1
|
Xu H, Luo Y, Li Q, Zhu H. Acupuncture influences multiple diseases by regulating gut microbiota. Front Cell Infect Microbiol 2024; 14:1371543. [PMID: 39040602 PMCID: PMC11260648 DOI: 10.3389/fcimb.2024.1371543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 06/13/2024] [Indexed: 07/24/2024] Open
Abstract
Acupuncture, an important green and side effect-free therapy in traditional Chinese medicine, is widely use both domestically and internationally. Acupuncture can interact with the gut microbiota and influence various diseases, including metabolic diseases, gastrointestinal diseases, mental disorders, nervous system diseases, and other diseases. This review presents a thorough analysis of these interactions and their impacts and examines the alterations in the gut microbiota and the potential clinical outcomes following acupuncture intervention to establish a basis for the future utilization of acupuncture in clinical treatments.
Collapse
Affiliation(s)
- Huimin Xu
- Department of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yingzhe Luo
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Qiaoqi Li
- Department of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hong Zhu
- Department of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
2
|
Yu W, Liang Y, Gao J, Xiong J. Study on risk factors and treatment strategies of hypoxemia after acute type a aortic dissection surgery. J Cardiothorac Surg 2024; 19:273. [PMID: 38702812 PMCID: PMC11067146 DOI: 10.1186/s13019-024-02775-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 04/24/2024] [Indexed: 05/06/2024] Open
Abstract
Acute type A aortic dissection is a life-threatening cardiovascular disease characterized by rapid onset and high mortality. Emergency surgery is the preferred and reliable treatment option. However, postoperative complications significantly impact patient prognosis. Hypoxemia, a common complication, poses challenges in clinical treatment, negatively affecting patient outcomes and increasing the risk of mortality. Therefore, it is crucial to study and comprehend the risk factors and treatment strategies for hypoxemia following acute type A aortic dissection to facilitate early intervention.
Collapse
Affiliation(s)
- Wenbo Yu
- The First Clinical Medical College of Gannan Medical University, Ganzhou, 341000, China
| | - Yuan Liang
- The First Clinical Medical College of Gannan Medical University, Ganzhou, 341000, China
| | - Jianfeng Gao
- The First Clinical Medical College of Gannan Medical University, Ganzhou, 341000, China
| | - Jianxian Xiong
- First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China.
| |
Collapse
|
3
|
Zhao Y, Zhang J, Lu H, Mao Y, Qin J, Wang Y, Wang X, Dai Z, Wang X, Yang Z, Hou L. CARDIOPULMONARY BYPASS-DERIVED PLASMA EXOSOMAL HMGB1 CONTRIBUTES TO ALVEOLAR EPITHELIAL CELL NECROPTOSIS VIA mtDNA/CGAS/STING PATHWAY. Shock 2022; 58:534-541. [PMID: 36516451 DOI: 10.1097/shk.0000000000002006] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
ABSTRACT Our previous study confirmed that cardiopulmonary bypass (CPB) leads to acute lung injury (ALI) via inducing high-mobility group box 1 (HMGB1) release. Recent research showed that HMGB1 promotes pulmonary injury mainly via exosomes transport. Currently, alveolar epithelial cell (AEC) necroptosis has been demonstrated to be involved in ALI. However, it is unknown whether exosomal inflammatory cytokine HMGB1 promotes ALI by inducing AEC necroptosis, and its underlying mechanisms remain elusive. Here, a prospective cohort study was carried out, in which plasma samples from 21 CPB patients were isolated at four specific time points: pre-CPB, 2, 12, and 24 h after initiation of CPB. Plasma exosomes were extracted via ultra-high-speed centrifugation and cocultured with AEC cell line-A549 cells at increasing concentrations of 50, 100, and 150 μg/mL. Then, HMGB1 antagonist-Box A and mtDNA deficiency ethidium bromide (EtBr) were applied to explore the underlying role of exosomal HMGB1 and cytoplasm mitochondrial DNA in AEC. Western blot analysis showed that plasma exosomal HMGB1 expression gradually increased and peaked at 24 h after CPB. Twenty-four-hour treatment of CPB-derived exosomes at 150 μg/mL for 24 h could induce necroptosis by promoting mitochondrial fission and further elevating cytoplasm mtDNA levels in A549 cells, which was successfully blocked by Box A or EtBr. Most importantly, EtBr significantly inhibited cytoplasm mtDNA downstream guanosine monophosphate (GMP)-AMP synthase (cGAS)/stimulator of interferon gene (STING) signal pathway. Collectively, these data demonstrate that CPB-derived plasma exosomal HMGB1 contributes to AEC necroptosis through the mtDNA/cGAS/STING pathway.
Collapse
Affiliation(s)
- Yupeng Zhao
- Department of Anesthesiology and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jinyuan Zhang
- Department of Anesthesiology and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Huihong Lu
- Department of Anesthesiology and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yiliang Mao
- Department of Anesthesiology and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jiawen Qin
- Department of Anesthesiology and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yinglin Wang
- Department of Anesthesiology and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xuebin Wang
- Department of Anesthesiology and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhiguang Dai
- Department of Anesthesiology, Shanghai East Hospital Ji'an Hospital, Ji'an City, Jiangxi Province, China
| | - Xiangrui Wang
- Department of Anesthesiology and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhongwei Yang
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiaotong University, Shanghai, China
| | | |
Collapse
|
4
|
Acupuncture Alleviates Corneal Inflammation in New Zealand White Rabbits with Dry Eye Diseases by Regulating α7nAChR and NF-κB Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:6613144. [DOI: 10.1155/2022/6613144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 02/07/2022] [Accepted: 02/22/2022] [Indexed: 11/15/2022]
Abstract
Purpose. The purpose of this study is to determine the mechanism of improvement in dry eye diseases (DEDs) treated by acupuncture. The inflammatory molecules and related pathways will be analyzed in our study. Methods. In order to establish the animal model for DEDs, healthy New Zealand white rabbits were treated with scopolamine (Scop) hydrobromide for 21 consecutive days. After 21 days, acupuncture, fluorometholone (Flu), and α7nAChR antagonist (α-BGT) treatments were performed, and the Scop injections were continued until day 35. The therapeutic effect of acupuncture on DED inflammation was evaluated by corneal fluorescence staining, tear film rupture time, tear flow measurement, in vivo confocal microscopy (IVCM), corneal histopathology, and cytokine protein chip technology. The influence of acupuncture on the corneal pathology and inflammatory factors ACh, α7nAChR, and NF-κB was detected by enzyme-linked immunosorbent assay (ELISA) and western blot. Results. Compared with the group Scop, acupuncture can significantly reduce corneal staining and increase the tear film rupture time and tear flow, which are accompanied by a decrease in corneal epithelial detachment and lymphocyte infiltration. Acupuncture can relieve the inflammation of corneal stroma and mitigate the expression of proinflammatory factors and chemokines. Acupuncture can upregulate the expression of ACh and α7nAChR and downregulate the expression of NF-κB. Conclusion. Our findings demonstrate that acupuncture can alleviate corneal inflammation in New Zealand white rabbits with DEDs via α7nAChR and NF-κB signaling pathway regulation. The expression indicates that α7nAChR/NF-κB signaling pathway may be active and that acupuncture is a potential therapeutic target for dry eye.
