1
|
Wang J, Li LL, Zhao ZA, Niu CY, Zhao ZG. NLRP3 Inflammasome-mediated pyroptosis in acute lung injury: Roles of main lung cell types and therapeutic perspectives. Int Immunopharmacol 2025; 154:114560. [PMID: 40184810 DOI: 10.1016/j.intimp.2025.114560] [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: 02/16/2025] [Revised: 03/14/2025] [Accepted: 03/23/2025] [Indexed: 04/07/2025]
Abstract
The NLRP3 inflammasome plays a pivotal role in the pathogenesis of acute lung injury (ALI) by regulating pyroptosis, a highly inflammatory form of programmed cell death. NLRP3-mediated pyroptosis leads to alveolar epithelial cell injury, increased pulmonary microvascular endothelial permeability, excessive alveolar macrophage activation, and neutrophil dysfunction, collectively driving ALI progression. In addition to the classical NLRP3-dependent pathway, the non-canonical pyroptosis pathway (caspase-4/5/11) also contributes to ALI by inducing pyroptotic cell death in AECs and ECs, further amplifying NLRP3 activation through damage-associated molecular patterns (DAMP) release. Moreover, neutrophils (NE) pyroptosis exhibits dual roles in ALI, as it enhances pathogen clearance but also exacerbates excessive inflammation and tissue damage, highlighting the complexity of its regulation. Targeting the NLRP3 inflammasome and pyroptotic pathways has emerged as a promising therapeutic strategy for ALI. Various NLRP3 inhibitors (e.g., MCC950, CY-09, OLT1177) and pyroptosis inhibitors have demonstrated significant anti-inflammatory and tissue-protective effects in preclinical models. However, the clinical translation of NLRP3-targeted therapies remains challenging due to off-target effects, potential immunosuppression, lack of patient stratification strategies, and compensatory activation of alternative inflammasomes (e.g., AIM2, NLRC4). Future studies should focus on optimizing the selectivity of NLRP3 inhibitors, developing personalized therapeutic approaches, and exploring combination strategies to enhance their clinical applicability in ALI.
Collapse
Affiliation(s)
- Jing Wang
- Department of Pathophysiology in Basic Medical College, Hebei Medical University, Shijiazhuang, Hebei 050017, China; Institute of Microcirculation, Hebei North University, Zuanshinan Road 11, Zhangjiakou, Hebei 075000, China
| | - Lu-Lu Li
- Institute of Microcirculation, Hebei North University, Zuanshinan Road 11, Zhangjiakou, Hebei 075000, China
| | - Zhen-Ao Zhao
- Institute of Microcirculation, Hebei North University, Zuanshinan Road 11, Zhangjiakou, Hebei 075000, China
| | - Chun-Yu Niu
- Department of Pathophysiology in Basic Medical College, Hebei Medical University, Shijiazhuang, Hebei 050017, China; Institute of Microcirculation, Hebei North University, Zuanshinan Road 11, Zhangjiakou, Hebei 075000, China; Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Zhangjiakou, Hebei 075000, China.
| | - Zi-Gang Zhao
- Institute of Microcirculation, Hebei North University, Zuanshinan Road 11, Zhangjiakou, Hebei 075000, China; Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Zhangjiakou, Hebei 075000, China.
| |
Collapse
|
2
|
Liu C, Zhou R, Chen B, Yan X, Guo L, Tang Y, Zuo X, Guo X, Yu H, Chen J, Guo Z, Wang F, Xu C. Inflammatory microenvironment-responsive nanomicelles for acute lung injury therapy: ROS-scavenging and macrophage repolarization. Mater Today Bio 2025; 31:101622. [PMID: 40104650 PMCID: PMC11919404 DOI: 10.1016/j.mtbio.2025.101622] [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: 12/03/2024] [Revised: 02/09/2025] [Accepted: 02/26/2025] [Indexed: 03/20/2025] Open
Abstract
The pathogenesis of acute lung injury (ALI) is characterized by an uncontrolled inflammatory response, marked by excessive production of reactive oxygen species (ROS) and the infiltration of inflammatory cells, particularly macrophages, which play a pivotal role in disease progression. The synergistic effect of ROS scavenging and macrophage repolarization provides a promising strategy for effective ALI treatment. Herein, we developed a novel type of self-assembling nanomicelles, which were composed of poly-L-glutamic acid (PLG) and 4-Hydroxymethyl phenylboronic acid (PBA). The nanomicelles (PPDex micelles) had a high drug-loading capacity for dexamethasone (Dex) based on boronic ester bonds, which exhibited reversible cleavage under inflammatory conditions characterized by elevated levels of ROS or decreased pH values. These PPDex micelles revealed rapid drug-responsive release behavior in the inflammatory environment, and in vivo studies demonstrated their efficacy in modulating cytokines, inhibiting oxidative stress, and promoting macrophage polarization from the pro-inflammatory M1 phenotype to the anti-inflammatory M2 phenotype, which ultimately suppressed the progression of ALI. Moreover, the PPDex micelles had the effective ability to effectively suppress the NF-кB and ROS/NLRP3 inflammatory pathways. Therefore, this study presented a novel and potent therapeutic strategy for ALI treatment, which could promote the clinical application of polymer nanomicelles in the treatment of ALI.
Collapse
Affiliation(s)
- Chang Liu
- College of Basic Medical Sciences, The Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Rui Zhou
- College of Basic Medical Sciences, The Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Baiqiao Chen
- College of Basic Medical Sciences, The Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Xinran Yan
- College of Basic Medical Sciences, The Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Lei Guo
- College of Basic Medical Sciences, The Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Yixin Tang
- College of Basic Medical Sciences, The Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Xu Zuo
- College of Basic Medical Sciences, The Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Xiaoping Guo
- College of Basic Medical Sciences, The Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Haiyang Yu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Jie Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Zhaopei Guo
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Fang Wang
- College of Basic Medical Sciences, The Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Caina Xu
- College of Basic Medical Sciences, The Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| |
Collapse
|
3
|
de Sales PVA, Godói IPD, Brito GADC, Leitão RC, de Araújo AA, de Medeiros CACX. Mechanisms of photobiomodulation therapy in treating and preventing antineoplastic-induced oral mucositis: a systematic review. Acta Cir Bras 2025; 40:e403125. [PMID: 40172371 PMCID: PMC11960598 DOI: 10.1590/acb403125] [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: 11/27/2024] [Accepted: 02/08/2025] [Indexed: 04/04/2025] Open
Abstract
PURPOSE To conduct a systematic review of the mechanisms of photobiomodulation therapy (PBMT) for treating or preventing oral mucositis (OM) caused by antineoplastic therapy. METHODS Following PRISMA 2020 guidelines, a search was conducted in Medline, Latin American and Caribbean Health Sciences Literature (LILACS), Scientific Electronic Library Online (SciELO), and Bibliografia Brasileira de Odontologia from August to September 2023 using descriptors related to OM and laser therapy. Studies on the mechanisms of photobiomodulation in OM were included. Randomized (RCTs) or non-randomized trials from the past 10 years were reviewed. Risk of bias was assessed using RoB 2.0 and ROBINS-I tools. RESULTS A total of 355 studies was identified. After the screening, seven met the eligibility criteria. The RCTs showed a low risk of bias. PBMT reduced OM incidence in patients undergoing chemotherapy/radiotherapy. PBMT decreased pro-inflammatory cytokines (interleukin-6, tumor necrosis factor-α) and increased anti-inflammatory cytokines (interleukin-4, interleukin-10). It also modulated inflammatory mediators, enhancing the antioxidant enzyme superoxide dismutase and overexpressing genes for keratinocyte differentiation, aiding injury repair. CONCLUSION The findings suggested that the mechanism of action of PBMT in OM involves modulation of the inflammatory response, balancing oxygen reactive species generation, and expression of factors related to healing or repair. Further studies are needed to elucidate these mechanisms and optimize treatment protocols.
Collapse
Affiliation(s)
- Paulo Victor Alves de Sales
- Universidade Federal do Rio de Janeiro – Institute of Pharmaceutical Sciences – Multidisciplinary Center of Macaé – Macaé (RJ) – Brazil
| | - Isabella Piassi Dias Godói
- Universidade Federal do Rio de Janeiro – Institute of Pharmaceutical Sciences – Multidisciplinary Center of Macaé – Macaé (RJ) – Brazil
- Universidade Federal do Rio de Janeiro – Health Technology Assessment Center – Management, Economics, Health Education and Pharmaceutical Services – Rio de Janeiro (RJ) – Brazil
| | - Gerly Anne de Castro Brito
- Universidade Federal do Ceará – Faculty of Medicine – Post Graduate Program of Morphofunctional Sciences – Fortaleza (CE) – Brazil
| | - Renata Carvalho Leitão
- Universidade Federal do Ceará – Faculty of Medicine – Post Graduate Program of Morphofunctional Sciences – Fortaleza (CE) – Brazil
| | - Aurigena Antunes de Araújo
- Universidade Federal do Rio Grande do Norte – Department of Biophysics and Pharmacology – Post Graduate Program in Pharmaceutical Science – Natal (RN) – Brazil
- Universidade Federal do Rio Grande do Norte – Department of Biophysics and Pharmacology – Post Graduate Program Dental Sciences – Natal (RN) – Brazil
| | - Caroline Addison Carvalho Xavier de Medeiros
- Universidade Federal do Rio de Janeiro – Institute of Pharmaceutical Sciences – Multidisciplinary Center of Macaé – Macaé (RJ) – Brazil
- Universidade Federal do Rio Grande do Norte – Post Graduate Program Biotechnology, Northeast Biotechnology Network – Natal (RN) – Brazil
- Universidade Federal do Rio Grande do Norte – Post Graduate Program Biochemistry and Molecular Biology – Natal (RN) – Brazil
| |
Collapse
|
4
|
Zhou MY, Feng HY, Wang TT, Xu ZS, Gu SL, Li LL, Cai L, Li R. TLR3 as an emerging molecule facilitating pyroptosis in the context of rheumatoid arthritis: A study combined bioinformatics and experimental validation. Cytokine 2025; 187:156875. [PMID: 39884182 DOI: 10.1016/j.cyto.2025.156875] [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: 10/06/2024] [Revised: 01/03/2025] [Accepted: 01/24/2025] [Indexed: 02/01/2025]
Abstract
BACKGROUND Rheumatoid arthritis (RA) is an inflammatory disease of the joints mediated by immune cells. As an immune-related mode of cell death, pyroptosis has yet to be fully understood in RA. This research identified novel pyroptosis-related markers in RA and confirmed its functional significance in RA. METHODS Initially, crucial pyroptosis-related genes of RA were identified through GEO database, and biological pathways were determined through enrichment analysis. Then, PPI network, WGCNA and CIBERSORT analysis was utilized to screen hub genes and evaluate immune cell infiltration levels. Finally, validation experiments determined hub genes expression and regulatory roles in RA pathogenesis, and screened potential therapeutic drugs. RESULTS A total of 46 DEPRGs in RA were identified, which involved in NOD-like receptor and Toll-like receptor signaling pathway. Further screening revealed 3 crucial hub genes: CCL5, LY96, and TLR3 had significantly increased expression in RA synovial tissue and FLS, which might become diagnostic markers of RA. Analysis of immune infiltration revealed that hub genes exhibited associations with plasma cells, T lymphocytes, and macrophages. Further study on the crucial hub gene TLR3 revealed that knocking down TLR3 significantly inhibited the RA FLS hyperproliferation and pyroptosis, and dexamethasone and doxorubicin, as potential drugs, could treat RA by inhibiting TLR3. CONCLUSION Our study indicates that high expression of TLR3 promotes FLS pyroptosis and RA progression, suggesting its potential as both a biomarker and a therapeutic target for RA.
