis-related inflammatory injury. ASTA, a functional food component, is a promising candidate for protection against tuberculosis-associated inflammatory lung injury.
Collapse
|
31
|
Wang J, Li J, Lou A, Lin Y, Xu Q, Cui W, Huang W, Wang G, Li Y, Sun J, Gong J, Guo Q, Qiu H, Meng Y, Li X. Sacubitril/valsartan alleviates sepsis-induced acute lung injury via inhibiting GSDMD-dependent macrophage pyroptosis in mice. FEBS J 2022; 290:2180-2198. [PMID: 36471663 DOI: 10.1111/febs.16696] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 10/25/2022] [Accepted: 12/05/2022] [Indexed: 12/12/2022]
Abstract
Sepsis-induced acute lung injury (ALI) is a life-threatening disorder with intricate pathogenesis. Macrophage pyroptosis reportedly plays a vital role in ALI. Although it has been established that angiotensin receptor blockers (ARBs) can reduce sepsis-induced organ injury, the efficacy of sacubitril/valsartan (SV) for sepsis has been largely understudied. Here, we aimed to investigate the role of SV in sepsis-induced ALI. Caecal ligation and puncture (CLP) were used to induce polymicrobial sepsis and related ALI. The therapeutic effects of SV in CLP mice were subsequently assessed. Gasdermin D (GSDMD)-/- mice were used to validate the signalling pathways affected by SV. In vitro, mouse bone marrow-derived macrophages (BMDMs) and Raw264.7 cells were treated with SV following exposure to lipopolysaccharide and adenosine triphosphate. Finally, the serum obtained from 42 septic patients was used for biochemical analysis. Compared to the other ARBs, SV yielded more pronounced anti-inflammatory effects on macrophages. In vivo, SV decreased mortality rates, significantly reduced lung damage and prevented the inflammatory response in CLP mice. In addition, SV suppressed GSDMD-mediated macrophage pyroptosis in mice. In BMDMs and Raw264.7 cells, the anti-inflammatory and anti-pyroptosis properties of SV were verified. SV treatment effectively inhibited NLRP3 inflammasome activation and prevented macrophage pyroptosis in a GSDMD-dependent manner. Furthermore, we found that septic individuals had considerably higher serum angiotensin II levels. Overall, we found that SV might prevent ALI in CLP mice by inhibiting GSDMD-mediated pyroptosis of macrophages. Thus, SV might be a viable drug for sepsis-induced ALI.
Collapse
Affiliation(s)
- Jun Wang
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jierui Li
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Anni Lou
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ying Lin
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qihan Xu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wanfu Cui
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Weichang Huang
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Guozhen Wang
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yang Li
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jing Sun
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiacheng Gong
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qiuping Guo
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hongshen Qiu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ying Meng
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xu Li
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Ministry of Education, Key Laboratory of Hainan Trauma and Disaster Rescue, College of Emergency and Trauma, Hainan Medical University, Haikou, China
| |
Collapse
|
32
|
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: 5.3] [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
|
33
|
Gong H, Chen Y, Chen M, Li J, Zhang H, Yan S, Lv C. Advanced development and mechanism of sepsis-related acute respiratory distress syndrome. Front Med (Lausanne) 2022; 9:1043859. [PMID: 36452899 PMCID: PMC9701739 DOI: 10.3389/fmed.2022.1043859] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 10/31/2022] [Indexed: 11/09/2023] Open
Abstract
The introduction of the Sepsis 3.0 guidelines in 2016 improved our understanding of sepsis diagnosis and therapy. Personalized treatment strategies and nursing methods for sepsis patients are recommended in the "Save Sepsis Campaign" in 2021. However, mortality in sepsis patients remains high. Patients with sepsis-related acute respiratory distress syndrome account for around 30% of them, with fatality rates ranging from 30 to 40%. Pathological specimens from individuals with sepsis-related ARDS frequently demonstrate widespread alveolar damage, and investigations have revealed that pulmonary epithelial and pulmonary endothelial injury is the underlying cause. As a result, the purpose of this work is to evaluate the mechanism and research progress of pulmonary epithelial and pulmonary endothelial damage in sepsis-related ARDS, which may provide new directions for future research, diagnosis, and therapy.
Collapse
Affiliation(s)
- Huankai Gong
- Emergency and Trauma College, Hainan Medical University, Haikou, China
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, China
| | - Yao Chen
- Department of Oncology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Meiling Chen
- Emergency and Trauma College, Hainan Medical University, Haikou, China
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, China
| | - Jiankang Li
- Emergency and Trauma College, Hainan Medical University, Haikou, China
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, China
| | - Hong Zhang
- Emergency and Trauma College, Hainan Medical University, Haikou, China
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, China
| | - Shijiao Yan
- Research Unit of Island Emergency Medicine, Chinese Academy of Medical Sciences (No. 2019RU013), Hainan Medical University, Haikou, China
- School of Public Health, Hainan Medical University, Haikou, China
| | - Chuanzhu Lv
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, China
- Research Unit of Island Emergency Medicine, Chinese Academy of Medical Sciences (No. 2019RU013), Hainan Medical University, Haikou, China
- Emergency Medicine Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
34
|
Wei T, Zhang C, Song Y. Molecular mechanisms and roles of pyroptosis in acute lung injury. Chin Med J (Engl) 2022; 135:2417-2426. [PMID: 36583860 PMCID: PMC9945565 DOI: 10.1097/cm9.0000000000002425] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Indexed: 12/31/2022] Open
Abstract
ABSTRACT Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS), which are characterized by excessive inflammation and accompanied by diffuse injury of alveoli, can result in severe respiratory failures. The morbidity and mortality of patients remain high because the major treatments for ALI/ARDS are mainly supportive due to the lack of effective therapies. Numerous studies have demonstrated that the aggravation of coronavirus disease 2019 (COVID-19) leads to severe pneumonia and even ARDS. Pyroptosis, a biological process identified as a type of programed cell death, is mainly triggered by inflammatory caspase activation and is directly meditated by the gasdermin protein family, as well as being associated with the secretion and release of pro-inflammatory cytokines. Clinical and experimental evidence corroborates that pyroptosis of various cells in the lung, such as immune cells and structural cells, may play an important role in the pathogenesis of "cytokine storms" in ALI/ARDS, including those induced by COVID-19. Here, with a focus on ALI/ARDS and COVID-19, we summarized the recent advances in this field and proposed the theory of an inflammatory cascade in pyroptosis to identify new targets and pave the way for new approaches to treat these diseases.