Collapse
|
5
|
Sun G, Zeng Y, Luo F, Zhang L, Tan J, Tong J, Yang L, Liu D, Liu L, Zhou J. Electroacupuncture Preconditioning Alleviates Lipopolysaccharides-Induced Acute Lung Injury by Downregulating LC3-II/I and Beclin 1 Expression. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:8997173. [PMID: 36310624 PMCID: PMC9613389 DOI: 10.1155/2022/8997173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 10/05/2022] [Indexed: 11/15/2022]
Abstract
Our study aimed to investigate the effect of electroacupuncture pretreatment on the inflammatory response and expression levels of LC3-II/I and Beclin 1 using a model of lipopolysaccharide (LPS)-induced acute lung injury (ALI). Eighteen male Sprague-Dawley (SD) rats were randomly divided into three groups: normal control group (NC, n = 6), LSP modeling group (LM, n = 6), and electroacupuncture group (EA, n = 6). Rats in the EA group received electroacupuncture pretreatment at bilateral Zusanli (ST36) and Chize (LU5) points for five days (30 min each time daily, frequency; 3 Hz/15 Hz, intensity; 1 mA). Rats in the EA and LM groups were then injected with 5 mg/kg LPS (Beijing, Solarbio Company, concentration; 5 mg/mL) through the tail vein, while those in the NC group were injected with 5 mg/kg saline. The animals were sacrificed six hours after LPS or saline injection through cervical vertebrae by dislocation under deep anesthesia. Orbital blood was collected for the analysis of serum inflammatory factors including interleukin-1β (IL-1β) and transforming growth factor-β (TGF-β). The lower left lung was excised, stained with hematoxylin-eosin (HE), and subjected to histopathological analysis. The mRNA and protein expression of Beclin 1 and LC3 II/I in the lower right lung tissues were detected via RT-qPCR and Western blot analyses, respectively. The results showed that lung injury score was significantly higher in the LM group than that of the NC group (P < 0.01) and EA group (P < 0.01). The IL-1β contents were significantly decreased in the EA group (P < 0.01) than in the LM group. In contrast, the GF-β contents were increased in the EA group significantly when compared with the LM group (P < 0.01). RT-qPCR and Western blot detection showed that the relative gene expression of LC3-II/I and Beclin 1 was significantly lower in the EA group than in the LM group (P < 0.01). However, the relative protein expression level of LC3-II/I and Beclin 1 was slightly lower in the EA group than the in LM group (P > 0.05). These results show that electroacupuncture pretreatment reduces the inflammatory response in ALI and can protect lung tissue by inhibiting the gene and protein expression levels of LC3-II/I and Beclin 1.
Collapse
Affiliation(s)
- Guanghua Sun
- The First Affiliated Hospital, Department of Rehabilitation, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
- The First Affiliated Hospital, Rehabilitation Medicine Center, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
- The First Affiliated Hospital, Rehabilitation Laboratory, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Yahua Zeng
- The First Affiliated Hospital, Department of Rehabilitation, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
- The First Affiliated Hospital, Rehabilitation Medicine Center, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
- The First Affiliated Hospital, Rehabilitation Laboratory, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Fu Luo
- The First Affiliated Hospital, Department of Rehabilitation, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
- The First Affiliated Hospital, Rehabilitation Medicine Center, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
- The First Affiliated Hospital, Rehabilitation Laboratory, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Lixian Zhang
- Children's Nerve and Development Center, Maternal and Child Health Hospital of Qingyuan City, Qingyuan 511500, Guangdong, China
| | - Jinqu Tan
- The First Affiliated Hospital, Department of Rehabilitation, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
- The First Affiliated Hospital, Rehabilitation Medicine Center, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
- The First Affiliated Hospital, Rehabilitation Laboratory, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Jie Tong
- The First Affiliated Hospital, Department of Rehabilitation, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
- The First Affiliated Hospital, Rehabilitation Medicine Center, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
- The First Affiliated Hospital, Rehabilitation Laboratory, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Lu Yang
- The First Affiliated Hospital, Department of Rehabilitation, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
- The First Affiliated Hospital, Rehabilitation Medicine Center, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
- The First Affiliated Hospital, Rehabilitation Laboratory, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Danni Liu
- The First Affiliated Hospital, Department of Rehabilitation, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
- The First Affiliated Hospital, Rehabilitation Medicine Center, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
- The First Affiliated Hospital, Rehabilitation Laboratory, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Liu Liu
- The First Affiliated Hospital, Department of Rehabilitation, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
- The First Affiliated Hospital, Rehabilitation Medicine Center, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
- The First Affiliated Hospital, Rehabilitation Laboratory, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Jun Zhou
- The First Affiliated Hospital, Department of Rehabilitation, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
- The First Affiliated Hospital, Rehabilitation Medicine Center, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
- The First Affiliated Hospital, Rehabilitation Laboratory, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| |
Collapse
|
6
|
Andersson U, Yang H. HMGB1 is a critical molecule in the pathogenesis of Gram-negative sepsis. JOURNAL OF INTENSIVE MEDICINE 2022; 2:156-166. [PMID: 36789020 PMCID: PMC9924014 DOI: 10.1016/j.jointm.2022.02.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/19/2022] [Accepted: 02/06/2022] [Indexed: 04/12/2023]
Abstract