Collapse
Affiliation(s)
- Meng-Yuan Zhou
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei 230032, Anhui Province, China
| | - Hong-Yan Feng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei 230032, Anhui Province, China
| | - Tian-Tian Wang
- Department of Pathology, School of Basic Medicine, Anhui Medical University, Hefei 230032, Anhui Province, China
| | - Ze-Shan Xu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei 230032, Anhui Province, China
| | - Sheng-Long Gu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei 230032, Anhui Province, China
| | - Ling-Ling Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei 230032, Anhui Province, China
| | - Li Cai
- Department of Pathology, School of Basic Medicine, Anhui Medical University, Hefei 230032, Anhui Province, China.
| | - Rong Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei 230032, Anhui Province, China; Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei 230026, Anhui Province, China.
| |
Collapse
|
5
|
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
|
6
|
Lyu J, Liu Y, Liu F, Liu G, Gao Y, Wei R, Cai Y, Shen X, Zhao D, Zhao X, Xie Y, Yu H, Chai Y, Zhang J, Zhang Y, Xie Y. Therapeutic effect and mechanisms of traditional Chinese medicine compound (Qilong capsule) in the treatment of ischemic stroke. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155781. [PMID: 38870749 DOI: 10.1016/j.phymed.2024.155781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 05/20/2024] [Accepted: 05/26/2024] [Indexed: 06/15/2024]
Abstract
Background Qilong capsule (QLC) is a well-known traditional Chinese medicine compound extensively used in clinical practice. It has been approved by the China's FDA for the treatment of ischemic stroke (IS). In our clinical trial involving QLC (ClinicalTrials.gov identifier: NCT03174535), we observed the potential of QLC to improve neurological function in IS patients at the 24th week, while ensuring their safety. However, the effectiveness of QLC beyond the initial 12-week period remains uncertain, and the precise mechanisms underlying its action in IS have not been fully elucidated. Purpose In order to further explore the clinical efficacy of QLC in treating IS beyond the initial 12-week period and systematically elucidate its underlying mechanisms. Study Design This study employed an interdisciplinary integration strategy that combines post hoc analysis of clinical trials, transcriptome sequencing, integrated bioinformatics analysis, and animal experiments. Methods In this study, we conducted a post-hoc analysis with 2302 participants to evaluate the effectiveness of QLC at the 12th week. The primary outcome was the proportion of patients achieving functional independence at the 12th week, defined as a score of 0-2 on the modified Rankin Scale (mRS), which ranges from 0 (no symptoms) to 6 (death). Subsequently, we employed RNA sequencing (RNA-Seq) and quantitative reverse transcription polymerase chain reaction (RT-qPCR) techniques in the QLC trial to investigate the potential molecular mechanisms underlying the therapeutic effect of QLC in IS. Simultaneously, we utilized integrated bioinformatics analyses driven by external multi-source data and algorithms to further supplement the exploration and validation of QLC's therapeutic mechanism in treating IS. This encompassed network pharmacology analysis and analyses at the mRNA, cellular, and pathway levels focusing on core targets. Additionally, we developed a disease risk prediction model using machine learning. By identifying differentially expressed core genes (DECGs) between the normal and IS groups, we quantitatively predicted IS occurrence. Furthermore, to assess its protective effects and determine the key regulated pathway, we conducted experiments using a middle cerebral artery occlusion and reperfusion (MACO/R) rat model. Results Our findings demonstrated that the combination of QLC and conventional treatment (CT) significantly improved the proportion of patients achieving functional independence (mRS score 0-2) at the 12th week compared to CT alone (n = 2,302, 88.65 % vs 87.33 %, p = 0.3337; n = 600, 91.33 % vs 84.67 %, p = 0.0165). Transcriptome data revealed that the potential underlying mechanism of QLC for IS is related to the regulation of the NF-κB inflammatory pathway. The RT-qPCR results demonstrated that the regulatory trends of key genes, such as MD-2, were consistent with those observed in the RNA-Seq analysis. Integrated bioinformatics analysis elucidated that QLC regulates the NF-κB signaling pathway by identifying core targets, and machine learning was utilized to forecast the risk of IS onset. The MACO/R rat model experiment confirmed that QLC exerts its anti-CIRI effects by inhibiting the MD-2/TLR-4/NF-κB signaling axis. Conclusion: Our interdisciplinary integration study has demonstrated that the combination of QLC with CT exhibits significant superiority over CT alone in improving functional independence in patients at the 12th week. The potential mechanism underlying QLC's therapeutic effect in IS involves the inhibition of the MD-2/TLR4/NF-κB inflammatory signaling pathway, thereby attenuating cerebral ischemia/reperfusion inflammatory injury and facilitating neurofunctional recovery. The novelty and innovative potential of this study primarily lie in the novel finding that QLC significantly enhances the proportion of patients achieving functional independence (mRS score 0-2) at the 12th week. Furthermore, employing a "multilevel-multimethod" integrated research approach, we elucidated the potential mechanism underlying QLC's therapeutic effect in IS.
Collapse
Affiliation(s)
- Jian Lyu
- NMPA Key Laboratory for Clinical Research and Evaluation of Traditional Chinese Medicine & National Clinical Research Center for Chinese Medicine Cardiology, XiYuan Hospital, China Academy of Chinese Medical Sciences, No.1 Xiyuan playground Road, Haidian District, Beijing, 100091, PR China.
| | - Yi Liu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, No.16 Nanxiaojie, Inner Dongzhimen, Dongcheng District, Beijing, 100700, PR China
| | - Fumei Liu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, No.16 Nanxiaojie, Inner Dongzhimen, Dongcheng District, Beijing, 100700, PR China
| | - Guangyu Liu
- NMPA Key Laboratory for Clinical Research and Evaluation of Traditional Chinese Medicine & National Clinical Research Center for Chinese Medicine Cardiology, XiYuan Hospital, China Academy of Chinese Medical Sciences, No.1 Xiyuan playground Road, Haidian District, Beijing, 100091, PR China
| | - Yang Gao
- Dongfang Hospital, Beijing University of Chinese Medicine, No. 6 Fangxingyuan, Fengtai District, Beijing, 100078, PR China
| | - Ruili Wei
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, No.16 Nanxiaojie, Inner Dongzhimen, Dongcheng District, Beijing, 100700, PR China
| | - Yefeng Cai
- Guangdong Provincial Hospital of Traditional Chinese Medicine, No.111 Dade Road, Yuexiu District, Guangzhou, 510120, Guangdong, PR China
| | - Xiaoming Shen
- The First Affiliated Hospital of Henan University of Chinese Medicine, No.19 Renmin Road, Jinshui District, Zhengzhou, 450000, Henan, PR China
| | - Dexi Zhao
- Affiliated Hospital of Changchun University of Chinese Medicine, No.1478 Gongnong Road, Chaoyang District, Changchun, 130021, Jilin, PR China
| | - Xingquan Zhao
- Beijing Tiantan Hospital, Capital Medical University, No.119 South Fourth Ring West Road, Fengtai District, Beijing,100070, PR China
| | - Yingzhen Xie
- Dongzhimen Hospital, Beijing University of Chinese Medicine, No.5 Hai Yun Cang, Dongcheng District, Beijing,100700, PR China
| | - Haiqing Yu
- Taiyuan Chinese Medicine Hospital, No. 2 Baling South Street, Xinghualing District, Taiyuan 030009, Shanxi, PR China
| | - Yan Chai
- Department of Epidemiology, University of California, Los Angeles, 405 Hilgard Avenue, CA90095, USA
| | - Jingxiao Zhang
- Center for Applied Statistics, School of Statistics, Renmin University of China, 100872, Beijing, China
| | - Yunling Zhang
- NMPA Key Laboratory for Clinical Research and Evaluation of Traditional Chinese Medicine & National Clinical Research Center for Chinese Medicine Cardiology, XiYuan Hospital, China Academy of Chinese Medical Sciences, No.1 Xiyuan playground Road, Haidian District, Beijing, 100091, PR China.
| | - Yanming Xie
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, No.16 Nanxiaojie, Inner Dongzhimen, Dongcheng District, Beijing, 100700, PR China.
| |
Collapse
|
7
|
Wang C, Yang Y, Jiang C, Xi C, Yin Y, Wu H, Qian C. Exosomes Derived from hucMSCs Primed with IFN-γ Suppress the NF-κB Signal Pathway in LPS-Induced ALI by Modulating the miR-199b-5p/AFTPH Axis. Cell Biochem Biophys 2024; 82:647-658. [PMID: 38216808 DOI: 10.1007/s12013-023-01208-2] [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: 08/03/2023] [Accepted: 11/30/2023] [Indexed: 01/14/2024]
Abstract
Exosomes (exos) are primarily responsible for the process of mesenchymal stem cells (MSCs) treatment for acute lung injury (ALI), but the mechanism remains unclear, particularly in altered microenvironment. Therefore, this study aimed to investigate the potential mechanism of exos derived from human umbilical cord mesenchymal stem cells (hucMSCs) primed with interferon-gamma (IFN-γ) on ALI and to propose a promising and cell-free strategy. This study extracted exos from hucMSCs supernatant primed and unprimed with IFN-γ marked with IFN-γ-exos and CON-exos, which were identified and traced. IFN-γ-exos administration to ALI models suppressed the NF-κB signaling pathway compared to CON-exos, which were quantified through western blot and immunohistochemical staining. Reverse transcription-quantitative polymerase chain reaction validated miR-199b-5p expression in the IFN-γ-exos and CON-exos treatment groups. Data analysis, a dual-luciferase reporter assay, and cell transfection were conducted to investigate the target binding between miR-199b-5p and Aftiphilin (AFTPH), with AFTPH expression analyzed via cell immunofluorescence and western blot. Co-immunoprecipitation was conducted for the interaction between AFTPH and NF-κB p65. The result revealed that miR-199b-5p was down-regulated in the IFN-γ-exos treatment group, which had a target binding site with AFTPH, and an interaction with NF-κB p65. Consequently, IFN-γ-exos inhibited the NF-κB signaling pathway in ALI in vitro and in vivo through the miR-199b-5p/AFTPH axis. Our results demonstrated new directions of novel and targeted treatment for ALI.
Collapse
Affiliation(s)
- Chun Wang
- Kunming Medical University, Kunming, Yunnan, China
- Department of Emergency Intensive Care Unit, Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Yiran Yang
- Kunming Medical University, Kunming, Yunnan, China
| | - Chen Jiang
- Kunming Medical University, Kunming, Yunnan, China
| | - Cheng Xi
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Yunxiang Yin
- Department of Emergency Intensive Care Unit, Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Haiying Wu
- Department of Emergency, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China.
| | - Chuanyun Qian
- Department of Emergency, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China.
| |
Collapse
|
8
|
Liu L, Lin L, Wang Y, Yan X, Li R, He M, Li H, Zhuo C, Li L, Zhang D, Wang X, Huang W, Li X, Mao Y, Chen H, Wu S, Jiang W, Zhu L. L-AP Alleviates Liver Injury in Septic Mice by Inhibiting Macrophage Activation via Suppressing NF-κB and NLRP3 Inflammasome/Caspase-1 Signal Pathways. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:8460-8475. [PMID: 38564364 DOI: 10.1021/acs.jafc.3c02781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Liver injury and progressive liver failure are severe life-threatening complications in sepsis, further worsening the disease and leading to death. Macrophages and their mediated inflammatory cytokine storm are critical regulators in the occurrence and progression of liver injury in sepsis, for which effective treatments are still lacking. l-Ascorbic acid 6-palmitate (L-AP), a food additive, can inhibit neuroinflammation by modulating the phenotype of the microglia, but its pharmacological action in septic liver damage has not been fully explored. We aimed to investigate L-AP's antisepticemia action and the possible pharmacological mechanisms in attenuating septic liver damage by modulating macrophage function. We observed that L-AP treatment significantly increased survival in cecal ligation and puncture-induced WT mice and attenuated hepatic inflammatory injury, including the histopathology of the liver tissues, hepatocyte apoptosis, and the liver enzyme levels in plasma, which were comparable to NLRP3-deficiency in septic mice. L-AP supplementation significantly attenuated the excessive inflammatory response in hepatic tissues of septic mice in vivo and in cultured macrophages challenged by both LPS and ATP in vitro, by reducing the levels of NLRP3, pro-IL-1β, and pro-IL-18 mRNA expression, as well as the levels of proteins for p-I-κB-α, p-NF-κB-p65, NLRP3, cleaved-caspase-1, IL-1β, and IL-18. Additionally, it impaired the inflammasome ASC spot activation and reduced the inflammatory factor contents, including IL-1β and IL-18 in plasma/cultured superannuants. It also prevented the infiltration/migration of macrophages and their M1-like inflammatory polarization while improving their M2-like polarization. Overall, our findings revealed that L-AP protected against sepsis by reducing macrophage activation and inflammatory cytokine production by suppressing their activation in NF-κB and NLRP3 inflammasome signal pathways in septic liver.