Collapse
Affiliation(s)
- Tianchang Wei
- Department of Pulmonary Medicine, Shanghai Key Laboratory of Lung Inflammation and Injury, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Cuiping Zhang
- Department of Pulmonary Medicine, Shanghai Key Laboratory of Lung Inflammation and Injury, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yuanlin Song
- Department of Pulmonary Medicine, Shanghai Key Laboratory of Lung Inflammation and Injury, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai 200032, China
- Shanghai Respiratory Research Institute, Shanghai 200032, China
- Jinshan Hospital of Fudan University, Shanghai 201508, China
| |
Collapse
|
35
|
Cui E, Zhang L, Pan X, Zhang Q, Zhang L, Wu F, Chen N, Lv L, Chen W, Chen H, Lin A, Wang F, Liang J, Pan R. RNA-Sequencing approach for exploring the therapeutic effect of umbilical cord mesenchymal stem/stromal cells on lipopolysaccharide-induced acute lung injury. Front Immunol 2022; 13:1021102. [PMID: 36341363 PMCID: PMC9632738 DOI: 10.3389/fimmu.2022.1021102] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 10/03/2022] [Indexed: 11/20/2022] Open
Abstract
Acute lung injury (ALI) is significantly associated with morbidity and mortality in patients with critical diseases. In recent years, studies have identified that mesenchymal stem/stromal cells (MSCs) ameliorate ALI and pulmonary fibrosis. However, the mechanism underlying this outcome in ALI has not yet been investigated. In this study, RNA sequencing technology was used to analyze the gene expression profile of lung tissue in lipopolysaccharide (LPS)-induced ALI rats following treatment with human umbilical cord MSC (HUCMSC). Differential expression analyses, gene ontology annotation, Kyoto Encyclopedia of Genes and Genomes enrichment, protein–protein interaction network identification, and hub gene analysis were also performed. HUCMSC treatment decreased inflammatory factor production and alveolar exudates, and attenuated lung damage in LPS-induced ALI rats. The RNA-Seq data indicated that HUCMSC treatment activated the IL-17, JAK-STAT, NF-κB, and TNF-α signaling pathways, increased oxygen transport, and decreased extracellular matrix organization. HUCMSC exert beneficial effects on ALI via these signaling pathways by reducing inflammation, inhibiting pulmonary fibrosis, and improving lung ventilation. Moreover, our study further revealed the hub genes (Tbx2, Nkx2-1, and Atf5) and signaling pathways involved in HUCMSC treatment, thus providing novel perspectives for future research into the molecular mechanisms underlying cell treatment of ALI. HUCMSC can regulate multiple genes and signaling pathways, which can prevent LPS-induced lung damage in an ALI rat model.
Collapse
Affiliation(s)
- Enhai Cui
- Department of Huzhou Central Hospital, Affiliated Huzhou Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Luwen Zhang
- Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, China
| | - Xin Pan
- Institute for Cell-Based Drug Development of Zhejiang Province, S-Evans Biosciences, Hangzhou, China
- Institute for Cell-Based Drug Development of Zhejiang Province, Key Laboratory of Cell-Based Drug and Applied Technology Development in Zhejiang Province, Hangzhou, China
| | - Qiang Zhang
- Institute for Cell-Based Drug Development of Zhejiang Province, S-Evans Biosciences, Hangzhou, China
- Institute for Cell-Based Drug Development of Zhejiang Province, Key Laboratory of Cell-Based Drug and Applied Technology Development in Zhejiang Province, Hangzhou, China
| | - Ling Zhang
- Institute for Cell-Based Drug Development of Zhejiang Province, S-Evans Biosciences, Hangzhou, China
- Institute for Cell-Based Drug Development of Zhejiang Province, Key Laboratory of Cell-Based Drug and Applied Technology Development in Zhejiang Province, Hangzhou, China
| | - Feifei Wu
- Institute for Cell-Based Drug Development of Zhejiang Province, S-Evans Biosciences, Hangzhou, China
- Institute for Cell-Based Drug Development of Zhejiang Province, Key Laboratory of Cell-Based Drug and Applied Technology Development in Zhejiang Province, Hangzhou, China
| | - Na Chen
- Department of Huzhou Central Hospital, Affiliated Huzhou Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Lu Lv
- Department of Huzhou Central Hospital, Affiliated Huzhou Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Wenyan Chen
- Department of Huzhou Central Hospital, Affiliated Huzhou Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Hong Chen
- Institute for Cell-Based Drug Development of Zhejiang Province, S-Evans Biosciences, Hangzhou, China
- Institute for Cell-Based Drug Development of Zhejiang Province, Key Laboratory of Cell-Based Drug and Applied Technology Development in Zhejiang Province, Hangzhou, China
| | - Aifu Lin
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Feng Wang
- Department of Nephrology, Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou Traditional Chinese Medicine (TCM) Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Jinfeng Liang
- Department of Drug Evaluation, Zhejiang Center for Drug & Cosmetic Evaluation, Hangzhou, China
- *Correspondence: Ruolang Pan, ; Jinfeng Liang,
| | - Ruolang Pan
- Institute for Cell-Based Drug Development of Zhejiang Province, S-Evans Biosciences, Hangzhou, China
- Institute for Cell-Based Drug Development of Zhejiang Province, Key Laboratory of Cell-Based Drug and Applied Technology Development in Zhejiang Province, Hangzhou, China
- *Correspondence: Ruolang Pan, ; Jinfeng Liang,
| |
Collapse
|
36
|
Sun J, Li Y. Pyroptosis and respiratory diseases: A review of current knowledge. Front Immunol 2022; 13:920464. [PMID: 36248872 PMCID: PMC9561627 DOI: 10.3389/fimmu.2022.920464] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Pyroptosis is a relatively newly discovered programmed cell death accompanied by an inflammatory response. In the classical view, pyroptosis is mediated by caspases-1,-4,-5,-11 and executed by GSDMD, however, recently it was demonstrated that caspase-3 and-8 also participate in the process of pyroptosis, by cleaving GSDMD/E and GSDMD respectively. Different from autophagy and apoptosis, many pores are formed on the cell membrane during pyroptosis, which makes the cell membrane lose its integrity, eventually leading to the release of cytokines interleukin(IL)-1β and IL-18. When the body is infected with pathogens or exposed to some stimulations, pyroptosis could play an immune defense role. It is found that pyroptosis exists widely in infectious and inflammatory respiratory diseases such as acute lung injury, bronchial dysplasia, chronic obstructive pulmonary disease, and asthma. Excessive pyroptosis may accompany airway inflammation, tissue injury, and airway damage, and induce an inflammatory reaction, leading to more serious damage and poor prognosis of respiratory diseases. This review summarizes the relationship between pyroptosis and related respiratory diseases.