Gram-negative sepsis is a severe clinical syndrome associated with significant morbidity and mortality. Lipopolysaccharide (LPS), expressed on Gram-negative bacteria, is a potent pro-inflammatory toxin that induces inflammation and coagulation via two separate receptor systems. One is Toll-like receptor 4 (TLR4), expressed on cell surfaces and in endosomes, and the other is the cytosolic receptor caspase-11 (caspases-4 and -5 in humans). Extracellular LPS binds to high mobility group box 1 (HMGB1) protein, a cytokine-like molecule. The HMGB1-LPS complex is transported via receptor for advanced glycated end products (RAGE)-endocytosis to the endolysosomal system to reach the cytosolic LPS receptor caspase-11 to induce HMGB1 release, inflammation, and coagulation that may cause multi-organ failure. The insight that LPS needs HMGB1 assistance to generate severe inflammation has led to successful therapeutic results in preclinical Gram-negative sepsis studies targeting HMGB1. However, to date, no clinical studies have been performed based on this strategy. HMGB1 is also actively released by peripheral sensory nerves and this mechanism is fundamental for the initiation and propagation of inflammation during tissue injury. Homeostasis is achieved when other neurons actively restrict the inflammatory response via monitoring by the central nervous system and the vagus nerve through the cholinergic anti-inflammatory pathway. The neuronal control in Gram-negative sepsis needs further studies since a deeper understanding of the interplay between HMGB1 and acetylcholine may have beneficial therapeutic implications. Herein, we review the synergistic overlapping mechanisms of LPS and HMGB1 and discuss future treatment opportunities in Gram-negative sepsis.
Collapse
Affiliation(s)
- Ulf Andersson
- Department of Women's and Children's Health, Karolinska Institute at Karolinska University Hospital, Stockholm 17176, Sweden
- Corresponding author: Ulf Andersson, Department of Women's and Children's Health, Karolinska Institute at Karolinska University Hospital, Stockholm 17176, Sweden.
| | - Huan Yang
- Institute for Bioelectronic Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY 11030, United States of America
| |
Collapse
|
7
|
Lee S, Kim SN. The Effect of Acupuncture on Modulating Inflammatory Cytokines in Rodent Animal Models of Respiratory Disease: A Systematic Review and Meta-Analysis. Front Immunol 2022; 13:878463. [PMID: 35784312 PMCID: PMC9241441 DOI: 10.3389/fimmu.2022.878463] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 05/11/2022] [Indexed: 11/13/2022] Open
Abstract
PurposeAlthough respiratory diseases (RD) are rapidly becoming a global health issue due to their high mortality and prevalence, there are limitations to the currently available treatments. Acupuncture has been recognized to mitigate many diseases by reducing inflammation and modulating cytokines. However, no systematic analysis has been performed to examine the effects of acupuncture on RD. We aimed to evaluate the effects of acupuncture on rodent animal models of RD.MethodsPubMed, EMBASE, MEDLINE, and the Research Information Service System were searched to retrieve studies that met our inclusion/exclusion criteria. The quality of each included study was evaluated using a 10-item checklist modified from the Collaborative Approach to Meta-Analysis and Review of Animal Data from Experimental Studies. With adequate data extracted, meta-analysis was performed using RevMan software.ResultsA total of 18 studies were included, and the mean quality assessment was 5.7. The meta-analysis revealed that acupuncture had a significant effect on changing the cytokine levels, including pro-/anti-inflammatory, Th1-, Th2- and Th17- specific cytokines.ConclusionAlthough there were limitations in the number of included studies, the results suggest that acupuncture can be a possible treatment for RD through its modulation of various cytokines, leading to reduced inflammation.
Collapse
|
8
|
Zhang Y, Zheng L, Deng H, Feng D, Hu S, Zhu L, Xu W, Zhou W, Wang Y, Min K, Zhou Q, Chen Y, Zhou H, Yang H, Lv X. Electroacupuncture Alleviates LPS-Induced ARDS Through α7 Nicotinic Acetylcholine Receptor-Mediated Inhibition of Ferroptosis. Front Immunol 2022; 13:832432. [PMID: 35222419 PMCID: PMC8866566 DOI: 10.3389/fimmu.2022.832432] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 01/19/2022] [Indexed: 12/12/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is an uncontrollable, progressive pulmonary inflammatory disease, and as a common clinical critical disease, there is no effective treatment available. Electroacupuncture (EA) therapy is a type of traditional Chinese medicine physiotherapy that can alleviate the inflammatory response. However, the potential mechanism of EA in the treatment of ARDS is not yet clear. Ferroptosis is a new type of programmed cell death characterized by intracellular iron accumulation and lipid peroxidation. Recently, emerging evidence has shown that ferroptosis is closely related to the occurrence and development of ARDS caused by various pathological factors. Here, we further investigated whether EA-mediated inhibition of ferroptosis in lung tissue could attenuate lipopolysaccharide (LPS)-induced ARDS and explored its underlying mechanisms. In this study, mice were administered LPS intraperitoneally to establish a model of LPS-induced ARDS. We found that EA stimulation could not only reduce the exudation of inflammatory cells and proteins in the alveolar lumen but also significantly alleviate the pathological changes of lung tissue, inhibit the production of proinflammatory cytokines and improve the survival rate of mice. Concurrently, we also found that ferroptosis events occurred in the lung tissue of LPS-induced ARDS mice, manifested by elevated iron levels, ROS production and lipid peroxidation. Intriguingly, our results showed that EA stimulation at the Zusanli (ST36) acupoint activated α7 nicotinic acetylcholine receptor (α7nAchR) in lung tissue mainly through the sciatic nerve and cervical vagus nerve, thus exerting anti-ferroptosis and pulmonary protective effects. Additionally, these effects were eliminated by methyllycaconitine (MLA), a selective antagonist of α7nAchR. In vitro experiments, activation of α7nAchR protected alveolar epithelial cells from LPS-induced ferroptosis. Furthermore, our experiments showed that the pulmonary protective effects of EA stimulation were effectively reversed by erastin, a ferroptosis activator. Collectively, we demonstrated that EA stimulation could alleviate LPS-induced ARDS by activating α7nAchR to inhibit LPS-induced ferroptosis in alveolar epithelial cells. Targeting and regulating ferroptosis in alveolar epithelial cells may be a potential intervention approach for the treatment of LPS-induced ALI/ARDS in the future.