Collapse
Affiliation(s)
- Linling Liu
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, and Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Lan Lin
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, and Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Yingling Wang
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, and Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Xin Yan
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, and Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, PR China
- Department of Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Ruli Li
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, and Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Min He
- Department of Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - He Li
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, and Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Caili Zhuo
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, and Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Lingyu Li
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, and Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Die Zhang
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, and Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Xuemei Wang
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, and Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Wenjing Huang
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, and Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Xinyue Li
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, and Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Yan Mao
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, and Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Hongying Chen
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, and Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, PR China
- Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Sisi Wu
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, and Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, PR China
- Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Wei Jiang
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, and Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Ling Zhu
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, and Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, PR China
| |
Collapse
|
9
|
Sung B, Hwang D, Baek A, Yang B, Lee S, Park J, Kim E, Kim M, Lee E, Chang Y. Gadolinium-Based Magnetic Resonance Theranostic Agent with Gallic Acid as an Anti-Neuroinflammatory and Antioxidant Agent. Antioxidants (Basel) 2024; 13:204. [PMID: 38397802 PMCID: PMC10885874 DOI: 10.3390/antiox13020204] [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: 11/29/2023] [Revised: 01/29/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
Studies in the field have actively pursued the incorporation of diverse biological functionalities into gadolinium-based contrast agents, aiming at the amalgamation of MRI imaging and therapeutic capabilities. In this research, we present the development of Gd-Ga, an anti-neuroinflammatory MR contrast agent strategically designed to target inflammatory mediators for comprehensive imaging diagnosis and targeted lesion treatment. Gd-Ga is a gadolinium complex composed of 1,4,7-tris(carboxymethylaza)cyclododecane-10-azaacetylamide (DO3A) conjugated with gallic acid (3,4,5-trihydroxybenzoic acid). Upon intravenous administration in LPS-induced mouse models, Gd-Ga demonstrated a remarkable three-fold increase in signal-to-noise (SNR) variation compared to Gd-DOTA, particularly evident in both the cortex and hippocampus 30 min post-MR monitoring. In-depth investigations, both in vitro and in vivo, into the anti-neuroinflammatory properties of Gd-Ga revealed significantly reduced protein expression levels of pro-inflammatory mediators compared to the LPS group. The alignment between in silico predictions and phantom studies indicates that Gd-Ga acts as an anti-neuroinflammatory agent by directly binding to MD2. Additionally, the robust antioxidant activity of Gd-Ga was confirmed by its effective scavenging of NO and ROS. Our collective findings emphasize the immense potential of this theranostic complex, where a polyphenol serves as an anti-inflammatory drug, presenting an exceptionally efficient platform for the diagnosis and treatment of neuroinflammation.
Collapse
Affiliation(s)
- Bokyung Sung
- ICT Convergence Research Center, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea;
| | - Dongwook Hwang
- Department of Biomedical Science, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu 41944, Republic of Korea;
- Theranocure Co., Ltd., 90 Chilgokjungang-daero 136-gil, Buk-gu, Daegu 41405, Republic of Korea; (B.Y.); (S.L.)
| | - Ahrum Baek
- Institute of Biomedical Engineering Research, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu 41944, Republic of Korea; (A.B.); (E.L.)
| | - Byeongwoo Yang
- Theranocure Co., Ltd., 90 Chilgokjungang-daero 136-gil, Buk-gu, Daegu 41405, Republic of Korea; (B.Y.); (S.L.)
- Department of Medical & Biological Engineering, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea
| | - Sangyun Lee
- Theranocure Co., Ltd., 90 Chilgokjungang-daero 136-gil, Buk-gu, Daegu 41405, Republic of Korea; (B.Y.); (S.L.)
- Department of Medical & Biological Engineering, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea
| | - Jangwoo Park
- Korea Radioisotope Center for Pharmaceuticals, Korea Institute of Radiological & Medical Sciences, Seoul 01812, Republic of Korea; (J.P.); (E.K.)
| | - Eunji Kim
- Korea Radioisotope Center for Pharmaceuticals, Korea Institute of Radiological & Medical Sciences, Seoul 01812, Republic of Korea; (J.P.); (E.K.)
- Center for Data Analytics Innovation, Office of National R&D Evaluation and Analysis, Korea Institute of S&T Evaluation and Planning, 1339, Wonjung-ro, Maengdong-myeon, Eumseong-gun 27740, Republic of Korea
| | - Minsup Kim
- TARS Scientific, Nowon-gu, Seoul 01662, Republic of Korea;
| | - Eunshil Lee
- Institute of Biomedical Engineering Research, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu 41944, Republic of Korea; (A.B.); (E.L.)
| | - Yongmin Chang
- Department of Biomedical Science, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu 41944, Republic of Korea;
- Institute of Biomedical Engineering Research, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu 41944, Republic of Korea; (A.B.); (E.L.)
- Department of Medical & Biological Engineering, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea
- Department of Molecular Medicine, School of Medicine, Kyungpook National University, 680 Guchaebosang-ro, Jung-gu, Daegu 41944, Republic of Korea
- Department of Radiology, Kyungpook National University Hospital, 130 Dongdeok-ro, Jung-gu, Daegu 41944, Republic of Korea
| |
Collapse
|
10
|
Chen J, Liu Z, Sun H, Liu M, Wang J, Zheng C, Cao X. MiR-203a-3p attenuates apoptosis and pyroptosis of chondrocytes by regulating the MYD88/NF-κB pathway to alleviate osteoarthritis progression. Aging (Albany NY) 2023; 15:14457-14472. [PMID: 38095638 PMCID: PMC10756106 DOI: 10.18632/aging.205373] [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: 08/16/2023] [Accepted: 11/20/2023] [Indexed: 12/21/2023]
Abstract
BACKGROUND Osteoarthritis (OA) is a degenerative joint disease that imposes a significant socioeconomic burden worldwide. Our previous studies revealed a down-regulation of miR-203a-3p in the knee tissues of OA patients. However, the underlying mechanism through which miR-203a-3p mediates the pathological process of OA remains unknown. Thus, we aimed to determine the effects of miR-203a-3p in the progression of OA. METHODS Rat primary chondrocytes were stimulated with 10 μg/mL lipopolysaccharide (LPS) for 24 hours, followed by transfection with 50 nM miR-203a-3p mimic, inhibitor, and siRNA for MYD88 or consistent negative controls for 48 hours. To evaluate the effects of miR-203a-3p on cartilage matrix degradation, oxidative stress, apoptosis, and pyroptosis in chondrocytes, various techniques such as immunofluorescence staining, biochemical analysis, Western blotting, and the TUNEL staining were utilized. In the rat OA model, all rats were randomly divided into four groups: Sham, OA, OA+Agomir negative control (NC), and OA+Agomir. They received intra-articular injections of 25 nmol miR-203a-3p agomir, agomir NC, or normal saline twice a week for the duration of 8 weeks after OA induction. Immunofluorescence staining was performed to evaluate the effects of miR-203a-3p on cartilage matrix degradation in rats. RESULTS MiR-203a-3p was down-regulated in LPS-treated rat chondrocytes and OA cartilage, and directly targeted MYD88. Moreover, miR-203a-3p significantly inhibited LPS-induced cartilage matrix degradation, oxidative stress, apoptosis, and pyroptosis of chondrocytes via targeting MYD88. Mechanistically, miR-203a-3p exerted protective effects via the inhibition of the MYD88/NF-κB pathway. In the rat OA model, intra-articular injections of miR-203a-3p agomir also significantly inhibited cartilage matrix degradation, thereby alleviating OA progression. Furthermore, the miR-203a-3p agomir-treated arthritic rat dramatically exhibited better articular tissue morphology and lower OARSI scores. CONCLUSIONS MiR-203a-3p plays a role in alleviating the progression of OA by regulating the MYD88/NF-κB pathway, thereby inhibiting cartilage matrix degradation, oxidative stress, apoptosis, and pyroptosis of chondrocytes. It highlights the potential significance of miR-203a-3p as an important regulator of OA.
Collapse
Affiliation(s)
- Jiayi Chen
- Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine, Zhongshan 528401, Guangdong, China
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510120, Guangdong China
| | - Zhutong Liu
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510120, Guangdong China
| | - He Sun
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510120, Guangdong China
| | - Mange Liu
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510120, Guangdong China
| | - Jiangliang Wang
- Liuyang Hospital of Traditional Chinese Medicine, Liuyang 410300, Hunan, China
| | - Chenxiao Zheng
- Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine, Zhongshan 528401, Guangdong, China
| | - Xuewei Cao
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510120, Guangdong China
| |
Collapse
|
11
|
Zheng Z, Zhou Y, Song Y, Ying P, Tan X. Genetic and immunological insights into COVID-19 with acute myocardial infarction: integrated analysis of mendelian randomization, transcriptomics, and clinical samples. Front Immunol 2023; 14:1286087. [PMID: 38022594 PMCID: PMC10657900 DOI: 10.3389/fimmu.2023.1286087] [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: 08/31/2023] [Accepted: 10/19/2023] [Indexed: 12/01/2023] Open
Abstract
Background Globally, most deaths result from cardiovascular diseases, particularly ischemic heart disease. COVID-19 affects the heart, worsening existing heart conditions and causing myocardial injury. The mechanistic link between COVID-19 and acute myocardial infarction (AMI) is still being investigated to elucidate the underlying molecular perspectives. Methods Genetic risk assessment was conducted using two-sample Mendelian randomization (TSMR) to determine the causality between COVID-19 and AMI. Weighted gene co-expression network analysis (WGCNA) and machine learning were used to discover and validate shared hub genes for the two diseases using bulk RNA sequencing (RNA-seq) datasets. Additionally, gene set enrichment analysis (GSEA) and single-cell RNA-seq (scRNA-seq) analyses were performed to characterize immune cell infiltration, communication, and immune correlation of the hub genes. To validate the findings, the expression patterns of hub genes were confirmed in clinical blood samples collected from COVID-19 patients with AMI. Results TSMR did not find evidence supporting a causal association between COVID-19 or severe COVID-19 and AMI. In the bulk RNA-seq discovery cohorts for both COVID-19 and AMI, WGCNA's intersection analysis and machine learning identified TLR4 and ABCA1 as significant hub genes, demonstrating high diagnostic and predictive value in the RNA-seq validation cohort. Single-gene GSEA and single-sample GSEA (ssGSEA) revealed immune and inflammatory roles for TLR4 and ABCA1, linked to various immune cell infiltrations. Furthermore, scRNA-seq analysis unveiled significant immune dysregulation in COVID-19 patients, characterized by altered immune cell proportions, phenotypic shifts, enhanced cell-cell communication, and elevated TLR4 and ABCA1 in CD16 monocytes. Lastly, the increased expression of TLR4, but not ABCA1, was validated in clinical blood samples from COVID-19 patients with AMI. Conclusion No genetic causal link between COVID-19 and AMI and dysregulated TLR4 and ABCA1 may be responsible for the development of immune and inflammatory responses in COVID-19 patients with AMI.
Collapse
Affiliation(s)
- Zequn Zheng
- Department of Cardiology, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
- Clinical Research Center, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Yueran Zhou
- Department of Cardiology, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
- Clinical Research Center, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Yongfei Song
- Ningbo Institute for Medicine &Biomedical Engineering Combined Innovation, Ningbo, Zhejiang, China
| | - Pengxiang Ying
- Department of Cardiology, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
- Centre for Precision Health, Edith Cowan University, Perth, WA, Australia
| | - Xuerui Tan
- Department of Cardiology, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
- Clinical Research Center, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| |
Collapse
|
12
|
He X, Bai Q, Zhang X, Zhang L. MgCl 2 Attenuates ox-LDL-Induced Vascular Smooth Muscle-Derived Foam Cells Pyroptosis by Downregulating the TLR4/NF-κB Signaling Pathway. Biol Trace Elem Res 2023; 201:5242-5256. [PMID: 36719541 DOI: 10.1007/s12011-023-03585-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 01/24/2023] [Indexed: 02/01/2023]
Abstract
Pyroptosis is a type of programmed cell death that is generally upregulated during atherosclerosis (AS). Magnesium, an important cation in the body, has exhibited an antiatherosclerotic effect. We collected AS model datasets from the Gene Expression Omnibus (GEO) and explored the correlation between pyroptosis and AS through a series of bioinformatics methods. We next investigated the impact of oxidized low-density lipoprotein (ox-LDL) on primary cultured vascular smooth muscle cells (VSMCs) foaminess and pyroptosis. Finally, foam cells were preconditioned with different concentrations of MgCl2 to explore its influence on ox-LDL-induced VSMCs pyroptosis. NLRP3-mediated pyroptosis plays a core role in regulating AS progression as shown by bioinformatic analysis. Ox-LDL (50/75/100 mg/L) increased CE/TE ratio (> 50%) in VSMCs and prompted VSMC-derived foam cell formation, and (75/100 mg/L) ox-LDL-induced pyroptosis. Compared to 1 mmol/L MgCl2, 10 mmol/L MgCl2 significantly downregulated the expression of pyroptosis related molecules in VSMCs induced by 75 mg/L ox-LDL, including NLRP3, ASC, caspase-1, and GSDMD. The secretion of IL-1β, IL-18, and LDH was also inhibited by MgCl2. According to CCK-8 and Hoechst 33,342/PI staining, the damage to VSMCs viability induced by ox-LDL was ameliorated by MgCl2. In addition, MgCl2 attenuated the upregulation of TLR4, IKKβ, and p65 and the downregulation of IκBα in VSMCs induced by ox-LDL. The present study demonstrated that pyroptosis-related genes were the core genes in AS. We also revealed the effect and underlying mechanism of MgCl2 on ox-LDL-induced VSMCs pyroptosis, suggesting that MgCl2 has promising clinical applications for AS pyroptosis prevention and treatment.