Collapse
|
37
|
Liu Y, Guo K, Ding M, Zhang B, Xiao N, Tang Z, Wang Z, Zhang C, Shubhra QTH. Engineered Magnetic Polymer Nanoparticles Can Ameliorate Breast Cancer Treatment Inducing Pyroptosis-Starvation along with Chemotherapy. ACS APPLIED MATERIALS & INTERFACES 2022; 14:42541-42557. [PMID: 36094305 DOI: 10.1021/acsami.2c13011] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Nanotechnology has shown a revolution in cancer treatments, including breast cancers. However, there remain some challenges and translational hurdles. Surgery, radiotherapy, and chemotherapy are the primary treatment methods for breast cancer, although drug combinations showed promising results in preclinical studies. Herein we report the development of a smart drug delivery system (DDS) to efficiently treat breast cancer by pyroptosis-starvation-chemotherapeutic combination. Cancer-starvation agent glucose oxidase was chemically attached to synthesized iron oxide nanoparticles which were entrapped inside poly(lactic-co-glycolic acid) along with apoptosis-associated speck-like protein containing a caspase recruitment domain plasmid and paclitaxel (PTX). An emulsion solvent evaporation method was used to prepare the DDS. The surface of the DDS was modified with chitosan to which aptamer was attached to achieve site-specific targeting. Hence, the prepared DDS could be targeted to a tumor site by both external magnet and aptamer to obtain an enhanced accumulation of drugs at the tumor site. The final size of the aptamer-decorated DDS was less than 200 nm, and the encapsulation efficiency of PTX was 76.5 ± 2.5%. Drug release from the developed DDS was much higher at pH 5.5 than at pH 7.4, ensuring the pH sensitivity of the DDS. Due to efficient dual targeting of the DDS, in vitro viability of 4T1 cells was reduced to 12.1 ± 1.6%, whereas the nontargeted group and free PTX group could reduce the viability of cells to 29.2 ± 2.4 and 46.2 ± 1.6%, respectively. Our DDS showed a synergistic effect in vitro and no severe side effects in vivo. This DDS has strong potential to treat various cancers.
Collapse
Affiliation(s)
- Yixuan Liu
- Department of Pathology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing 21116, China
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou 350117, China
| | - Kai Guo
- Stomatology Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510140, China
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou 350117, China
| | - Min Ding
- Department of Pathology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing 21116, China
| | - Bingchen Zhang
- Dongguan Hospital, Southern Medical University, Dongguan 523795, China
| | - Nanyang Xiao
- Department of Microbiology, The University of Chicago, Chicago, Illinois 60637, United States
| | - Zonghao Tang
- Drug Discovery Research Center, Southwest Medical University, Luzhou 646000, China
- Cedars-Sinai Medical Center, Los Angeles 90048, United States
| | - Zhengming Wang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Chengfei Zhang
- Department of Pathology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing 21116, China
- Sir Run Run Hospital, Nanjing Medical University, Nanjing 211166, China
| | - Quazi T H Shubhra
- Stomatology Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510140, China
- Translational Medicine Engineering Research Center of Guangdong Province, Foshan First People's Hospital, Foshan 528000, China
| |
Collapse
|
38
|
Mo W, Li Q, Zhou H, Shi X, Yang H, Xiao Z, Wei J, Lv X. Bibliometric analysis of global research trends on pyroptosis in lung disease. Front Immunol 2022; 13:978552. [PMID: 36177039 PMCID: PMC9513361 DOI: 10.3389/fimmu.2022.978552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 08/22/2022] [Indexed: 11/21/2022] Open
Abstract
Background Pyroptosis is a lytic pro-inflammatory programmed cell death mode that depends on caspase, inflammasome, and Gasdermin D (GSDMD). A growing number of studies have shown that pyroptosis is closely related to the pathophysiological mechanism of lung. The purpose of this study is to analyze the literature from Science Citation Index Expanded (SCI-expanded) of Web of Science Core Collection (WoSCC) and visualize the current trends and hotspots in the research of pyroptosis in lung disease. Methods On February 20, 2022, we retrieved all articles on pyroptosis in lung disease from SCI-expanded of WoSCC. Original articles and reviews published in English from 2007 to 2021 were included in the analysis. VOSviewer 1.6.17 and CiteSpace 5.8.R2 were used to analyze the retrieved data and visualize the results. Result 1798 qualified original articles and reviews on pyroptosis in lung disease were included in the bibliometric analysis. So far, the research in this field is still in a period of growth, and the number of global publications has increased yearly. Among the 66 countries that have published relevant articles, China ranked first in the number of publications, and the USA ranked first in the number of cited articles. Holian,A. was the author with the largest number of articles, including 21 published. The University of California System in the USA was the organization with the largest number of articles, totaling 55. Frontiers in Immunology was the journal with the most publications in pyroptosis. After bibliometric analysis, the frequently used keywords are: “NOD-like receptor3 (NLRP3) inflammasome”, “inflammation”, “oxidative stress”, and “acute lung injury (ALI)”. Conclusion The research on pyroptosis in lung disease is in its growth stage. The information released in this article may help researchers better understand the hotspots and developmental trends in this field, the cooperation network information of authors, countries, and institutions, and the citation correlation between articles. With the in-depth study of the mechanism of pyroptosis, the focus has shifted to increasing research on the connections and influences of different diseases. So far, increasing attention has been paid to the research field of the relationship between ALI and pyroptosis related to COVID-19.