Collapse
Affiliation(s)
- Yiguo Zhang
- Graduate School, Wannan Medical College, Wuhu, China
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Li Zheng
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Huimin Deng
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Di Feng
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Song Hu
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lina Zhu
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wenting Xu
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wenyu Zhou
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yu Wang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Keting Min
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qing Zhou
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yuanli Chen
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Huanping Zhou
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hao Yang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xin Lv
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
9
|
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: 411] [Impact Index Per Article: 137.0] [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
|
10
|
Li N, Guo Y, Gong Y, Zhang Y, Fan W, Yao K, Chen Z, Dou B, Lin X, Chen B, Chen Z, Xu Z, Lyu Z. The Anti-Inflammatory Actions and Mechanisms of Acupuncture from Acupoint to Target Organs via Neuro-Immune Regulation. J Inflamm Res 2022; 14:7191-7224. [PMID: 34992414 PMCID: PMC8710088 DOI: 10.2147/jir.s341581] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 12/01/2021] [Indexed: 12/17/2022] Open
Abstract
Inflammation plays a significant role in the occurrence and development of multiple diseases. This study comprehensively reviews and presents literature from the last five years, showing that acupuncture indeed exerts strong anti-inflammatory effects in multiple biological systems, namely, the immune, digestive, respiratory, nervous, locomotory, circulatory, endocrine, and genitourinary systems. It is well known that localized acupuncture-mediated anti-inflammatory effects involve the regulation of multiple populations and functions of immune cells, including macrophages, granulocytes, mast cells, and T cells. In acupuncture stimulation, macrophages transform from the M1 to the M2 phenotype and the negative TLR4 regulator PPARγ is activated to inhibit the intracellular TLR/MyD88 and NOD signaling pathways. The downstream IκBα/NF-κB and P38 MAPK pathways are subsequently inhibited by acupuncture, followed by suppressed production of inflammasome and proinflammatory mediators. Acupuncture also modulates the balance of helper T cell populations. Furthermore, it inhibits oxidative stress by enhancing SOD activity via the Nrf2/HO-1 pathway and eliminates the generation of oxygen free radicals, thereby preventing inflammatory cell infiltration. The anti-inflammatory effects of acupuncture on different biological systems are also specific to individual organ microenvironments. As part of its anti-inflammatory action, acupuncture deforms connective tissue and upregulates the secretion of various molecules in acupoints, further activating the NF-κB, MAPK, and ERK pathways in mast cells, fibroblasts, keratinocytes, and monocytes/macrophages. The somatic afferents present in acupuncture-activated acupoints also convey sensory signals to the spinal cord, brainstem, and hypothalamic neurons. Upon information integration in the brain, acupuncture further stimulates multiple neuro-immune pathways, including the cholinergic anti-inflammatory, vagus-adrenal medulla-dopamine, and sympathetic pathways, as well as the hypothalamus-pituitary-adrenal axis, ultimately acting immune cells via the release of crucial neurotransmitters and hormones. This review provides a scientific and reliable basis and viewpoints for the clinical application of acupuncture in various inflammatory conditions.
Collapse
Affiliation(s)
- Ningcen Li
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin City, People's Republic of China.,School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin City, People's Republic of China
| | - Yi Guo
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin City, People's Republic of China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin City, People's Republic of China.,School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin City, People's Republic of China
| | - Yinan Gong
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin City, People's Republic of China.,School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin City, People's Republic of China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin City, People's Republic of China
| | - Yue Zhang
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin City, People's Republic of China.,School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin City, People's Republic of China
| | - Wen Fan
- Suzuka University of Medical Science, Suzuka City, Japan
| | - Kaifang Yao
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin City, People's Republic of China.,School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin City, People's Republic of China
| | - Zhihan Chen
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin City, People's Republic of China.,School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin City, People's Republic of China
| | - Baomin Dou
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin City, People's Republic of China.,School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin City, People's Republic of China
| | - Xiaowei Lin
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin City, People's Republic of China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin City, People's Republic of China.,School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin City, People's Republic of China
| | - Bo Chen
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin City, People's Republic of China.,School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin City, People's Republic of China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin City, People's Republic of China
| | - Zelin Chen
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin City, People's Republic of China.,School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin City, People's Republic of China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin City, People's Republic of China
| | - Zhifang Xu
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin City, People's Republic of China.,School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin City, People's Republic of China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin City, People's Republic of China
| | - Zhongxi Lyu
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin City, People's Republic of China.,School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin City, People's Republic of China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin City, People's Republic of China
| |
Collapse
|
11
|
Abdel-Rafei MK, Thabet NM, El Tawel G, El Bakary NM, El Fatih NM, Sh Azab K. Role of leptin/STAT3 signaling and RIP-kinases in fucoxanthin influences on mice exposed to LPS and gamma radiation. TOXIN REV 2021. [DOI: 10.1080/15569543.2021.2008451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Mohamed K. Abdel-Rafei
- Radiation Biology Department, National Centre for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Noura M. Thabet