Collapse
Affiliation(s)
- Xiao He
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, 23 You Zheng Street, Harbin, 150001, Heilongjiang Province, China
| | - Qingquan Bai
- Department of Hepatology & Gastroenterology, Charité University Medical Center, Augustenburger Pl. 1, 13353, Berlin, Germany
| | - Xiaosi Zhang
- Metro-Medic Clinic, 1538 Sherbrooke Ouest, Suite 100, Montreal, QC, H3G 1L5, Canada.
| | - Liming Zhang
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, 23 You Zheng Street, Harbin, 150001, Heilongjiang Province, China.
| |
Collapse
|
13
|
Mohamed HA, Abdelkafy AE, Khairy RMM, Abdelraheim SR, Kamel BA, Marey H. MicroRNAs and cytokines as potential predictive biomarkers for COVID-19 disease progression. Sci Rep 2023; 13:3531. [PMID: 36864077 PMCID: PMC9979137 DOI: 10.1038/s41598-023-30474-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 02/23/2023] [Indexed: 03/04/2023] Open
Abstract
Host microRNAs can influence the cytokine storm associated SARS-CoV-2 infection and proposed as biomarkers for COVID-19 disease. In the present study, serum MiRNA-106a and miRNA-20a were quantified by real time-PCR in 50 COVID-19 patients hospitalized at Minia university hospital and 30 healthy volunteers. Profiles of serum inflammatory cytokines (TNF-α, IFN-γ, and IL-10) and TLR4 were analyzed by Eliza in patients and controls. A highly significant decrease (P value = 0.0001) in the expressions of miRNA-106a and miRNA-20a was reported in COVID-19 patients compared to controls. A significant decrease in the levels of miRNA-20a was also reported in patients with lymphopenia, patients having chest CT severity score (CSS) > 19 and in patients having O2 saturation less than 90%. Significantly higher levels of TNF-α, IFN-γ, IL-10 and TLR4 were reported in patients compared to controls. IL-10 and TLR4 levels were significantly higher in patients having lymphopenia. TLR-4 level was higher in patients with CSS > 19 and in patients with hypoxia. Using univariate logistic regression analysis, miRNA-106a, miRNA-20a, TNF-α, IFN-γ, IL-10 and TLR4 were identified as good predictors of disease. Receiver operating curve showed that the downregulation of miRNA-20a in patients having lymphopenia, patients with CSS > 19 and patients with hypoxia could be a potential biomarker with AUC = 0.68 ± 0.08, AUC = 0.73 ± 0.07 and AUC = 0.68 ± 0.07 respectively. Also, ROC curve showed accurate association between the increase of serum IL-10 and TLR-4 and lymphopenia among COVID-19 patients with AUC = 0.66 ± 0.08 and AUC = 0.73 ± 0.07 respectively. ROC curve showed also that serum TLR-4 could be a potential marker for high CSS with AUC = 0.78 ± 0.06. A negative correlation was detected between miRNA-20a with TLR-4 (r = - 0.30, P value = 0.03). We concluded that, miR-20a, is a potential biomarker of COVID-19 severity and blockade of IL-10 and TLR4 may constitute a novel therapy for COVID-19 patients.
Collapse
Affiliation(s)
- Hatem A. Mohamed
- grid.411806.a0000 0000 8999 4945Department of Biochemistry, Faculty of Medicine, Minia University, Minia, Egypt
| | - Aya Eid Abdelkafy
- grid.411806.a0000 0000 8999 4945Department of Biochemistry, Faculty of Medicine, Minia University, Minia, Egypt
| | - Rasha M. M. Khairy
- grid.411806.a0000 0000 8999 4945Department of Microbiology and Immunology, Faculty of Medicine, Minia University, Minia, 61511 Egypt
| | - Salama R. Abdelraheim
- grid.411806.a0000 0000 8999 4945Department of Biochemistry, Faculty of Medicine, Minia University, Minia, Egypt
| | - Bothina Ahmed Kamel
- grid.411806.a0000 0000 8999 4945Department of Biochemistry, Faculty of Medicine, Minia University, Minia, Egypt
| | - Heba Marey
- grid.411806.a0000 0000 8999 4945Department of Biochemistry, Faculty of Medicine, Minia University, Minia, Egypt
| |
Collapse
|
14
|
Zhao Y, Liu P, Luan H, Jiang H, Xu Y, Zhang Y, Zhang Y, Li R. Demethyleneberberine alleviated the inflammatory response by targeting MD-2 to inhibit the TLR4 signaling. Front Immunol 2023; 14:1130404. [PMID: 37168866 PMCID: PMC10165096 DOI: 10.3389/fimmu.2023.1130404] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 03/30/2023] [Indexed: 05/13/2023] Open
Abstract
Introduction The colitis induced by trinitrobenzenesulfonic acid (TNBS) is a chronic and systemic inflammatory disease that leads to intestinal barrier dysfunction and autoimmunedisorders. However, the existing treatments of colitis are associated with poor outcomes, and the current strategies remain deep and long-time remission and the prevention of complications. Recently, demethyleneberberine (DMB) has been reported to be a potential candidate for the treatment of inflammatory response that relied on multiple pharmacological activities, including anti-oxidation and antiinflammation. However, the target and potential mechanism of DMB in inflammatory response have not been fully elucidated. Methods This study employed a TNBS-induced colitis model and acute sepsis mice to screen and identify the potential targets and molecular mechanisms of DMB in vitro and in vivo. The purity and structure of DMB were quantitatively analyzed by high-performance liquid chromatography (HPLC), mass spectrometry (MS), Hydrogen nuclear magnetic resonance spectroscopy (1H-NMR), and infrared spectroscopy (IR), respectively. The rats were induced by a rubber hose inserted approximately 8 cm through their anus to be injected with TNBS. Acute sepsis was induced by injection with LPS via the tail vein for 60 h. These animals with inflammation were orally administrated with DMB, berberine (BBR), or curcumin (Curc), respectively. The eukaryotic and prokaryotic expression system of myeloid differentiation protein-2 (MD-2) and its mutants were used to evaluate the target of DMB in inflammatory response. Resluts DMB had two free phenolic hydroxyl groups, and the purity exceeded 99% in HPLC. DMB alleviated colitis and suppressed the activation of TLR4 signaling in TNBS-induced colitis rats and LPS-induced RAW264.7 cells. DMB significantly blocked TLR4 signaling in both an MyD88-dependent and an MyD88-independent manner by embedding into the hydrophobic pocket of the MD-2 protein with non-covalent bonding to phenylalanine at position 76 in a pi-pi T-shaped interaction. DMB rescued mice from sepsis shock induced by LPS through targeting the TLR4-MD-2 complex. Conclusion Taken together, DMB is a promising inhibitor of the MD-2 protein to suppress the hyperactivated TLR4 signaling in inflammatory response.
Collapse
Affiliation(s)
- Yaxing Zhao
- State Key Laboratory of Natural Medicines, Department of Biochemistry, China Pharmaceutical University, Nanjing, China
- Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Peng Liu
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Pharmaceutical Sciences, Jiangnan University, Wuxi, China
| | - Haofan Luan
- State Key Laboratory of Natural Medicines, Department of Biochemistry, China Pharmaceutical University, Nanjing, China
| | - Hua Jiang
- State Key Laboratory of Natural Medicines, Department of Biochemistry, China Pharmaceutical University, Nanjing, China
| | - Yingmei Xu
- State Key Laboratory of Natural Medicines, Department of Biochemistry, China Pharmaceutical University, Nanjing, China
| | - Yuanqiang Zhang
- State Key Laboratory of Natural Medicines, Department of Biochemistry, China Pharmaceutical University, Nanjing, China
| | - Yubin Zhang
- State Key Laboratory of Natural Medicines, Department of Biochemistry, China Pharmaceutical University, Nanjing, China
| | - Ruiyan Li
- State Key Laboratory of Natural Medicines, Department of Biochemistry, China Pharmaceutical University, Nanjing, China
- *Correspondence: Ruiyan Li,
| |
Collapse
|
15
|
Li W, Huang Q, Yu J, Yang Y, Yu J, Liu Y, Song H, Cui L, Niu X. Schisandrin improves lipopolysaccharide‐induced acute lung injury by inhibiting the inflammatory response in vivo and in vitro. J Food Biochem 2022; 46:e14141. [DOI: 10.1111/jfbc.14141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 02/28/2022] [Accepted: 03/03/2022] [Indexed: 11/27/2022]
Affiliation(s)
- Weifeng Li
- School of Pharmacy Xi'an Jiaotong University Xi'an China
| | - Qiuxia Huang
- School of Pharmacy Xi'an Jiaotong University Xi'an China
| | - Jinjin Yu
- School of Pharmacy Xi'an Jiaotong University Xi'an China
| | - Yajie Yang
- School of Pharmacy Xi'an Jiaotong University Xi'an China
| | - Jiabao Yu
- School of Pharmacy Xi'an Jiaotong University Xi'an China
| | - Yang Liu
- School of Pharmacy Xi'an Jiaotong University Xi'an China
| | - Huixin Song
- School of Pharmacy Xi'an Jiaotong University Xi'an China
| | - Langjun Cui
- School of Life Sciences Shaanxi Normal University Xi'an China
| | - Xiaofeng Niu
- School of Pharmacy Xi'an Jiaotong University Xi'an China
| |
Collapse
|
16
|
Xiong Y, Yang J, Tong H, Zhu C, Pang Y. HMGB1 augments cognitive impairment in sepsis-associated encephalopathy by binding to MD-2 and promoting NLRP3-induced neuroinflammation. Psychogeriatrics 2022; 22:167-179. [PMID: 34931753 DOI: 10.1111/psyg.12794] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 11/07/2021] [Accepted: 11/23/2021] [Indexed: 12/28/2022]
Abstract
BACKGROUND Sepsis-associated encephalopathy (SAE) always manifests with severe inflammatory symptoms and cognitive impairment. High mobility group box 1 (HMGB1) is a pro-inflammatory cytokine. In this study we investigated the role of HMGB1 in SAE. METHODS An SAE mouse model was established through cecal ligation and puncture surgery and then injected with adenovirus short hairpin RNA (Ad-sh)-HMGB1 or Ad-sh-myeloid differentiation protein (MD-2). The cognitive impairment and pathological injury in mice of different groups were evaluated using the Morris water maze experiment, Y-maze test, tail suspension test, fear conditioning test, and haematoxylin-eosin staining. The expressions of HMGB1 (fully reduced and disulfide (ds)HMGB1), MD-2, and NLRP3 in SAE mice were determined. Then, levels of inflammatory cytokines were measured. The binding relation between HMGB1 and MD-2 was predicted and certified. Additionally, MD-2 was downregulated to verify the role of the binding of HMGB1 and MD-2 in neuroinflammation and cognitive impairment in SAE. RESULTS Expressions of HMGB1, MD-2, NLRP3, and inflammatory cytokines were enhanced in the SAE mouse model, which were in parallel with impaired cognitive function. HMGB1 silencing resulted in downregulated NLRP3 expression and alleviated neuroinflammation and cognitive impairment in SAE mice. Mechanically, dsHMGB1 bound to MD-2 to activate NLRP3, thereby exacerbating neuroinflammation and cognitive impairment in SAE mice. The limited binding of HMGB1 and MD-2 downregulated NLRP3 expression to alleviate neuroinflammation and cognitive impairment in SAE mice. CONCLUSION HMGB1 was overexpressed in SAE, and dsHMGB1 bound to MD-2 to activate NLRP3 inflammasome, inducing neuroinflammation and cognitive impairment in SAE.