Collapse
Affiliation(s)
- Wei Mo
- Graduate School, Wannan Medical College, Wuhu, China
| | - Quanfu Li
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Huanping Zhou
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xuan Shi
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Hao Yang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhuoran Xiao
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Juan Wei
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- *Correspondence: Xin Lv, ; Juan Wei,
| | - Xin Lv
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- *Correspondence: Xin Lv, ; Juan Wei,
| |
Collapse
|
39
|
Surolia R, Antony VB. Pathophysiological Role of Vimentin Intermediate Filaments in Lung Diseases. Front Cell Dev Biol 2022; 10:872759. [PMID: 35573702 PMCID: PMC9096236 DOI: 10.3389/fcell.2022.872759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 04/13/2022] [Indexed: 11/17/2022] Open
Abstract
Vimentin intermediate filaments, a type III intermediate filament, are among the most widely studied IFs and are found abundantly in mesenchymal cells. Vimentin intermediate filaments localize primarily in the cytoplasm but can also be found on the cell surface and extracellular space. The cytoplasmic vimentin is well-recognized for its role in providing mechanical strength and regulating cell migration, adhesion, and division. The post-translationally modified forms of Vimentin intermediate filaments have several implications in host-pathogen interactions, cancers, and non-malignant lung diseases. This review will analyze the role of vimentin beyond just the epithelial to mesenchymal transition (EMT) marker highlighting its role as a regulator of host-pathogen interactions and signaling pathways for the pathophysiology of various lung diseases. In addition, we will also examine the clinically relevant anti-vimentin compounds and antibodies that could potentially interfere with the pathogenic role of Vimentin intermediate filaments in lung disease.
Collapse
Affiliation(s)
| | - Veena B. Antony
- Division of Pulmonary, Allergy and Critical Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
40
|
Fu Z, Wu X, Zheng F, Zhang Y. Sevoflurane anesthesia ameliorates LPS-induced acute lung injury (ALI) by modulating a novel LncRNA LINC00839/miR-223/NLRP3 axis. BMC Pulm Med 2022; 22:159. [PMID: 35473680 PMCID: PMC9044806 DOI: 10.1186/s12890-022-01957-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 04/19/2022] [Indexed: 11/16/2022] Open
Abstract
Background Sevoflurane is considered as a lung-protective factor in acute lung injury (ALI), but the underlying molecular mechanism remains largely unknown. The present study identified for the first time that sevoflurane ameliorated lipopolysaccharide (LPS)-induced ALI through regulating a novel long non-coding RNA LINC00839, and uncovered its regulatory mechanism. Methods LPS-induced ALI models were established in mice or mouse pulmonary microvascular endothelial cells (MPVECs), and they were administered with sevoflurane. Real-Time quantitative PCR, western blot and bioinformatics analysis were performed to screen the aberrantly expressed long non-coding RNA and the downstream molecules in sevoflurane-treated ALI models, and their roles in the protection effect of sevoflurane were verified by functional recovery experiments. Results Sevoflurane relieved LPS-induced lung injury, cell pyroptosis and inflammation in vitro and in vivo. LINC00839 was significantly suppressed by sevoflurane, and overexpression of LINC00839 abrogated the protective effects of sevoflurane on LPS-treated MPVECs. Mechanismly, LINC00839 positively regulated NOD-like receptor protein 3 (NLRP3) via sequestering miR-223. MiR-223 inhibitor reversed the inhibitory effects of LINC00839 knockdown on NLRP3-mediated pyroptosis in LPS-treated MPVECs. Furthermore, both miR-223 ablation and NLRP3 overexpression abrogated the protective effects of sevoflurane on LPS-treated MPVECs. Conclusion In general, our work illustrates that sevoflurane regulates the LINC00839/miR-223/NLRP3 axis to ameliorate LPS-induced ALI, which might provide a novel promising candidate for the prevention of ALI.
Collapse
Affiliation(s)
- Zhiling Fu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, 110004, Liaoning, China
| | - Xiuying Wu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, 110004, Liaoning, China
| | - Fushuang Zheng
- Department of Thoracic Surgery, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, 110004, Liaoning, China
| | - Yan Zhang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, 110004, Liaoning, China.
| |
Collapse
|
41
|
Zhang Y, Zhang J, Fu Z. Molecular hydrogen is a potential protective agent in the management of acute lung injury. Mol Med 2022; 28:27. [PMID: 35240982 PMCID: PMC8892414 DOI: 10.1186/s10020-022-00455-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 02/14/2022] [Indexed: 11/21/2022] Open
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome, which is a more severe form of ALI, are life-threatening clinical syndromes observed in critically ill patients. Treatment methods to alleviate the pathogenesis of ALI have improved to a great extent at present. Although the efficacy of these therapies is limited, their relevance has increased remarkably with the ongoing pandemic caused by the novel coronavirus disease 2019 (COVID-19), which causes severe respiratory distress syndrome. Several studies have demonstrated the preventive and therapeutic effects of molecular hydrogen in the various diseases. The biological effects of molecular hydrogen mainly involve anti-inflammation, antioxidation, and autophagy and cell death modulation. This review focuses on the potential therapeutic effects of molecular hydrogen on ALI and its underlying mechanisms and aims to provide a theoretical basis for the clinical treatment of ALI and COVID-19.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
| | - Jin Zhang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
| | - Zhiling Fu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China.