- Radiation Biology Department, National Centre for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Ghada El Tawel
- Radiation Biology Department, National Centre for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Nermeen M. El Bakary
- Radiation Biology Department, National Centre for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Neama M. El Fatih
- Radiation Biology Department, National Centre for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Khaled Sh Azab
- Radiation Biology Department, National Centre for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo, Egypt
| |
Collapse
|
12
|
Andersson U, Tracey KJ, Yang H. Post-Translational Modification of HMGB1 Disulfide Bonds in Stimulating and Inhibiting Inflammation. Cells 2021; 10:cells10123323. [PMID: 34943830 PMCID: PMC8699546 DOI: 10.3390/cells10123323] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/23/2021] [Accepted: 11/25/2021] [Indexed: 12/16/2022] Open
Abstract
High mobility group box 1 protein (HMGB1), a highly conserved nuclear DNA-binding protein, is a “damage-associated molecular pattern” molecule (DAMP) implicated in both stimulating and inhibiting innate immunity. As reviewed here, HMGB1 is an oxidation-reduction sensitive DAMP bearing three cysteines, and the post-translational modification of these residues establishes its proinflammatory and anti-inflammatory activities by binding to different extracellular cell surface receptors. The redox-sensitive signaling mechanisms of HMGB1 also occupy an important niche in innate immunity because HMGB1 may carry other DAMPs and pathogen-associated molecular pattern molecules (PAMPs). HMGB1 with DAMP/PAMP cofactors bind to the receptor for advanced glycation end products (RAGE) which internalizes the HMGB1 complexes by endocytosis for incorporation in lysosomal compartments. Intra-lysosomal HMGB1 disrupts lysosomal membranes thereby releasing the HMGB1-transported molecules to stimulate cytosolic sensors that mediate inflammation. This HMGB1-DAMP/PAMP cofactor pathway slowed the development of HMGB1-binding antagonists for diagnostic or therapeutic use. However, recent discoveries that HMGB1 released from neurons mediates inflammation via the TLR4 receptor system, and that cancer cells express fully oxidized HMGB1 as an immunosuppressive mechanism, offer new paths to targeting HMGB1 for inflammation, pain, and cancer.
Collapse
Affiliation(s)
- Ulf Andersson
- Department of Women’s and Children’s Health, Karolinska Institute, Karolinska University Hospital, 17176 Stockholm, Sweden
- Correspondence: ; Tel.: +46-(70)-7401740
| | - Kevin J. Tracey
- Institute for Bioelectronic Medicine, The Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY 11030, USA; (K.J.T.); (H.Y.)
| | - Huan Yang
- Institute for Bioelectronic Medicine, The Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY 11030, USA; (K.J.T.); (H.Y.)
| |
Collapse
|
13
|
Electroacupuncture Alleviates Inflammation of Dry Eye Diseases by Regulating the α7nAChR/NF- κB Signaling Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6673610. [PMID: 33897942 PMCID: PMC8052151 DOI: 10.1155/2021/6673610] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 02/21/2021] [Accepted: 03/08/2021] [Indexed: 01/30/2023]
Abstract
Purpose We tried to investigate whether electroacupuncture (EA) can reduce inflammation of dry eye disease (DED) by regulating α7nAChR and inhibiting the NF-κB signaling pathway. Methods Healthy New Zealand white rabbits were treated with scopolamine hydrobromide (Scop) for 21 consecutive days to establish the DED animal model. After 21 days, EA, fluorometholone (Flu), and α7nAChR antagonist (α-BGT) treatments were performed, and the Scop injection was continued until day 35. During treatment, the fluorescence staining of the corneal epithelium and the level of tear flow were observed. The influence of EA on the LG pathology and inflammatory factors ACh, α7nAChR, and NF-κB was detected using the LG histopathology, transmission electron microscopy (TEM), cytokine protein chip technology, enzyme-linked immunosorbent assay (ELISA), and Western blot. Results The EA stimulation can reduce the corneal epithelial damage and repair epithelial cell ultrastructure, promote the tear secretion, relieve the LG atrophy and decrease lipid droplet accumulation in LG acinar cell, and reduce the levels of inflammatory cytokines (i.e., IL-1, MIP-1b, TNF-α, and IL-8) in the LG. The protective effect of EA on the inflammation and the ocular surface is similar to the corticosteroid Flu. EA and Flu can upregulate the expression of the α7nAChR and downregulate the expression of NF-κB. The α7nAChR antagonist α-BGT can reverse the protective effect of EA on the LG and the inhibitory effect on the NF-κB pathway and the expression of inflammatory factors but cannot affect the expression of Flu on the NF-κB pathway and inflammatory factors. Conclusion These results prove that EA can alleviate DEDs by stimulating the acupoints around the eyes. These beneficial effects are related to the upregulation of α7nAChR and the downregulation of NF-κB in the LG. The protective effect of LG is mediated through the anti-inflammatory pathway mediated by α7nAChR. EA can reduce the NF-κB P65 nuclear transcription and reduce inflammatory factors by regulating α7nAChR. This expression indicates that the α7nAChR/NF-κB signaling pathway may serve as a potential therapeutic target for EA to treat DEDs.
Collapse
|
14
|
Hou L, Zhang J, Liu Y, Fang H, Liao L, Wang Z, Yuan J, Wang X, Sun J, Tang B, Chen H, Ye P, Ding Z, Lu H, Wang Y, Wang X. MitoQ alleviates LPS-mediated acute lung injury through regulating Nrf2/Drp1 pathway. Free Radic Biol Med 2021; 165:219-228. [PMID: 33539948 DOI: 10.1016/j.freeradbiomed.2021.01.045] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/18/2021] [Accepted: 01/23/2021] [Indexed: 12/12/2022]
Abstract
Lipopolysaccharide (LPS) has been known to cause alveolar epithelial cell (AEC) apoptosis and barrier breakdown that characterize acute lung injury (ALI) and acute respiratory distress syndrome. We aimed to investigate whether mitoquinone (MitoQ), a mitochondria-targeted antioxidant, could alleviate LPS-induced AEC damage in ALI and its underlying mechanisms. In vitro studies in AEC A549 cell line, we noted that LPS could induce dynamin-related protein 1 (Drp1)-mediated mitochondrial fission, AEC apoptosis and barrier breakdown, which could be reversed with MitoQ and mitochondrial division inhibitor 1 treatment. Moreover, the protective role of MitoQ was attenuated with Drp1 overexpression. Nuclear factor E2-related factor 2 (Nrf2) downregulation could block the effect of MitoQ by decreasing the expression of Nrf2 target genes in LPS-treated AEC, such as heme oxygenase-1 (HO-1) and NAD(P)H:quinone oxidoreductase 1 (NQO1). Nrf2 gene knockdown in LPS-treated A549 cells prevented the protective effect of MitoQ from decreasing Drp1-mediated mitochondrial fission, AEC apoptosis and barrier breakdown. The lung protective effect of MitoQ by regulating the Drp1-mediated mitochondrial fission, AEC apoptosis and barrier breakdown was further confirmed in vivo with LPS-induced ALI mouse model. Additionally, the protective effect of MitoQ was inhibited by Nrf2 inhibitor ML385. We therefore conclude that MitoQ exerts ALI-protective effects by preventing Nrf2/Drp1-mediated mitochondrial fission, AEC apoptosis as well as barrier breakdown.