Collapse
Affiliation(s)
- Yanan Xiong
- Department of Emergency, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, P.R. China
| | - Jilin Yang
- Department of Emergency, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, P.R. China
| | - Haiyang Tong
- Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, P.R. China
| | - Chenting Zhu
- Department of Emergency, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, P.R. China
| | - Yinhu Pang
- Department of Emergency, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, P.R. China
| |
Collapse
|
17
|
Wei Y, Yang L, Pandeya A, Cui J, Zhang Y, Li Z. Pyroptosis-Induced Inflammation and Tissue Damage. J Mol Biol 2022; 434:167301. [PMID: 34653436 PMCID: PMC8844146 DOI: 10.1016/j.jmb.2021.167301] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 09/23/2021] [Accepted: 10/05/2021] [Indexed: 02/07/2023]
Abstract
Programmed cell deaths are pathways involving cells playing an active role in their own destruction. Depending on the signaling system of the process, programmed cell death can be divided into two categories, pro-inflammatory and non-inflammatory. Pyroptosis is a pro-inflammatory form of programmed cell death. Upon cell death, a plethora of cytokines are released and trigger a cascade of responses from the neighboring cells. The pyroptosis process is a double-edged sword, could be both beneficial and detrimental in various inflammatory disorders and disease conditions. A physiological outcome of these responses is tissue damage, and sometimes death of the host. In this review, we focus on the inflammatory response triggered by pyroptosis, and resulting tissue damage in selected organs.
Collapse
Affiliation(s)
- Yinan Wei
- Department of Chemistry, College of Arts and Sciences, University of Kentucky, Lexington, KY, USA.
| | - Ling Yang
- Department of Chemistry, College of Arts and Sciences, University of Kentucky, Lexington, KY, USA
| | - Ankit Pandeya
- Department of Chemistry, College of Arts and Sciences, University of Kentucky, Lexington, KY, USA
| | - Jian Cui
- Department of Chemistry, College of Arts and Sciences, University of Kentucky, Lexington, KY, USA
| | - Yan Zhang
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky, Lexington, KY, USA.,Department of Oncology, the First Affiliated Hospital of Soochow University, Suzhou,China
| | - Zhenyu Li
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
18
|
Pandur E, Balatinácz A, Micalizzi G, Mondello L, Horváth A, Sipos K, Horváth G. Anti-inflammatory effect of lavender (Lavandula angustifolia Mill.) essential oil prepared during different plant phenophases on THP-1 macrophages. BMC Complement Med Ther 2021; 21:287. [PMID: 34819075 PMCID: PMC8611982 DOI: 10.1186/s12906-021-03461-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 11/02/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Pseudomonas aeruginosa is the most common Gram-negative bacterium associated with nosocomial respiratory infections. Lavender essential oil is mainly used in aromatherapy, but it has several pharmacological and therapeutic properties. Furthermore, it possesses antifungal and antibacterial activities. The anti-inflammatory activity of essential oils may depend on the composition and the ratio of the compounds. The constitution of the essential oils extracted from the different stages of flowering period varies, which makes it plausible that the collection time of the flowers influences the anti-inflammatory effects. Different types of essential oils reduce inflammation acting similarly by modulating the activity and action of the NFκB signalling pathway, which is the major regulator of the transcription of pro-inflammatory cytokines. METHODS Lavender essential oils were distilled from lavender plant cultivated in Hungary and the flowers were harvested at the beginning and at the end of flowering period. The experiments were carried out on THP-1 human monocyte/macrophage cell line as in vitro cell culture model for monitoring the effects of lavender essential oils and the main compound linalool on P. aeruginosa LPS stimulated inflammation. The mRNA and protein levels of four pro-inflammatory cytokines, IL-6, IL-1β, IL-8 and TNFα were determined by Real Time PCR and ELISA measurements. The effects of essential oils were compared to the response to two NFκB inhibitors, luteolin and ACHP. RESULTS Linalool and lavender essential oil extracted from plants at the beginning of flowering period were successful in decreasing pro-inflammatory cytokine production following LPS pretreatment. In case of IL-8 and IL-1β lavender oil showed stronger effect compared to linalool and both of them acted similarly to NFκB inhibitors. Pretreatments with linalool and lavender essential oil/beginning of flowering period prevented pro-inflammatory cytokine production compared to LPS treatment alone. Although lavender essential oil/end of flowering period decreased IL-6, IL-1β and IL-8 mRNA expression in case of LPS pretreatment, it was not capable to reduce cytokine secretion. CONCLUSION Based on our results it has been proven that lavender essential oil extracted at the beginning of flowering period is a potent inhibitor of the synthesis of four pro-inflammatory cytokines IL-6, IL-8, IL-β and TNFα of THP-1 cells. This supports the relevance of the collection of the lavender flowers from early blooming period for essential oil production and for the utilization as an anti-inflammatory treatment.
Collapse
Affiliation(s)
- Edina Pandur
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Pécs, H-7624, Rókus u. 2, Pécs, Hungary
| | - Alex Balatinácz
- Department of Pharmacognosy, Faculty of Pharmacy, University of Pécs, H-7624, Rókus u. 2, Pécs, Hungary
| | - Giuseppe Micalizzi
- Chromaleont s.r.l., c/o Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98168, Messina, Italy
| | - Luigi Mondello
- Chromaleont s.r.l., c/o Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98168, Messina, Italy.,Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98168, Messina, Italy.,Unit of Food Science and Nutrition, Department of Medicine, University Campus Bio-Medico of Rome, 00128, Rome, Italy
| | - Adrienn Horváth
- Department of Pharmacognosy, Faculty of Pharmacy, University of Pécs, H-7624, Rókus u. 2, Pécs, Hungary
| | - Katalin Sipos
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Pécs, H-7624, Rókus u. 2, Pécs, Hungary
| | - Györgyi Horváth
- Department of Pharmacognosy, Faculty of Pharmacy, University of Pécs, H-7624, Rókus u. 2, Pécs, Hungary.
| |
Collapse
|
19
|
Silencing ROCK1 ameliorates ventilator-induced lung injury in mice by inhibiting macrophages' NLRP3 signaling. Int Immunopharmacol 2021; 101:108208. [PMID: 34619496 DOI: 10.1016/j.intimp.2021.108208] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 09/17/2021] [Accepted: 09/27/2021] [Indexed: 01/19/2023]
Abstract
Rho kinase, including two subtypes, ROCK1 and ROCK2, controls a variety of biological processes helping coordinate the tissues response to stress and injury. Some authors believe that alveolar macrophages (AMs) play a key role in the early phase of ventilator-induced lung injury (VILI), which is closely related to the activation of NLRP3 inflammasome and NF-κB signaling. However, there is currently little known about the relationship between ROCK signaling and NLRP3 inflammasome. Accordingly, we focused on exploring the effect of ROCK for NLRP3 inflammasome, the results showed that VILI in C57BL/6 mice significantly increased NF-κB, NLRP3, ASC, caspase1 expression, and the secretion of cytokines, which was reversed by applying the ROCK Inhibitor-Y27632. Moreover, the use of AMs and mechanical stretching suggested that ROCK regulated transcriptional level of NF-κB and NLRP3 inflammasome in AMs. Specifically, we silenced the ROCK1 and ROCK2 respectively, and found that the inflammation of MH-S cells after LPS and ATP priming could be regulated by ROCK1 and ROCK2, while the NLRP3 was only dependent upon ROCK1. Meantime, the related genes of NLRP3 signal are also regulated by ROCK1. Collectively, our data suggest that silencing ROCK1 ameliorates VILI in mice in part by inhibiting AMs' NLRP3 signaling pathway.
Collapse
|
20
|
Fisetin Inhibits NLRP3 Inflammasome by Suppressing TLR4/MD2-Mediated Mitochondrial ROS Production. Antioxidants (Basel) 2021; 10:antiox10081215. [PMID: 34439462 PMCID: PMC8389007 DOI: 10.3390/antiox10081215] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 07/25/2021] [Accepted: 07/26/2021] [Indexed: 12/25/2022] Open
Abstract
Fisetin has numerous therapeutic properties, such as anti-inflammatory, antioxidative, and anticancer effects. However, the mechanism by which fisetin inhibits NLRP3 inflammasome remains unclear. In this study, we observed that fisetin bound to TLR4 and occluded the hydrophobic pocket of MD2, which in turn inhibited the binding of LPS to the TLR4/MD2 complex. This prevented the initiation of scaffold formation by the inhibition of MyD88/IRAK4 and subsequently downregulated the NF-κB signaling pathway. The result also demonstrated that fisetin downregulated the activation of the NLRP3 inflammasome induced by LPS and ATP (LPS/ATP) and the subsequent maturation of IL-1β. Fisetin also activated mitophagy and prevented the accumulation of damaged mitochondria and the excessive production of mitochondrial reactive oxygen species. The transient knockdown of p62 reversed the inhibitory activity of fisetin on the LPS/ATP-induced formation of the NLRP3 inflammasome. This indicated that fisetin induces p62-mediated mitophagy for eliminating damaged mitochondria. Recently, the existence of inflammasomes in non-mammalian species including zebrafish have been identified. Treatment of an LPS/ATP-stimulated zebrafish model with fisetin aided the recovery of the impaired heart rate, decreased the recruitment of macrophage to the brain, and gradually downregulated the expression of inflammasome-related genes. These results indicated that fisetin inhibited the TLR4/MD2-mediated activation of NLRP3 inflammasome by eliminating damaged mitochondria in a p62-dependent manner.
Collapse
|
21
|
Johnson CT, Bradshaw HB. Modulatory Potential of Cannabidiol on the Opioid-Induced Inflammatory Response. Cannabis Cannabinoid Res 2021; 6:211-220. [PMID: 34115948 DOI: 10.1089/can.2020.0181] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Opioids are effective analgesics; however, there are many negative consequences of chronic use. One important side effect of chronic opioid use is the continuous engagement of the immune response that can exacerbate chronic pain. The opioid, morphine, initiates a Toll-like receptor 4 (TLR4) signaling cascade that drives the activation of NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflammasome proteins, resulting in cytokine production and effectively creating a positive feedback loop for continuous TLR4 activation. In addition to driving cytokine production, morphine drives changes in proinflammatory lipid signaling. The alteration of both cytokine and lipid signaling systems by morphine suggests that its chronic use leads to a pathological immune response that would benefit from targeted therapy. Engaging the endogenous cannabinoid system has shown therapeutic benefit, particularly regarding its anti-inflammatory and immunosuppressive effects. Promising preclinical and clinical investigations suggest that cannabidiol (CBD) is an effective adjuvant for treatment of symptoms of opioid use disorders; however, the mechanism through which CBD drives this outcome is unclear. One potential source of insight into this mechanism is in how CBD regulates immune regulators such as cytokines and lipid signaling systems, including endocannabinoids and related immune-responsive lipids. In this review, we outline the immune response to chronic opioid use as well as CBD in the context of a lipopolysaccharide-induced immune response and speculate on the mechanism of CBD as a modulator of chronic opioid-induced immune system dysregulation.
Collapse
Affiliation(s)
- Clare T Johnson
- Department of Psychological & Brain Science, Indiana University, Bloomington, Indiana, USA
| | - Heather B Bradshaw
- Department of Psychological & Brain Science, Indiana University, Bloomington, Indiana, USA
| |
Collapse
|
22
|
Li X, Zou Y, Fu YY, Xing J, Wang KY, Wan PZ, Wang M, Zhai XY. Ibudilast Attenuates Folic Acid-Induced Acute Kidney Injury by Blocking Pyroptosis Through TLR4-Mediated NF-κB and MAPK Signaling Pathways. Front Pharmacol 2021; 12:650283. [PMID: 34025417 PMCID: PMC8139578 DOI: 10.3389/fphar.2021.650283] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 04/19/2021] [Indexed: 02/06/2023] Open
Abstract
Folic acid (FA)-induced renal tubule damage, which is characterized by extensive inflammation, is a common model of acute kidney injury (AKI). Pyroptosis, a pro-inflammatory form of cell death due to the activation of inflammatory caspases, is involved in AKI progression. Ibudilast, a TLR4 antagonist, has been used in the clinic to exert an anti-inflammatory effect on asthma. However, researchers have not explored whether ibudilast exerts a protective effect on AKI by inhibiting inflammation. In the present study, ibudilast reversed FA-induced AKI in mice, as indicated by the reduced serum creatinine and urea nitrogen levels, and improved renal pathology, as well as the downregulation of kidney injury marker-1. In addition, ibudilast significantly increased the production of the anti-inflammatory factor IL-10 while suppressing the secretion of the pro-inflammatory cytokine TNF-α and macrophage infiltration. Moreover, in the injured kidney, ibudilast reduced the levels of both inflammasome markers (NLRP3) and pyroptosis-related proteins (caspase-1, IL1-β, IL-18, and GSDMD cleavage), and decreased the number of TUNEL-positive cells. Further mechanistic studies showed that ibudilast administration inhibited the FA-induced upregulation of TLR4, blocked NF-κB nuclear translocation, and reduced the phosphorylation of NF-κB and IκBα, p38, ERK, and JNK. Thus, this study substantiates the protective effect of ibudilast on FA-induced AKI in mice and suggests that protection might be achieved by reducing pyroptosis and inflammation, likely through the inhibition of TLR4-mediated NF-κB and MAPK signaling pathways.