| |
Collapse
|
42
|
Corry J, Kettenburg G, Upadhyay AA, Wallace M, Marti MM, Wonderlich ER, Bissel SJ, Goss K, Sturgeon TJ, Watkins SC, Reed DS, Bosinger SE, Barratt-Boyes SM. Infiltration of inflammatory macrophages and neutrophils and widespread pyroptosis in lung drive influenza lethality in nonhuman primates. PLoS Pathog 2022; 18:e1010395. [PMID: 35271686 PMCID: PMC8939778 DOI: 10.1371/journal.ppat.1010395] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 03/22/2022] [Accepted: 02/24/2022] [Indexed: 01/04/2023] Open
Abstract
Severe influenza kills tens of thousands of individuals each year, yet the mechanisms driving lethality in humans are poorly understood. Here we used a unique translational model of lethal H5N1 influenza in cynomolgus macaques that utilizes inhalation of small-particle virus aerosols to define mechanisms driving lethal disease. RNA sequencing of lung tissue revealed an intense interferon response within two days of infection that resulted in widespread expression of interferon-stimulated genes, including inflammatory cytokines and chemokines. Macaques with lethal disease had rapid and profound loss of alveolar macrophages (AMs) and infiltration of activated CCR2+ CX3CR1+ interstitial macrophages (IMs) and neutrophils into lungs. Parallel changes of AMs and neutrophils in bronchoalveolar lavage (BAL) correlated with virus load when compared to macaques with mild influenza. Both AMs and IMs in lethal influenza were M1-type inflammatory macrophages which expressed neutrophil chemotactic factors, while neutrophils expressed genes associated with activation and generation of neutrophil extracellular traps (NETs). NETs were prominent in lung and were found in alveolar spaces as well as lung parenchyma. Genes associated with pyroptosis but not apoptosis were increased in lung, and activated inflammatory caspases, IL-1β and cleaved gasdermin D (GSDMD) were present in bronchoalveolar lavage fluid and lung homogenates. Cleaved GSDMD was expressed by lung macrophages and alveolar epithelial cells which were present in large numbers in alveolar spaces, consistent with loss of epithelial integrity. Cleaved GSDMD colocalized with viral NP-expressing cells in alveoli, reflecting pyroptosis of infected cells. These novel findings reveal that a potent interferon and inflammatory cascade in lung associated with infiltration of inflammatory macrophages and neutrophils, elaboration of NETs and cell death by pyroptosis mediates lethal H5N1 influenza in nonhuman primates, and by extension humans. These innate pathways represent promising therapeutic targets to prevent severe influenza and potentially other primary viral pneumonias in humans. Influenza can cause acute lung injury and death, but the mechanisms resulting in lethal influenza in humans are not well understood. We used a novel model of lethal influenza in nonhuman primates caused by aerosol infection with highly pathogenic avian influenza virus that closely resembles human disease to define how the virus causes severe pneumonia. We found that a potent innate immune response starting with high-level production of interferons and inflammatory factors in the lung drives severe disease. Inflammatory cells including macrophages and neutrophils were recruited into lung because of this early response, which in turn led to release of neutrophil extracellular traps that blocked lung alveoli. In addition, a particularly inflammatory form of cell death known as pyroptosis occurred in lungs during lethal influenza. These new findings show that an intense interferon response leading to an inflammatory cascade of macrophages and neutrophils, release of neutrophil extracellular traps, and cell death by pyroptosis is responsible for acute lung injury in lethal influenza. These innate pathways could be targeted by drugs to prevent lung injury in critically ill influenza patients.
Collapse
Affiliation(s)
- Jacqueline Corry
- Department of Infectious Diseases & Microbiology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- * E-mail: (JC); (SMBB)
| | - Gwenddolen Kettenburg
- Department of Infectious Diseases & Microbiology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Amit A. Upadhyay
- Yerkes NHP Genomics Core Laboratory, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Megan Wallace
- Department of Infectious Diseases & Microbiology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Michelle M. Marti
- Department of Infectious Diseases & Microbiology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Elizabeth R. Wonderlich
- Department of Infectious Diseases & Microbiology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Stephanie J. Bissel
- Division of Neuropathology, Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Kyndal Goss
- Yerkes NHP Genomics Core Laboratory, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Timothy J. Sturgeon
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Simon C. Watkins
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Douglas S. Reed
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Steven E. Bosinger
- Yerkes NHP Genomics Core Laboratory, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Simon M. Barratt-Boyes
- Department of Infectious Diseases & Microbiology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- * E-mail: (JC); (SMBB)
| |
Collapse
|
43
|
Liu B, Wang Z, He R, Xiong R, Li G, Zhang L, Fu T, Li C, Li N, Geng Q. Buformin alleviates sepsis-induced acute lung injury via inhibiting NLRP3-mediated pyroptosis through an AMPK-dependent pathway. Clin Sci (Lond) 2022; 136:273-289. [PMID: 35132999 DOI: 10.1042/cs20211156] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 02/02/2022] [Accepted: 02/08/2022] [Indexed: 11/17/2022]
Abstract
BACKGROUND NOD-like receptor family pyrin domain containing 3 (NLRP3)-mediated macrophage pyroptosis plays an important role in sepsis-induced acute lung injury (ALI). Inhibition of pyroptosis may be a way to alleviate inflammation as well as tissue damage triggered after lipopolysaccharide (LPS) stimulation. The aim of the present study was to explore whether buformin (BF), a hypoglycemic agent, could alleviate sepsis-induced ALI by inhibiting pyroptosis. METHODS Wildtype C57BL/6 mice were randomly divided into control group, BF group, LPS group and LPS+BF group. BF group and LPS+BF group were pretreated with BF at a dose of 25 mg/kg, and the changes were observed. In addition, BF was used to interfere with THP-1 cells. The therapeutic effect of BF has been verified by intraperitoneal injection of BF in vivo after LPS stimulation. RESULTS Inflammation and injury was significantly reduced in BF pretreated mice, and the indexes related to pyroptosis were suppressed. The phosphorylation of AMP-activated protein kinase (AMPK) in lung tissues of mice in the BF and LPS+BF groups was significantly higher. In THP-1 cells, the AMPK inhibitor, Compound C was added to demonstrate that BF worked via AMPK to inhibit NLRP3 inflammasome. It was further demonstrated that BF up-regulated autophagy, which in turn promoted NLRP3 inflammasome degradation. On the other hand, BF decreased NLRP3 mRNA level by increasing nuclear factor-erythroid 2 related factor 2 (Nrf2). And BF showed a therapeutic effect after LPS challenge. CONCLUSION Our study confirmed that BF inhibited NLRP3-mediated pyroptosis in sepsis-induced ALI by up-regulating autophagy and Nrf2 protein level through an AMPK-dependent pathway. This provides a new strategy for clinical mitigation of sepsis-induced ALI.