Collapse
Affiliation(s)
- Lei Hou
- Department of Anesthesiology and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong, Shanghai, 200120, China
| | - Jinyuan Zhang
- Department of Anesthesiology and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong, Shanghai, 200120, China
| | - Yajing Liu
- Department of Anesthesiology and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong, Shanghai, 200120, China
| | - Hongwei Fang
- Department of Anesthesiology and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong, Shanghai, 200120, China
| | - Lijun Liao
- Department of Anesthesiology and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong, Shanghai, 200120, China
| | - Zhankui Wang
- Department of Orthopedics, The First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Jie Yuan
- Department of Pain, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, 563000, China
| | - Xuebin Wang
- Department of Anesthesiology and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong, Shanghai, 200120, China
| | - Jixiong Sun
- Department of Anesthesiology and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong, Shanghai, 200120, China
| | - Bing Tang
- Department of Anesthesiology and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong, Shanghai, 200120, China
| | - Hongfei Chen
- Department of Anesthesiology and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong, Shanghai, 200120, China
| | - Pengcheng Ye
- Department of Anesthesiology and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong, Shanghai, 200120, China
| | - Zhenmin Ding
- Department of Anesthesiology and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong, Shanghai, 200120, China
| | - Huihong Lu
- Department of Anesthesiology and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong, Shanghai, 200120, China.
| | - Yinglin Wang
- Department of Anesthesiology and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong, Shanghai, 200120, China.
| | - Xiangrui Wang
- Department of Anesthesiology and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong, Shanghai, 200120, China.
| |
Collapse
|
15
|
Cardiopulmonary Bypass Induces Acute Lung Injury via the High-Mobility Group Box 1/Toll-Like Receptor 4 Pathway. DISEASE MARKERS 2020; 2020:8854700. [PMID: 33062073 PMCID: PMC7532999 DOI: 10.1155/2020/8854700] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 08/23/2020] [Accepted: 08/31/2020] [Indexed: 11/25/2022]
Abstract
During cardiopulmonary bypass (CPB), pulmonary ischemia/reperfusion (I/R) injury can cause acute lung injury (ALI). Our previous research confirmed that abnormal high-mobility group box 1 (HMGB1) release after CPB was closely related to ALI. However, the mechanism underlying the HMGB1-mediated induction of ALI after CPB is unclear. Our previous study found that HMGB1 binds Toll-like receptor 4 (TLR4), leading to lung injury, but direct evidence of a role for these proteins in the mechanism of CPB-induced lung injury has not been shown. We examined the effects of inhibiting HMGB1 or reducing TLR4 expression on CPB-induced lung injury in rats administered anti-HMBG1 antibody or TLR4 short-hairpin RNA (shTLR4), respectively. In these rat lungs, we studied the histologic changes and levels of interleukin- (IL-) 1β, tumour necrosis factor- (TNF-) α, HMGB1, and TLR4 after CPB. After CPB, the lung tissues from untreated rats showed histologic features of injury and significantly elevated levels of IL-1β, TNF-α, HMGB1, and TLR4. Treatment with anti-HMGB1 attenuated the CPB-induced morphological inflammatory response and protein levels of IL-1β, TNF-α, HMGB1, and TLR4 in the lung tissues and eventually alleviated the ALI after CPB. Treatment with shTLR4 attenuated the CPB-induced morphological inflammatory response and protein levels of IL-1β, TNF-α, and TLR4 in the lung tissues and eventually alleviated the ALI after CPB, but could not alleviate the HMGB1 protein levels induced by CPB. In summary, the present study demonstrated that the HMGB1/TLR4 pathway mediated the development of ALI induced by CPB.
Collapse
|
16
|
Electroacupuncture Alleviates Osteoarthritis by Suppressing NLRP3 Inflammasome Activation in Guinea Pigs. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:5476064. [PMID: 32952587 PMCID: PMC7487102 DOI: 10.1155/2020/5476064] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 08/09/2020] [Accepted: 08/10/2020] [Indexed: 12/21/2022]
Abstract
Osteoarthritis (OA) is an increasingly prevalent disease affecting synovial joints, which includes joint degeneration, inflammation, and joint pain. The activation of nucleotide-binding and oligomerization domain-like receptor containing protein 3 (NLRP3) could promote synovial inflammation. Previous studies have shown that electroacupuncture (EA) has potential anti-inflammatory effect. However, the effect of EA treatment on OA remains unclear. The aim of this study was to investigate the effect of applied EA on OA and joint pain and its relationship with NLRP3 inflammasome. The Hartley guinea pigs with naturally occurring OA at age 18 months were chosen as the OA model and treated with EA for 4 weeks. Mechanical allodynia was quantified by using von Frey filaments. The expression of NLRP3 inflammasome and the downstream proinflammatory factors in the cartilage tissue were quantified. Our results showed that EA treatment significantly reduces mechanical allodynia, improves the articular cartilage structure, and decreases the fibrillation on the cartilage surface in guinea pigs with spontaneous osteoarthritis. Moreover, we also found that EA treatment attenuates the NLRP3 inflammasome activation and suppresses the protein expression levels of caspase-1 and IL-1β in the cartilage tissue. Our findings suggest that EA treatment attenuates OA and joint pain by suppressing NLRP3 inflammasome activation and support further investigation of the potential therapeutic tactics.