Collapse
Affiliation(s)
- Xue Li
- Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, China.,Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yu Zou
- Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, China
| | - Yuan-Yuan Fu
- Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, China
| | - Jia Xing
- Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, China
| | - Kai-Yue Wang
- Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, China
| | - Peng-Zhi Wan
- Department of Nephrology, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Mo Wang
- Department of Surgery, Yale School of Medicine, New Haven, CT, United States
| | - Xiao-Yue Zhai
- Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, China.,Institute of Nephropathology, China Medical University, Shenyang, China
| |
Collapse
|
23
|
Jiang Y, Zhang W. LncRNA ZFAS1 plays a role in regulating the inflammatory responses in sepsis-induced acute lung injury via mediating miR-193a-3p. INFECTION GENETICS AND EVOLUTION 2021; 92:104860. [PMID: 33848686 DOI: 10.1016/j.meegid.2021.104860] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/24/2021] [Accepted: 04/08/2021] [Indexed: 12/31/2022]
Abstract
OBJECTIVE To explore the role of lncRNA ZFAS1-mediated miR-193a-3p in the regulation of inflammatory responses in rats with sepsis-induced acute lung injury (ALI). METHODS Sepsis-induced ALI models were constructed by LPS induction and then injected with ZFAS1 overexpression plasmid. Thereafter, lung injury score and the W/D weight ratio were calculated. Besides, bronchoalveolar lavage fluid (BALF) was isolated from rats to perform the cell count and protein quantification, while qRT-PCR and ELISA were performed to detect the inflammatory cytokines expressions. In vitro, NR8383 cells were transfected and then treated with LPS, followed by the measurement of inflammatory cytokines, cell viability and cell apoptosis. RESULTS In comparison with the Control group, rats in the LPS group presented sharp increases in the W/D weight ratio and injury score of lung, total protein concentration and the count of neutrophils and macrophages in BALF. Besides, rats in LPS group also resulted in a decrease in ZFAS1 expression and increase in miR-193a-3p expression in lung tissues, with the increased pro-inflammatory cytokines. Dual luciferase reporter gene assay confirmed a target relation between miR-193a-3p and ZFAS1. As compared to the Blank group, NR8383 cells in the LPS group had up-regulated pro-inflammatory cytokines with declined cell viability and elevated cell apoptosis; and meanwhile, ZFAS1 and Bcl-2 were decreased but miR-193a-3p and Bax were increased. Overexpression of ZFAS1 could significantly improve LPS-induced ALI in vivo and in vitro with reduced levels of pro-inflammatory cytokines. CONCLUSION Overexpression of ZFAS1, possibly via targeting the expression of miR-193a-3p, could inhibit the apoptosis and ameliorate the inflammatory responses of ALI in sepsis.
Collapse
Affiliation(s)
- Yan Jiang
- Department of Critical Care Medicine, Yantaishan Hospital, Yantai 264001, China
| | - Wei Zhang
- Department of Critical Care Medicine, Yantaishan Hospital, Yantai 264001, China.
| |
Collapse
|
24
|
Chen G, Liu Y, Xu Y, Zhang M, Guo S, Zhang G. Isoimperatorin exerts anti-inflammatory activity by targeting the LPS-TLR4/MD-2-NF-κB pathway. EUR J INFLAMM 2021. [DOI: 10.1177/20587392211000573] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Isoimperatorin (QHS) is a phytoconstituent found in the methanolic extracts obtained from the roots of Angelica dahurica, which contains anti-inflammatory, anti-bacterial, analgesic, anti-tumor, and vasodilatory activities. QHS possesses potent antagonistic activity against lipopolysaccharide (LPS)-induced inflammation; however, the mechanism of action remains unclear. In this study, we investigated the anti-inflammatory effect of QHS and explored the underlying mechanisms. The QHS was purchased from Jiangsu Yongjian Pharmaceutical Co., Ltd. (Jiangsu, China). We performed MTT assay, real-time PCR, ELISA, and western blotting experiments to assess the anti-inflammatory activity and the possible mechanism of QHS in vitro. Molecular docking was performed to study the binding of QHS and myeloid differentiation protein-2 (MD-2) and elucidate the possible anti-inflammatory mechanism. QHS had no significant effect on cell viability. Moreover, pre-treatment with QHS significantly decreased the release of inflammatory cytokines and mediators including NO, TNF-α, IL-6, and IL-1β. In addition, real-time PCR showed that QHS decreased the mRNA expressions of iNOS, COX-2 TNF-α, IL-6, and IL-1β. Western blotting indicated that QHS could inhibit the expression of the proteins associated with the LPS-TLR4/MD-2-NF-κB signaling pathway. Lastly, molecular docking revealed a possible binding mechanism between QHS and MD-2. QHS exhibited anti-inflammatory activity when combined with MD-2, regulating the LPS-TLR4/MD-2-NF-κB signaling pathway, and inhibiting the release and expression of inflammatory cytokines and mediators. Furthermore, QHS can be used as a potential TLR4 antagonist, which blocks MD-2 binding, for treating inflammatory responses induced by LPS.
Collapse
Affiliation(s)
- Guirong Chen
- 967th Hospital of the Joint Logistics Support Force of the Chinese People’s Liberation Army, Dalian, Liaoning, China
- Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
| | - Yunong Liu
- Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
| | - Yubin Xu
- Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, China
| | - Mingbo Zhang
- Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
| | - Song Guo
- Department of Computer Application, Shenyang Sport University, Shenyang, Liaoning, China
| | - Gang Zhang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
25
|
Liu B, He R, Zhang L, Hao B, Jiang W, Wang W, Geng Q. Inflammatory Caspases Drive Pyroptosis in Acute Lung Injury. Front Pharmacol 2021; 12:631256. [PMID: 33613295 PMCID: PMC7892432 DOI: 10.3389/fphar.2021.631256] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 01/06/2021] [Indexed: 12/16/2022] Open
Abstract
Acute lung injury (ALI), a critical respiratory disorder that causes diffuse alveolar injury leads to high mortality rates with no effective treatment. ALI is characterized by varying degrees of ventilation/perfusion mismatch, severe hypoxemia, and poor pulmonary compliance. The diffuse injury to cells is one of most important pathological characteristics of ALI. Pyroptosis is a form of programmed cell death distinguished from apoptosis induced by inflammatory caspases, which can release inflammatory cytokines to clear cells infected by pathogens and promote monocytes to reassemble at the site of injury. And pyroptosis not only promotes inflammation in certain cell types, but also regulates many downstream pathways to perform different functions. There is increasing evidence that pyroptosis and its related inflammatory caspases play an important role in the development of acute lung injury. The main modes of activation of pyroptosis is not consistent among different types of cells in lung tissue. Meanwhile, inhibition of inflammasome, the key to initiating pyroptosis is currently the main way to treat acute lung injury. The review summarizes the relationship among inflammatory caspases, pyroptosis and acute lung injury and provides general directions and strategies to conduct further research.
Collapse
Affiliation(s)
- Bohao Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ruyuan He
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lin Zhang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Bo Hao
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wenyang Jiang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wei Wang
- 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
|
26
|
Aboudounya MM, Heads RJ. COVID-19 and Toll-Like Receptor 4 (TLR4): SARS-CoV-2 May Bind and Activate TLR4 to Increase ACE2 Expression, Facilitating Entry and Causing Hyperinflammation. Mediators Inflamm 2021; 2021:8874339. [PMID: 33505220 PMCID: PMC7811571 DOI: 10.1155/2021/8874339] [Citation(s) in RCA: 240] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 12/16/2020] [Accepted: 12/22/2020] [Indexed: 01/08/2023] Open
Abstract
Causes of mortality from COVID-19 include respiratory failure, heart failure, and sepsis/multiorgan failure. TLR4 is an innate immune receptor on the cell surface that recognizes pathogen-associated molecular patterns (PAMPs) including viral proteins and triggers the production of type I interferons and proinflammatory cytokines to combat infection. It is expressed on both immune cells and tissue-resident cells. ACE2, the reported entry receptor for SARS-CoV-2, is only present on ~1-2% of the cells in the lungs or has a low pulmonary expression, and recently, the spike protein has been proposed to have the strongest protein-protein interaction with TLR4. Here, we review and connect evidence for SARS-CoV-1 and SARS-CoV-2 having direct and indirect binding to TLR4, together with other viral precedents, which when combined shed light on the COVID-19 pathophysiological puzzle. We propose a model in which the SARS-CoV-2 spike glycoprotein binds TLR4 and activates TLR4 signalling to increase cell surface expression of ACE2 facilitating entry. SARS-CoV-2 also destroys the type II alveolar cells that secrete pulmonary surfactants, which normally decrease the air/tissue surface tension and block TLR4 in the lungs thus promoting ARDS and inflammation. Furthermore, SARS-CoV-2-induced myocarditis and multiple-organ injury may be due to TLR4 activation, aberrant TLR4 signalling, and hyperinflammation in COVID-19 patients. Therefore, TLR4 contributes significantly to the pathogenesis of SARS-CoV-2, and its overactivation causes a prolonged or excessive innate immune response. TLR4 appears to be a promising therapeutic target in COVID-19, and since TLR4 antagonists have been previously trialled in sepsis and in other antiviral contexts, we propose the clinical trial testing of TLR4 antagonists in the treatment of severe COVID-19. Also, ongoing clinical trials of pulmonary surfactants in COVID-19 hold promise since they also block TLR4.
Collapse
Affiliation(s)
- Mohamed M. Aboudounya
- Department of Cardiology, The Rayne Institute, St Thomas' Hospital, British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, King's College London, UK
| | - Richard J. Heads
- Department of Cardiology, The Rayne Institute, St Thomas' Hospital, British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, King's College London, UK
| |
Collapse
|
27
|
Zhao J, Xie F, Chen R, Zhang Z, Dai R, Zhao N, Wang R, Sun Y, Chen Y. Transcription factor NF-κB promotes acute lung injury via microRNA-99b-mediated PRDM1 down-regulation. J Biol Chem 2020; 295:18638-18648. [PMID: 33109608 PMCID: PMC7939479 DOI: 10.1074/jbc.ra120.014861] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 10/08/2020] [Indexed: 01/12/2023] Open
Abstract
Acute lung injury (ALI), is a rapidly progressing heterogenous pulmonary disorder that possesses a high risk of mortality. Accumulating evidence has implicated the activation of the p65 subunit of NF-κB [NF-κB(p65)] activation in the pathological process of ALI. microRNAs (miRNAs), a group of small RNA molecules, have emerged as major governors due to their post-transcriptional regulation of gene expression in a wide array of pathological processes, including ALI. The dysregulation of miRNAs and NF-κB activation has been implicated in human diseases. In the current study, we set out to decipher the convergence of miR-99b and p65 NF-κB activation in ALI pathology. We measured the release of pro-inflammatory cytokines (IL-1β, IL-6, and TNFα) in bronchoalveolar lavage fluid using ELISA. MH-S cells were cultured and their viability were detected with cell counting kit 8 (CCK8) assays. The results showed that miR-99b was up-regulated, while PRDM1 was down-regulated in a lipopolysaccharide (LPS)-induced murine model of ALI. Mechanistic investigations showed that NF-κB(p65) was enriched at the miR-99b promoter region, and further promoted its transcriptional activity. Furthermore, miR-99b targeted PRDM1 by binding to its 3'UTR, causing its down-regulation. This in-creased lung injury, as evidenced by increased wet/dry ratio of mouse lung, myeloperoxidase activity and pro-inflammatory cytokine secretion, and enhanced infiltration of inflammatory cells in lung tissues. Together, our findings indicate that NF-κB(p65) promotion of miR-99b can aggravate ALI in mice by down-regulating the expression of PRDM1.