Collapse
Affiliation(s)
- Bohao Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhong Wang
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ruyuan He
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Rui Xiong
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Guorui Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lin Zhang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Tinglv Fu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Chenyuan Li
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ning Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
44
|
Chen R, Kang R, Tang D. The mechanism of HMGB1 secretion and release. Exp Mol Med 2022; 54:91-102. [PMID: 35217834 PMCID: PMC8894452 DOI: 10.1038/s12276-022-00736-w] [Citation(s) in RCA: 356] [Impact Index Per Article: 118.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 10/13/2021] [Accepted: 11/04/2021] [Indexed: 02/08/2023] Open
Abstract
High mobility group box 1 (HMGB1) is a nonhistone nuclear protein that has multiple functions according to its subcellular location. In the nucleus, HMGB1 is a DNA chaperone that maintains the structure and function of chromosomes. In the cytoplasm, HMGB1 can promote autophagy by binding to BECN1 protein. After its active secretion or passive release, extracellular HMGB1 usually acts as a damage-associated molecular pattern (DAMP) molecule, regulating inflammation and immune responses through different receptors or direct uptake. The secretion and release of HMGB1 is fine-tuned by a variety of factors, including its posttranslational modification (e.g., acetylation, ADP-ribosylation, phosphorylation, and methylation) and the molecular machinery of cell death (e.g., apoptosis, pyroptosis, necroptosis, alkaliptosis, and ferroptosis). In this minireview, we introduce the basic structure and function of HMGB1 and focus on the regulatory mechanism of HMGB1 secretion and release. Understanding these topics may help us develop new HMGB1-targeted drugs for various conditions, especially inflammatory diseases and tissue damage. A nuclear protein that gets released after cell death or is actively secreted by immune cells offers a promising therapeutic target for treating diseases linked to excessive inflammation. Daolin Tang from the University of Texas Southwestern Medical Center in Dallas, USA, and colleagues review how cellular stresses can trigger the accumulation of HMGB1, a type of alarm signal protein that promotes the recruitment and activation of inflammation-promoting immune cells. The researchers discuss various mechanisms that drive both passive and active release of HMGB1 into the space around cells. These processes, which include enzymatic modifications of the HMGB1 protein, cell–cell interactions and molecular pathways of cell death, could be targeted by drugs to lessen tissue damage and inflammatory disease caused by HMGB1-induced immune responses
Collapse
Affiliation(s)
- Ruochan Chen
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China. .,Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
45
|
Luo B, Lin J, Cai W, Wang M. Identification of the Pyroptosis-Related Gene Signature and Risk Score Model for Colon Adenocarcinoma. Front Genet 2021; 12:771847. [PMID: 34938319 PMCID: PMC8686197 DOI: 10.3389/fgene.2021.771847] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 11/12/2021] [Indexed: 01/10/2023] Open
Abstract
The prognosis of advanced colon adenocarcinoma (COAD) remains poor. However, existing methods are still difficult to assess patient prognosis. Pyroptosis, a lytic and inflammatory process of programmed cell death caused by the gasdermin protein, is involved in the development and progression of various tumors. Moreover, there are no related studies using pyroptosis-related genes to construct a model to predict the prognosis of COAD patients. Thus, in this study, bioinformatics methods were used to analyze the data of COAD patients downloaded from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases to construct a risk model for the patient prognosis. TCGA database was used as the training set, and GSE39582 downloaded from GEO was used as the validation set. A total of 24 pyroptosis-related genes shown significantly different expression between normal and tumor tissues in COAD and seven genes (CASP4, CASP5, CASP9, IL6, NOD1, PJVK, and PRKACA) screened by univariate and LASSO cox regression analysis were used to construct the risk model. The receiver operating characteristic (ROC) and Kaplan–Meier (K–M curves) curves showed that the model based on pyroptosis-related genes can be used to predict the prognosis of COAD and can be validated by the external cohort well. Then, the clinicopathological factors were combined with the risk score to establish a nomogram with a C-index of 0.774. In addition, tissue validation results also showed that CASP4, CASP5, PRKACA, and NOD1 were differentially expressed between tumor and normal tissues from COAD patients. In conclusion, the risk model based on the pyroptosis-related gene can be used to assess the prognosis of COAD patients well, and the related genes may become the potential targets for treatment.
Collapse
Affiliation(s)
- Bixian Luo
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianwei Lin
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Cai
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingliang Wang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
46
|
Zhang Z, Zhou J, Verma V, Liu X, Wu M, Yu J, Chen D. Crossed Pathways for Radiation-Induced and Immunotherapy-Related Lung Injury. Front Immunol 2021; 12:774807. [PMID: 34925345 PMCID: PMC8672113 DOI: 10.3389/fimmu.2021.774807] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 11/11/2021] [Indexed: 12/19/2022] Open
Abstract
Radiation-induced lung injury (RILI) is a form of radiation damage to normal lung tissue caused by radiotherapy (RT) for thoracic cancers, which is most commonly comprised of radiation pneumonitis (RP) and radiation pulmonary fibrosis (RPF). Moreover, with the widespread utilization of immunotherapies such as immune checkpoint inhibitors as first- and second-line treatments for various cancers, the incidence of immunotherapy-related lung injury (IRLI), a severe immune-related adverse event (irAE), has rapidly increased. To date, we know relatively little about the underlying mechanisms and signaling pathways of these complications. A better understanding of the signaling pathways may facilitate the prevention of lung injury and exploration of potential therapeutic targets. Therefore, this review provides an overview of the signaling pathways of RILI and IRLI and focuses on their crosstalk in diverse signaling pathways as well as on possible mechanisms of adverse events resulting from combined radiotherapy and immunotherapy. Furthermore, this review proposes potential therapeutic targets and avenues of further research based on signaling pathways. Many new studies on pyroptosis have renewed appreciation for the value and importance of pyroptosis in lung injury. Therefore, the authors posit that pyroptosis may be the common downstream pathway of RILI and IRLI; discussion is also conducted regarding further perspectives on pyroptosis as a crucial signaling pathway in lung injury treatment.