Collapse
|
17
|
Electroacupuncture Pretreatment Alleviates LPS-Induced Acute Respiratory Distress Syndrome via Regulating the PPAR Gamma/NF-Kappa B Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:4594631. [PMID: 32774418 PMCID: PMC7396021 DOI: 10.1155/2020/4594631] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 06/27/2020] [Accepted: 06/30/2020] [Indexed: 11/18/2022]
Abstract
Electroacupuncture (EA) is reported to possess anti-inflammatory properties and has beneficial effects on acute respiratory distress syndrome (ARDS). However, the underlying mechanisms of the effects of EA on ARDS remain unclear. This study aims to investigate the protective effect of EA on LPS-induced ARDS. In this study, Sprague-Dawley male rats were treated with EA at Hegu (LI4) for 45 minutes before LPS instillation (0.4 mg/kg, 100 ul). H&E staining, wet-to-dry weight (W/D) ratio, PaO2, and protein content in BALF were employed to determine the function of lung tissues. Inflammatory cytokines in serum and BALF were detected by enzyme-linked immunoassay assay (ELISA). The levels of oxidative stress markers were detected to determine the oxidative stress status. Cell apoptosis was observed by terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL) staining and western blot. Here, we found that EA pretreatment effectively alleviated lung pathological damage. Moreover, EA suppressed the oxidative stress damage by upregulating glutathione and superoxide dismutase and downregulating malondialdehyde. EA pretreatment also regulated apoptosis-related proteins, such as Bax and Bcl-2. We found that peroxisome proliferators-activated receptors γ (PPARγ) play a critical role during ARDS, EA up-regulated the expression of PPARγ, which inhibited the activation of nuclear factor-kappa B (NF-κB) and decreased the inflammatory cytokines (interleukin-1β, interleukin-6, and tumor necrosis factor-α). When rats were treated with GW9662, a selective PPARγ antagonist, these effects of EA were reversed. Our study demonstrated that EA pretreatment had a beneficial effect on LPS-induced ARDS in rats by anti-inflammatory, antioxidative, and antiapoptotic properties which was regulated via PPARγ/NF-κB signaling pathway.
Collapse
|
18
|
Dexmedetomidine attenuates inflammation and pancreatic injury in a rat model of experimental severe acute pancreatitis via cholinergic anti-inflammatory pathway. Chin Med J (Engl) 2020; 133:1073-1079. [PMID: 32265428 PMCID: PMC7213633 DOI: 10.1097/cm9.0000000000000766] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Excessive inflammatory responses play a critical role in the development of severe acute pancreatitis (SAP), and controlling such inflammation is vital for managing this often fatal disease. Dexmedetomidine has been reported to possess protective properties in inflammatory diseases. Therefore, this study aimed to investigate whether dexmedetomidine pre-treatment exerts an anti-inflammatory effect in rats with SAP induced by sodium taurocholate, and if so, to determine the potential mechanism. METHODS SAP was induced with sodium taurocholate. Rats received an intraperitoneal injection of dexmedetomidine 30 min before sodium taurocholate administration. α-bungarotoxin, a selective alpha-7 nicotinic acetylcholine receptor (α7nAchR) antagonist, was injected intra-peritoneally 30 min before dexmedetomidine administration. The role of the vagus nerve was evaluated by performing unilateral cervical vagotomy before the administration of dexmedetomidine. Efferent discharge of the vagal nerve was recorded by the BL-420F Data Acquisition & Analysis System. Six hours after onset, serum pro-inflammatory cytokine (tumor necrosis factor α [TNF-α] and interleukin 6 [IL-6]) levels and amylase levels were determined using an enzyme-linked immunosorbent assay and an automated biochemical analyzer, respectively. Histopathological changes in the pancreas were observed after hematoxylin and eosin staining and scored according to Schmidt criteria. RESULTS Pre-treatment with dexmedetomidine significantly decreased serum levels of TNF-α, IL-6, and amylase, strongly alleviating pathological pancreatic injury in the rat model of SAP (TNF-α: 174.2 ± 30.2 vs. 256.1±42.4 pg/ml; IL-6: 293.3 ± 46.8 vs. 421.7 ± 48.3 pg/ml; amylase: 2102.3 ± 165.3 vs. 3186.4 ± 245.2 U/L). However, the anti-inflammatory and pancreatic protective effects were abolished after vagotomy or pre-administration of α-bungarotoxin. Dexmedetomidine also significantly increased the discharge frequency and amplitude of the cervical vagus nerve in the SAP rat model (discharge frequency: 456.8 ± 50.3 vs. 332.4 ± 25.1 Hz; discharge amplitude: 33.4 ± 5.3 vs. 20.5 ± 2.9 μV). CONCLUSIONS Dexmedetomidine administration attenuated the systemic inflammatory response and local pancreatic injury caused by SAP in rats through the cholinergic anti-inflammatory pathway involving vagus- and α7nAChR-dependent mechanisms.
Collapse
|
19
|
Sheng W, Yang H, Niu Z, Yin H. Anti-apoptosis effect of heme oxygenase-1 on lung injury after cardiopulmonary bypass. J Thorac Dis 2020; 12:1393-1403. [PMID: 32395277 PMCID: PMC7212168 DOI: 10.21037/jtd.2020.03.48] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Background This study aimed to investigate the anti-apoptosis effects of heme oxygenase-1 (HO-1) on lung injury (LI) after cardiopulmonary bypass (CPB) and its probable mechanisms. Methods One hundred and forty-four male Wistar rats were divided into 3 groups randomly: group A (control group), group B (cobalt protoporphyrin, CoPP), and group C [CoPP plus zinc protoporphyrin (ZnPP)]. Lung tissues were harvested at different time: before CPB (T0), 0 min after CPB (T1), 2 h after CPB (T2), 6 h (T3), 12 h (T4), and 24 h (T5). Results The HO-1 protein expressions in lung tissue in group B were higher than those in group A and group C in any given time, and the same as HO-1 activity (P<0.05). The expressions of Bcl-2 protein in group B at all time point after bypass were higher than those in group A and group C, and the difference was statistically significant (P<0.05). Apoptosis index (AI) in group B at any time point after bypass were lower than those in group A and group C (P<0.05). Conclusions CoPP can significantly increase the expression of HO-1 protein in lung tissue. HO-1 is still highly expressed after CPB, so as to play an important part in anti-apoptosis, and reduce LI.