Collapse
Affiliation(s)
- Jie Zhao
- The Second Department of Pediatric, Cangzhou Central Hospital, Cangzhou, P. R. China.
| | - Fei Xie
- The Six Department of Pediatric, Cangzhou Central Hospital, Cangzhou, P. R. China
| | - Ruidong Chen
- The Six Department of Pediatric, Cangzhou Central Hospital, Cangzhou, P. R. China
| | - Zhen Zhang
- The Second Department of Pediatric, Cangzhou Central Hospital, Cangzhou, P. R. China
| | - Rujun Dai
- The Second Department of Pediatric, Cangzhou Central Hospital, Cangzhou, P. R. China
| | - Na Zhao
- The Second Department of Pediatric, Cangzhou Central Hospital, Cangzhou, P. R. China
| | - Rongxin Wang
- The Second Department of Pediatric, Cangzhou Central Hospital, Cangzhou, P. R. China
| | - Yanhong Sun
- The Second Department of Pediatric, Cangzhou Central Hospital, Cangzhou, P. R. China
| | - Yue Chen
- The Second Department of Pediatric, Cangzhou Central Hospital, Cangzhou, P. R. China
| |
Collapse
|
28
|
PIM2 deletion alleviates lipopolysaccharide (LPS)-induced respiratory distress syndrome (ARDS) by suppressing NLRP3 inflammasome. Biochem Biophys Res Commun 2020; 533:1419-1426. [PMID: 33333710 DOI: 10.1016/j.bbrc.2020.08.109] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 08/30/2020] [Indexed: 11/24/2022]
Abstract
Inflammation has an essential role in regulating the pathogenesis of acute respiratory distress syndrome (ARDS). The serine/threonine kinase PIM2 is highly expressed in human macrophages, and exhibits regulatory role in inflammatory response. However, its effect on ARDS progression has not been investigated and still remains unclear. In the study, we attempted to investigate the potential of PIM2 during ARDS progression, and to reveal the underlying molecular mechanisms. Here, we found that PIM2 expression was dramatically up-regulated in lipopolysaccharide (LPS)-exposed murine macrophages through a dose- and time-dependent manner. Additionally, we found that PIM2 knockdown greatly alleviated LPS-triggered activation of Caspase-1, interleukin (IL)-1β, NOD-like receptor pyrin domain 3 (NLRP3) and apoptosis-associated speck-like protein (ASC) in macrophages, along with suppressed inflammatory response. Importantly, we identified that PIM2 could directly interact with NLRP3. PIM2 over-expression could further promote LPS-triggered inflammation and NLRP3 inflammasome in macrophages. Furthermore, PIM2 knockout significantly alleviated the severity of ARDS in LPS-challenged mice. Evidently decreased inflammatory response and NLRP3 inflammasome were detected in pulmonary tissues of LPS-treated mice with PIM2 deficiency. Together, our findings demonstrated that PIM2 as a promising therapeutic target for ARDS treatment through regulating NLRP3 inflammasome.
Collapse
|
29
|
Li Q, Tan Y, Chen S, Xiao X, Zhang M, Wu Q, Dong M. Irisin alleviates LPS-induced liver injury and inflammation through inhibition of NLRP3 inflammasome and NF-κB signaling. J Recept Signal Transduct Res 2020; 41:294-303. [PMID: 32814473 DOI: 10.1080/10799893.2020.1808675] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Lipopolysaccharide (LPS) provokes severe inflammation and cell death in sepsis, with liver being the major affected organ. Up-to-date, neither the mechanism of action nor target treatment is readily available for LPS-induced liver injury. This study examined the effect of irisin, an endogenous hormonal peptide, on LPS-induced liver injury using animal and cell models, and the mechanism involved with a special focus on pyroptosis. Irisin is known to regulate glucose metabolism, inflammation, and immune response, while our earlier work denoted the anti-inflammatory and anti-apoptotic properties for irisin. Inflammatory factors and AST/ALT were also detected. Pyroptosis, apoptosis, and reactive oxygen species (ROS) were evaluated using PI staining, TUNEL staining, DCFH-DA fluorescence, and western blot, respectively. Our results indicated that irisin attenuated LPS-induced liver injury and release of inflammatory cytokines. Increased activity of NLRP3 inflammasome was discovered in LPS-challenged Raw264.7 cells, along with elevated levels of inflammation and apoptosis, the effects of which were mediated by activation of ROS and nuclear factor κB (NF-κB) signaling. These changes were reversed following irisin treatment. Our study demonstrated that irisin countered LPS-mediated liver injury via inhibiting apoptosis, NLRP3 inflammasome activation and NF-κB signaling. These findings revealed the role of irisin as a promising new anti-pyroptosis/apoptosis agent to reconcile the onset and progression of septic liver injury.
Collapse
Affiliation(s)
- Qian Li
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ying Tan
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou
| | - Sainan Chen
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaochan Xiao
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Mingming Zhang
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qi Wu
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Maolong Dong
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
30
|
Wang T, Hao Z, Liu C, Yuan L, Li L, Yin M, Li Q, Qi Z, Wang Z. LEF1 mediates osteoarthritis progression through circRNF121/miR-665/MYD88 axis via NF-кB signaling pathway. Cell Death Dis 2020; 11:598. [PMID: 32732957 PMCID: PMC7393488 DOI: 10.1038/s41419-020-02769-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 07/07/2020] [Accepted: 07/10/2020] [Indexed: 12/20/2022]
Abstract
Osteoarthritis (OA) is a joint disease that causes great pain to patients and imposes a tremendous burden on the world’s medical resources. Regulatory noncoding RNAs, including circular RNAs (circRNAs) and microRNAs (miRNAs), play an important role in OA progression. Here, we identified differential expression of transcription factor LEF1 that increased circRNA circRNF121 levels in normal and OA cartilage tissues. The expression of LEF1 and circRNF121 was positively associated with Mankin’s scores. Alteration of circRNF121 mediated the degradation of extracellular mechanisms (ECM), apoptosis, and proliferation of chondrocytes. MiR-665 was identified as a direct regulatory target of circRNF121 and MYD88. Functional analysis showed that circRNF121 and MYD88 modulated ECM degradation, apoptosis, and proliferation of chondrocytes, which could be reversed by miR-665. MYD88 regulated the activity of the NF-кB signaling pathway by circRNF121 via sponging miR-665. Collectively, these data indicated that LEF1 impacted OA progression by modulating the circRNF121/miR-665/MYD88 axis via NF-кB pathway. Our research proposed a new molecular mechanism for the development of OA, and provided a prospective therapeutic target for OA.
Collapse
Affiliation(s)
- Tianfu Wang
- Department of Sports Medicine, Dalian Municipal Central Hospital, Dalian, 116033, Liaoning Province, China.,Department of Spinal Surgery, The Second Hospital of Dalian Medical University, Dalian, 116033, Liaoning Province, China
| | - Zhiyu Hao
- Department of Medical Imageology, Dalian Medical University, Dalian, 116044, Liaoning Province, China
| | - Changcheng Liu
- Department of Sports Medicine, Dalian Municipal Central Hospital, Dalian, 116033, Liaoning Province, China
| | - Lebin Yuan
- Department of Sports Medicine, Dalian Municipal Central Hospital, Dalian, 116033, Liaoning Province, China
| | - Li Li
- Department of Sports Medicine, Dalian Municipal Central Hospital, Dalian, 116033, Liaoning Province, China
| | - Menghong Yin
- Department of Sports Medicine, Dalian Municipal Central Hospital, Dalian, 116033, Liaoning Province, China
| | - Qing Li
- Department of Sports Medicine, Dalian Municipal Central Hospital, Dalian, 116033, Liaoning Province, China
| | - Zhiming Qi
- Department of Sports Medicine, Dalian Municipal Central Hospital, Dalian, 116033, Liaoning Province, China
| | - Zi Wang
- Department of Sports Medicine, Dalian Municipal Central Hospital, Dalian, 116033, Liaoning Province, China.
| |
Collapse
|
31
|
Jiaweishaoyao Decoction Alleviates DSS-Induced Ulcerative Colitis via Inhibiting Inflammation. Gastroenterol Res Pract 2020; 2020:7182874. [PMID: 32565784 PMCID: PMC7273494 DOI: 10.1155/2020/7182874] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/09/2020] [Accepted: 05/11/2020] [Indexed: 02/07/2023] Open
Abstract
Purpose Jiaweishaoyao decoction (JWSYD) is a traditional prescription of Chinese medicine that is initially used for the treatment of diarrhea. This study is aimed at investigating the effects of JWSYD on DSS-induced ulcerative colitis (UC). Methods DSS-induced UC mice and LPS-induced RAW264.7 cells were used as the UC model in vivo and in vitro. UC was assessed by body weight, disease activity index (DAI), colon length, spleen weight, and histopathological score (HE staining). The levels of TNF-α, IL-1β, and IL-6 were analyzed by ELISA and qRT-PCR. The levels of NLRP3 inflammasome- and NF-κB pathway-associated proteins were measured by western blot. Results JWSYD alleviated DSS-induced UC in respect to body weight, DAI, colon length, spleen weight, and histopathological score. JWSYD reduced the levels of TNF-α, IL-1β, and IL-6 in DSS-induced UC mice and the supernatants of LPS-induced RAW264.7 cells. JWSYD suppressed the protein levels of inflammasome-associated proteins, including NLRP3, ASC1, Procaspase-1, Cleaved caspase-1, and Cleaved IL-1β in DSS-induced UC mice and LPS-induced RAW264.7 cells. In addition, JWSYD suppressed the NF-κB pathway in vitro and in vivo. Conclusion JWSYD alleviated DSS-induced UC via inhibiting the NLRP3 inflammasome and NF-κB pathway.
Collapse
|
32
|
Hu X, Ding C, Ding X, Fan P, Zheng J, Xiang H, Li X, Qiao Y, Xue W, Li Y. Inhibition of myeloid differentiation protein 2 attenuates renal ischemia/reperfusion-induced oxidative stress and inflammation via suppressing TLR4/TRAF6/NF-kB pathway. Life Sci 2020; 256:117864. [PMID: 32474021 DOI: 10.1016/j.lfs.2020.117864] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 05/22/2020] [Accepted: 05/26/2020] [Indexed: 12/18/2022]
Abstract
As a major risk factor of acute kidney injury, renal ischemia/reperfusion (I/R) has a high mortality rate. Myeloid differentiation protein 2 (MD-2) is a secretory glycoprotein that plays an important role in inflammation. Our study aimed to explore the roles of MD-2 in I/R-induced inflammation and oxidative stress in vivo and in vitro. For the in vivo studies, male C57BL/6 mice were randomly divided into four groups: 1) sham, 2) I/R, 3) negative control for siRNA (siNC) and I/R treatment, or 4) MD-2 siRNA (siMD-2) and I/R. Levels of blood urea nitrogen and creatinine in the plasma were tested, and hematoxylin and eosin staining was performed at 24 h after I/R injury. The inflammatory cytokines TNF-α, IL-6, and MCP-1 were measured using ELISA and Real-time qPCR (RT-qPCR). Malondialdehyde (MDA) content and superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPx) activity were estimated. For the in vitro studies, HK-2 cells were transfected with siMD-2 and then exposed to hypoxia/reoxygenation (H/R). Inflammatory cytokine expression and oxidative stress then were evaluated. We found decreased levels of blood urea nitrogen and creatinine levels after MD-2 silencing. MD-2 deficiency improved histological damage. MD-2 downregulation attenuated levels of inflammatory cytokines. Inhibition of MD-2 resulted in reduced MDA content and increased SOD, CAT, and GPx activity. Loss of function of MD-2 inhibited the H/R-induced production and expression of inflammatory cytokines. MD-2 silencing reduced MDA content after H/R, and MD-2 suppression enhanced SOD, CAT, and GPx activity. MD-2 deficiency also blocked H/R-mediated activation of the TLR4/TRAF6/NF-κB pathway, and pyrrolidinedithiocarbamate (PDTC) pretreatment strengthened the anti-inflammatory and antioxidant damage effects of MD-2 silencing. Taken together, our study revealed that MD-2 deficiency ameliorated renal I/R-induced inflammation and oxidative stress via inhibition of TLR4/TRAF6/NF-κB pathway.
Collapse
Affiliation(s)
- Xiaojun Hu
- Department of Renal Transplantation, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Chenguang Ding
- Department of Renal Transplantation, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Xiaoming Ding
- Department of Renal Transplantation, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Ping Fan
- Department of Rheumatism and Immunology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Jin Zheng
- Department of Renal Transplantation, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Heli Xiang
- Department of Renal Transplantation, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Xiao Li
- Department of Renal Transplantation, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Yuxi Qiao
- Department of Renal Transplantation, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Wujun Xue
- Department of Renal Transplantation, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China.
| | - Yang Li
- Department of Renal Transplantation, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China.
| |
Collapse
|
33
|
Yang Y, Ding Z, Wang Y, Zhong R, Feng Y, Xia T, Xie Y, Yang B, Sun X, Shu Z. Systems pharmacology reveals the mechanism of activity of Physalis alkekengi L. var. franchetii against lipopolysaccharide-induced acute lung injury. J Cell Mol Med 2020; 24:5039-5056. [PMID: 32220053 PMCID: PMC7205831 DOI: 10.1111/jcmm.15126] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 01/03/2020] [Accepted: 01/21/2020] [Indexed: 12/11/2022] Open
Abstract
Acute lung injury (ALI) is an important cause of mortality of patients with sepsis, shock, trauma, pneumonia, multiple transfusions and pancreatitis. Physalis alkekengi L. var. franchetii (Mast.) Makino (PAF) has been extensively used in Chinese folk medicine because of a good therapeutic effect in respiratory diseases. Here, an integrated approach combining network pharmacology, proton nuclear magnetic resonance-based metabolomics, histopathological analysis and biochemical assays was used to elucidate the mechanism of PAF against ALI induced by lipopolysaccharide (LPS) in a mouse model. We found that the compounds present in PAF interact with 32 targets to effectively improve the damage in the lung undergoing ALI. We predicted the putative signalling pathway involved by using the network pharmacology and then used the orthogonal signal correction partial least-squares discriminant analysis to analyse the disturbances in the serum metabolome in mouse. We also used ELISA, RT-qPCR, Western blotting, immunohistochemistry and TUNEL assay to confirm the potential signalling pathways involved. We found that PAF reduced the release of cytokines, such as TNF-α, and the accumulation of oxidation products; decreased the levels of NF-κB, p-p38, ERK, JNK, p53, caspase-3 and COX-2; and enhanced the translocation of Nrf2 from the cytoplasm to the nucleus. Collectively, PAF significantly reduced oxidative stress injury and inflammation, at the same time correcting the energy metabolism imbalance caused by ALI, increasing the amount of antioxidant-related metabolites and reducing the apoptosis of lung cells. These observations suggest that PAF may be an effective candidate preparation alleviating ALI.