Collapse
Affiliation(s)
- Zengfu Zhang
- Department of Radiation Oncology, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Radiation Oncology, Laboratory of Radio-Immunology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Jialin Zhou
- Department of Radiation Oncology, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Radiation Oncology, Laboratory of Radio-Immunology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Vivek Verma
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Xu Liu
- Department of Radiation Oncology, Laboratory of Radio-Immunology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Meng Wu
- Department of Radiation Oncology, Laboratory of Radio-Immunology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Jinming Yu
- Department of Radiation Oncology, Laboratory of Radio-Immunology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Dawei Chen
- Department of Radiation Oncology, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Radiation Oncology, Laboratory of Radio-Immunology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
47
|
Sun H, Hu H, Xu X, Fang M, Tao T, Liang Z. Protective effect of dexmedetomidine in cecal ligation perforation-induced acute lung injury through HMGB1/RAGE pathway regulation and pyroptosis activation. Bioengineered 2021; 12:10608-10623. [PMID: 34747306 PMCID: PMC8810048 DOI: 10.1080/21655979.2021.2000723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 10/26/2021] [Accepted: 10/27/2021] [Indexed: 12/02/2022] Open
Abstract
Dexmedetomidine (DEX) has been reported to attenuate cecal ligation perforation (CLP)-stimulated acute lung injury (ALI) by downregulating HMGB1 and RAGE. This study aimed to further investigate the specific mechanisms of RAGE and its potential-related mechanisms of DEX on ALI models in vitro and in vivo. The in vitro and in vivo ALI models were established by lipopolysaccharide treatment in MLE-12 cells and CLP in mice, respectively. The effect of DEX on pathological alteration was investigated by HE staining. Thereafter, the myeloperoxidase (MPO) activity and inflammatory cytokine levels were respectively detected to assess the lung injury of mice using commercial kits. The expression levels of HMGB1, RAGE, NF-κB, and pyroptosis-related molecules were detected by RT-qPCR and Western blot. HE staining showed that lung injury, increased inflammatory cell infiltration, and lung permeability was found in the ALI mice, and DEX treatment significantly attenuated lung tissue damage induced by CLP. The MPO activity and inflammatory cytokines (TNF-α, IL-1β, and NLRP3) levels were also significantly reduced after DEX treatment compared with those in the ALI mice. Moreover, DEX activated the HMGB1/RAGE/NF-κB pathway and upregulated the pyroptosis-related proteins. However, the protective DEX effect was impaired by RAGE overexpression in ALI mice and MLE-12 cells. Additionally, DEX treatment significantly suppressed HMGB1 translocation from the nucleus region to the cytoplasm, and this effect was reversed by RAGE overexpression. These findings suggested that DEX may be a useful ALI treatment, and the protective effects on ALI mice may be through the inhibition of HMGB1/RAGE/NF-κB pathway and cell pyroptosis.
Collapse
Affiliation(s)
- Huaqin Sun
- Department of Anesthesiology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Hongyi Hu
- Department of Anesthesiology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Xiaoping Xu
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Laboratory Animal Research Center, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Mingsun Fang
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Laboratory Animal Research Center, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Tao Tao
- Department of Anesthesiology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Zhehao Liang
- Department of Ultrasound, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| |
Collapse
|
48
|
Bai T, Cui Y, Yang X, Cui X, Yan C, Tang Y, Cao X, Dong C. miR-302a-3p targets FMR1 to regulate pyroptosis of renal tubular epithelial cells induced by hypoxia-reoxygenation injury. Exp Physiol 2021; 106:2531-2541. [PMID: 34605097 DOI: 10.1113/ep089887] [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: 07/12/2021] [Accepted: 09/29/2021] [Indexed: 12/26/2022]
Abstract
NEW FINDINGS What is the central question of this study? How does miR-302a-3p play a role in hypoxia-reoxygenation-induced pyroptosis of renal tubular epithelial cells? What is the main finding and its importance? Hypoxia-reoxygenation treatment upregulated the expression of miR-302a-3p in HK-2 cells, and then inhibited the transcription of FMRP translational regulator 1 (FMR1), so as to promote the activation of the NLRP3 inflammasome and aggravate the pyroptosis of HK-2 cells. miR-302a-3p was used as a molecular target in this study, which provides a new theoretical basis for the treatment of renal failure. ABSTRACT Hypoxia-reoxygenation (H/R) induction can affect miRNA expression and then control NLR family pyrin domain containing 3 (NLRP3) inflammasome-mediated pyroptosis. This study investigated the mechanism of miR-302a-3p in H/R-induced renal tubular epithelial cell (RTEC) pyroptosis. Human HK-2 RTECs were induced by H/R. Lactate dehydrogenase content, cell activity and pyroptosis, and levels of NLRP3, GSDMD-N, caspase-1, interleukin (IL)-1β, IL-18, superoxide dismutase, and malondialdehyde were detected to verify the effect of H/R on HK-2 cells. The NLRP3 inflammasome action was evaluated after H/R-induced HK-2 cells were treated with BAY11-7082, an inflammasome inhibitor. After inhibiting miR-302a-3p expression, the changes of pyroptosis were observed. The binding relation between miR-302a-3p and FMRP translational regulator 1 (FMR1) was verified. A function-rescue experiment verified the role of FMR1 in the regulation of pyroptosis. H/R-induced HK-2 cells showed significant pyroptosis injury, and the NLRP3 inflammasome was activated. After inhibiting the NLRP3 inflammasome, H/R-induced apoptosis was inhibited. After H/R treatment, miR-302a-3p in HK-2 cells was increased, and miR-302a-3p downregulation limited H/R-induced NLRP3 inflammasome-mediated pyroptosis. FMR1 is the target of miR-302a-3p. Inhibition of FMR1 alleviated the inhibition of H/R-induced HK-2 cell pyroptosis by miR-302a-3p inhibitor. Collectively, inhibiting miR-302a-3p can weaken its targeted inhibition on FMR1, thereby inhibiting the activation of NLRP3 inflammasomes and reducing caspase-1-dependent pyroptosis in HK-2 cells.