Collapse
Affiliation(s)
- Wei Sheng
- Department of Cardiovascular Surgery, Qingdao Municipal Hospital, Medical College of Qingdao University, Qingdao 266071, China
| | - Haiqin Yang
- Department of Mental Intervention, Qingdao Preferential Hospital, Qingdao 266071, China
| | - Zhaozhuo Niu
- Department of Cardiovascular Surgery, Qingdao Municipal Hospital, Medical College of Qingdao University, Qingdao 266071, China
| | - Hong Yin
- Department of Cardiovascular Surgery, Qingdao Municipal Hospital, Medical College of Qingdao University, Qingdao 266071, China
| |
Collapse
|
20
|
Huang D, Chen M, Wang Z, Hou L, Yu W. Electroacupuncture Pretreatment Attenuates Inflammatory Lung Injury After Cardiopulmonary Bypass by Suppressing NLRP3 Inflammasome Activation in Rats. Inflammation 2019; 42:895-903. [PMID: 30680695 DOI: 10.1007/s10753-018-0944-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cardiopulmonary bypass (CPB) can induce inflammatory lung injury, which is a common complication during cardiac surgery. Nod-like receptor family, pyrin domain-containing 3 (NLRP3) inflammasome-induced inflammation plays a crucial role in lung injury after CPB. Previous studies have shown that electroacupuncture (EA) has potential anti-inflammatory activity. However, the role of EA in CPB is poorly understood. The aim of this study was to determine whether EA was associated with CPB-induced inflammatory lung injury. In the present study, rats were treated with EA for 5 days before CPB. Two hours after CPB, the lung tissue, serum, and bronchoalveolar lavage fluid (BALF) were prepared for assessment. Our results showed that the expression of NLRP3 in the lung tissue increased significantly after CPB. The EA pretreatment suppressed NLRP3 inflammasome activation, reduced lung edema, and inhibited IL-1β release into the serum and BALF after CPB. Our findings suggest that EA pretreatment attenuates inflammatory lung injury after CPB by suppressing NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Dongxiao Huang
- The Third Affiliated Hospital of Soochow University, No.185 Juqian Street, Changzhou, Jiangsu, China.,Department of Anesthesiology, Wuxi People's Hospital, No.299 Qingyang Road, Wuxi, Jiangsu, China
| | - Mo Chen
- The Third Affiliated Hospital of Soochow University, No.185 Juqian Street, Changzhou, Jiangsu, China
| | - Zhankui Wang
- Department of Orthopedics, The First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Lei Hou
- Department of Anesthesiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, No.160 Pudian Road, Shanghai, China
| | - Weifeng Yu
- Department of Anesthesiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, No.160 Pudian Road, Shanghai, China. .,Department of Anesthesiology, Shanghai Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, No.225 Changhai Road, Shanghai, 200438, People's Republic of China.
| |
Collapse
|
21
|
Dhar R, Zhang L, Li Y, Rana MN, Hu Z, Li Z, Cui H, Tang H. Electroacupuncture ameliorates cardiopulmonary bypass induced apoptosis in lung via ROS/Nrf2/NLRP3 inflammasome pathway. Life Sci 2019; 238:116962. [PMID: 31628913 DOI: 10.1016/j.lfs.2019.116962] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 10/04/2019] [Accepted: 10/12/2019] [Indexed: 02/06/2023]
Abstract
AIMS Electroacupuncture (EAc) has a pulmonary protective effect during cardiopulmonary bypass (CPB), but its molecular mechanisms including inflammasome activation signaling pathways remains unclear. MATERIALS AND METHODS Male Sprague Dawley rats were divided into control, CPB + EAc and CPB groups. Lung injury model was developed by CPB treatment and EAc (2/100 Hz) was carried out before CPB in the CPB + EAc group. Lung tissues were collected at two time points (0.5 h; 2 h) to determine cytokines release by ELISA kits, and protein expressions by Western blot. Serum collected at two time points (0.5 h; 2 h) from CPB and CPB + EAc treated groups were used in NR8383 cells to confirm the effect of EAc. KEY FINDINGS CPB significantly increased the inflammatory mediators, histological damage and expression of inflammasome related protein and apoptosis, when compared with control group. The level of tumor necrosis factor-α(TNF-α), interleukin (IL)-18 and IL-1β in the CPB + EAc treated group was significantly decreased along with histological changes compared to CPB. Moreover, EAc inhibited the activation of Nod like receptor protein-3 (NLRP3) inflammasome complex, caspase-8 and activated NF-E2-related factor 2 (p-Nrf2). In addition, serum from the CPB + EAc group prevented CPB induced activation of inflammasome and related mediators, reducing ROS generation and apoptosis in NR8383 macrophages. SIGNIFICANCE These findings indicate that EAc had a critical anti-apoptotic role by suppression of ROS/Nrf2/NLRP3 inflammasome pathway. EAc might be a possible therapeutic treatment for CPB-induced acute lung injury.
Collapse
Affiliation(s)
- Rana Dhar
- Department of Pharmacology, School of Basic Medical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Lejun Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yajun Li
- Department of Pharmacology, School of Basic Medical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Mohammad Nasiruddin Rana
- Department of Pharmacology, School of Basic Medical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Zhengqiang Hu
- Department of Pharmacology, School of Basic Medical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Zigang Li
- Department of Anesthesiology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Huashun Cui
- Department of Acupuncture, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Huifang Tang
- Department of Pharmacology, School of Basic Medical Sciences, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
22
|
|