Collapse
Affiliation(s)
- Tao Bai
- Pathology Department, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yanzhi Cui
- Medical oncology, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xian Yang
- Pathology Department, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xinyue Cui
- Pathology Department, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Congmin Yan
- Pathology Department, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Ying Tang
- Pathology Department, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiaoming Cao
- Urology Department, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Chunhui Dong
- Department of urinary surgery, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
49
|
Wang Y, Wang X, Zhang H, Han B, Ye Y, Zhang M, Wang Y, Xue J, Wang C. Transforming Growth Factor-β1 Promotes M1 Alveolar Macrophage Polarization in Acute Lung Injury by Up-Regulating DNMT1 to Mediate the microRNA-124/PELI1/IRF5 Axis. Front Cell Infect Microbiol 2021; 11:693981. [PMID: 34504806 PMCID: PMC8421846 DOI: 10.3389/fcimb.2021.693981] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 06/29/2021] [Indexed: 12/12/2022] Open
Abstract
Objective Macrophages function as key orchestrators in the pathogenesis of acute lung injury (ALI). The current study sets out to investigate the molecular mechanism of transforming growth factor-β (TGFβ1) in the regulation of M1 alveolar macrophage polarization in ALI by modulating DNA methyltransferase 1 (DNMT1), along with the microRNA (miR)-124/Pellino 1 (PELI1)/interferon regulatory factor 5 (IRF5) axis. Methods First, ALI mouse models were established, and the proportion of M1 and M2 macrophages in mouse lung tissues was detected using flow cytometry. The targeting relationship between miR-124 and PELI1 was verified with the help of a dual luciferase gene reporter assay. Following TGFβ1 knockdown, RT-qPCR and Western blot assay were performed to analyze the expression patterns of TGFβ1, DNMT1, miR-124, and PELI1 and M1/M2 polarization markers in the lung tissues of ALI mice. Immunofluorescence was further employed to detect nuclear translocation of IRF5 in macrophages. Results The polarization of M1 macrophages was found to be positively correlated with the severity of lung injury. TGFβ1, DNMT1, PELI1 were highly expressed, while miR-124 was down-regulated in ALI mice, and IRF5 was primarily distributed in the nucleus. TGFβ1 promoted the polarization of M1 alveolar macrophages by up-regulating DNMT1. Furthermore, DNMT1 down-regulated the expression of miR-124, which led to enhancement of M1 alveolar macrophage polarization. Meanwhile, over-expression of miR-124 inhibited the nuclear translocation of IRF5 and suppressed M1 alveolar macrophage polarization. On the other hand, over-expression of PELI1 reversed the above trends. Conclusion Collectively, our findings indicated that TGFβ1 can promote the expression of DNMT1, which down-regulates miR-124 to activate PELI1 and nuclear translocation of IRF5, thereby aggravating ALI in mice.
Collapse
Affiliation(s)
- Yongqi Wang
- Department of Anesthesiology, The First Hospital of Lanzhou University, Lanzhou, China
| | - Xiaoqing Wang
- Department of Anesthesiology, The First Hospital of Lanzhou University, Lanzhou, China
| | - Hong Zhang
- Department of Anesthesiology, The First Hospital of Lanzhou University, Lanzhou, China
| | - Biao Han
- Department of Thoracic Surgery, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yuanmei Ye
- Department of Anesthesiology, The First Hospital of Lanzhou University, Lanzhou, China
| | - Mengjie Zhang
- Department of Anesthesiology, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yingbin Wang
- Department of Anesthesiology, Lanzhou University Second Hospital, Lanzhou, China
| | - Jianjun Xue
- Department of Anesthesiology, Gansu Provincial Hospital of TCM, Lanzhou, China
| | - Chun'ai Wang
- Department of Anesthesiology, Gansu Provincial Hospital of TCM, Lanzhou, China
| |
Collapse
|
50
|
Ultramicronized Palmitoylethanolamide Inhibits NLRP3 Inflammasome Expression and Pro-Inflammatory Response Activated by SARS-CoV-2 Spike Protein in Cultured Murine Alveolar Macrophages. Metabolites 2021; 11:metabo11090592. [PMID: 34564408 PMCID: PMC8472716 DOI: 10.3390/metabo11090592] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 08/29/2021] [Accepted: 08/30/2021] [Indexed: 02/06/2023] Open
Abstract
Despite its possible therapeutic potential against COVID-19, the exact mechanism(s) by which palmitoylethanolamide (PEA) exerts its beneficial activity is still unclear. PEA has demonstrated analgesic, anti-allergic, and anti-inflammatory activities. Most of the anti-inflammatory properties of PEA arise from its ability to antagonize nuclear factor-κB (NF-κB) signalling pathway via the selective activation of the PPARα receptors. Acting at this site, PEA can downstream several genes involved in the inflammatory response, including cytokines (TNF-α, Il-1β) and other signal mediators, such as inducible nitric oxide synthase (iNOS) and COX2. To shed light on this, we tested the anti-inflammatory and immunomodulatory activity of ultramicronized(um)-PEA, both alone and in the presence of specific peroxisome proliferator-activated receptor alpha (PPAR-α) antagonist MK886, in primary cultures of murine alveolar macrophages exposed to SARS-CoV-2 spike glycoprotein (SP). SP challenge caused a significant concentration-dependent increase in proinflammatory markers (TLR4, p-p38 MAPK, NF-κB) paralleled to a marked upregulation of inflammasome-dependent inflammatory pathways (NLRP3, Caspase-1) with IL-6, IL-1β, TNF-α over-release, compared to vehicle group. We also observed a significant concentration-dependent increase in angiotensin-converting enzyme-2 (ACE-2) following SP challenge. um-PEA concentration-dependently reduced all the analyzed proinflammatory markers fostering a parallel downregulation of ACE-2. Our data show for the first time that um-PEA, via PPAR-α, markedly inhibits the SP induced NLRP3 signalling pathway outlining a novel mechanism of action of this lipid against COVID-19.
Collapse
|