Collapse
Affiliation(s)
- Yanni Yang
- Guangdong Standardized Processing Engineering Technology Research Center of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- Department of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Zihe Ding
- Guangdong Standardized Processing Engineering Technology Research Center of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- Department of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yi Wang
- Guangdong Standardized Processing Engineering Technology Research Center of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Renxing Zhong
- Guangdong Standardized Processing Engineering Technology Research Center of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- Department of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yanlin Feng
- Department of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Tianyi Xia
- Guangdong Standardized Processing Engineering Technology Research Center of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- Department of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yuanyuan Xie
- Department of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Bingyou Yang
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xiaobo Sun
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Zunpeng Shu
- Guangdong Standardized Processing Engineering Technology Research Center of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- Department of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| |
Collapse
|
34
|
Propofol post-conditioning lessens renal ischemia/reperfusion-induced acute lung injury associated with autophagy and apoptosis through MAPK signals in rats. Gene 2020; 741:144562. [PMID: 32169629 DOI: 10.1016/j.gene.2020.144562] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 03/08/2020] [Indexed: 12/14/2022]
Abstract
Renal Ischemia/Reperfusion (rI/R)-induced acute lung injury (ALI) is a major problem in rI/R. The objective of the current study was to explore the defensive roles of propofol (Pro), an intravenous anesthetic, on rI/R-induced ALI through mitogen-activated protein kinase (MAPK) signaling. Rats were divided into Sham, Pro (10 mg/kg), rI/R, rI/R + Pro (5 mg/kg), and rI/R + Pro (10 mg/kg) groups. Rats were treated with Pro at 1 h after rI/R treatment. Serum and lung tissues at 24 h after rI/R were collected to evaluate morphological changes and the expression of myeloperoxidase (MPO), inflammatory cytokines, and crucial proteins in the MAPK pathway. Pro attenuated the production of mediators, resulting in reduced levels of autophagy and apoptosis by restricting the MAPK pathway in rI/R-induced ALI model. Pro represses rI/R-induced pulmonary autophagy and apoptosis by decreasing the production of inflammatory molecules, and the effects of Pro are involved in the inhibition of the MAPK pathway.
Collapse
|
35
|
The HO-1 Signal Prevents HMGB1-Mediated Activation of NLRP3 Inflammasomes in Lipopolysaccharide-Induced Acute Lung Injury In Vitro. J Surg Res 2020; 247:335-343. [DOI: 10.1016/j.jss.2019.10.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Revised: 09/08/2019] [Accepted: 10/01/2019] [Indexed: 01/02/2023]
|
36
|
Cheng H, Zhu J, Shan J, Ouyang X, Yang H, Wei R, Zeng J. Involvement of toll-like receptor 2/myeloid differentiation factor 88/nuclear factor kappa B/NLR family pyrin domain-containing 3 signaling pathways in the hepatoprotective effect of Lagotis brachystachys in rats with alcoholic liver disease. Pharmacogn Mag 2020. [DOI: 10.4103/pm.pm_557_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
37
|
Zhang Z, Tian L, Jiang K. Propofol attenuates inflammatory response and apoptosis to protect d-galactosamine/lipopolysaccharide induced acute liver injury via regulating TLR4/NF-κB/NLRP3 pathway. Int Immunopharmacol 2019; 77:105974. [PMID: 31735662 DOI: 10.1016/j.intimp.2019.105974] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 10/09/2019] [Accepted: 10/11/2019] [Indexed: 02/08/2023]
Abstract
OBJECTIVE Propofol has been reported to be protective against liver injury due to its anti-inflammatory, anti-oxidative and anti-apoptotic activities. The purpose of this study was to examine the protective effects of propofol on d-galactosamine/lipopolysaccharide (d-GalN/LPS) induced acute liver injury. METHODS Mice were given an intraperitoneal injection of propofol before d-GalN/LPS treatment. Liver injury was confirmed by serum biochemical analysis and liver histopathological analysis. Relevant molecular events were determined by ELISA, western blot, and test kits. Cell apoptosis were evaluated by TUNEL assay. RESULTS The results showed that propofol significantly prevented d-GalN/LPS-induced liver damage by preventing associated increases of serum alanine transaminase (ALT) and aspartate transaminase (AST) and restoring liver histopathological changes. Propofol markedly inhibited the production of inflammatory cytokines and oxidative stress-related factors. Propofol markedly reduced hepatocyte apoptosis, decreased Bax, Bad, cleaved caspase-3 and increased Bcl-2 expression. Besides, NLRP3 inflammasome and TLR4/NF-κB pathway were inactivated under the treatment of propofol according to the expression of pathways-related proteins. CONCLUSION Taken together, propofol contributed to liver protection against d-GalN/LPS-induced liver injury in mice by inhibiting inflammation, oxidative stress and hepatocyte apoptosis through regulating TLR4/NF-κB/NLRP3 pathway.
Collapse
Affiliation(s)
- Zhaojian Zhang
- Department of Anaesthesiology, The First People's Hospital of Lianyungang, 222000, China
| | - Liang Tian
- Department of Anaesthesiology, The First People's Hospital of Lianyungang, 222000, China
| | - Kai Jiang
- Department of Hepatobiliary and Pancreatic Surgery & Minimally Invasive Surgery, Zhejiang Provincial People's Hospital, Hangzhou 310058, China.
| |
Collapse
|
38
|
Hernandez A, Patil NK, Stothers CL, Luan L, McBride MA, Owen AM, Burelbach KR, Williams DL, Sherwood ER, Bohannon JK. Immunobiology and application of toll-like receptor 4 agonists to augment host resistance to infection. Pharmacol Res 2019; 150:104502. [PMID: 31689522 DOI: 10.1016/j.phrs.2019.104502] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 10/04/2019] [Accepted: 10/15/2019] [Indexed: 12/19/2022]
Abstract
Infectious diseases remain a threat to critically ill patients, particularly with the rise of antibiotic-resistant bacteria. Septic shock carries a mortality of up to ∼40% with no compelling evidence of promising therapy to reduce morbidity or mortality. Septic shock survivors are also prone to nosocomial infections. Treatment with toll-like receptor 4 (TLR4) agonists have demonstrated significant protection against common nosocomial pathogens in various clinically relevant models of infection and septic shock. TLR4 agonists are derived from a bacteria cell wall or synthesized de novo, and more recently novel small molecule TLR4 agonists have also been developed. TLR4 agonists augment innate immune functions including expansion and recruitment of innate leukocytes to the site of infection. Recent studies demonstrate TLR4-induced leukocyte metabolic reprogramming of cellular metabolism to improve antimicrobial function. Metabolic changes include sustained augmentation of macrophage glycolysis, mitochondrial function, and tricarboxylic acid cycle flux. These findings set the stage for the use of TLR4 agonists as standalone therapeutic agents or antimicrobial adjuncts in patient populations vulnerable to nosocomial infections.
Collapse
Affiliation(s)
- Antonio Hernandez
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Naeem K Patil
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Cody L Stothers
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Liming Luan
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Margaret A McBride
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Allison M Owen
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Katherine R Burelbach
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - David L Williams
- Department of Surgery, East Tennessee State University, James H. Quillen College of Medicine, Johnson City, TN, USA
| | - Edward R Sherwood
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Julia K Bohannon
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
39
|
Huang JJ, Xia J, Huang LL, Li YC. HIF‑1α promotes NLRP3 inflammasome activation in bleomycin‑induced acute lung injury. Mol Med Rep 2019; 20:3424-3432. [PMID: 31432144 DOI: 10.3892/mmr.2019.10575] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Accepted: 07/12/2019] [Indexed: 11/05/2022] Open
Abstract
The inflammatory response is one of the most important factors in the occurrence and development of acute lung injury (ALI). Hypoxia‑inducible factor‑1α (HIF‑1α) and the NOD‑like receptor 3 (NLRP3) inflammasome have been demonstrated to serve an important role in the pathogenesis of ALI. The objective of the present study was to investigate whether HIF‑1α could regulate activation of the NLRP3 inflammasome and its potential function and specific mechanism in bleomycin (BLM)‑induced ALI. Activation of the NLRP3 inflammasome and secretion of IL‑1β were detected following silencing of HIF‑1α or NF‑κB, respectively, in BLM‑treated A549 and RLE‑6TN cells. The results demonstrated that the NLRP3 inflammasome could be activated after BLM treatment. HIF‑1α and NF‑κB expression significantly increased in the BLM group. The levels of NF‑κB‑ and NLRP3 inflammasome‑associated proteins, including NLRP3, apoptosis‑associated speck‑like protein containing CARD and caspase‑1, markedly decreased after treating A549 and RLE‑6TN cells with HIF‑1α small interfering RNA. Activation of the NLRP3 inflammasome was also inhibited after silencing NF‑κB. Furthermore, the levels of IL‑1β markedly decreased in the cellular culture supernatants following inhibition of HIF‑1α and NF‑κB. Therefore, the present study indicated that HIF‑1α could modulate the activation of the NLRP3 inflammasome and the secretion of IL‑1β through NF‑κB signaling in BLM‑induced ALI. The current results improve understanding of the mechanism of ALI and may provide new ideas for identifying therapeutic targets of ALI.
Collapse
Affiliation(s)
- Jun-Jun Huang
- Department of Geriatric Rehabilitation, Geriatric Rehabilitation Hospital of Nantong, Branch of Nantong University's Affiliated Hospital, Nantong, Jiangsu 226001, P.R. China
| | - Jie Xia
- Department of Gastroenterology, Changzhou No. 2 People's Hospital, Changzhou, Jiangsu 213164, P.R. China
| | - Li-Li Huang
- Department of Geriatric Rehabilitation, Geriatric Rehabilitation Hospital of Nantong, Branch of Nantong University's Affiliated Hospital, Nantong, Jiangsu 226001, P.R. China
| | - Ya-Chun Li
- Department of Anesthesiology, The Central Hospital of Songjiang, Songjiang Branch of Shanghai General Hospital Affiliated to Shanghai Jiaotong University, Shanghai 201600, P.R. China
| |
Collapse
|
40
|
Li C, Wang X, Kuang M, Li L, Wang Y, Yang F, Wang G. UFL1 modulates NLRP3 inflammasome activation and protects against pyroptosis in LPS-stimulated bovine mammary epithelial cells. Mol Immunol 2019; 112:1-9. [DOI: 10.1016/j.molimm.2019.04.023] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 04/15/2019] [Accepted: 04/27/2019] [Indexed: 12/13/2022]
|
41
|
He Y, Xu K, Wang Y, Chao X, Xu B, Wu J, Shen J, Ren W, Hu Y. AMPK as a potential pharmacological target for alleviating LPS-induced acute lung injury partly via NLRC4 inflammasome pathway inhibition. Exp Gerontol 2019; 125:110661. [PMID: 31319131 DOI: 10.1016/j.exger.2019.110661] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 06/09/2019] [Accepted: 07/14/2019] [Indexed: 12/12/2022]
Abstract
Old people are spectacularly susceptible to acute lung injury (ALI) and the accompanying complications. An acute aggravated inflammatory response is a characteristic feature of ALI, and inflammasomes play a critical role in the inflammatory response. Metformin has been shown to be an effective anti-inflammatory agent in ALI. However, the mechanism of this regulation still remains poorly understood. In this study, 18- to 19-month-old male mice were treated by intratracheal instillation of lipopolysaccharide (LPS) or PBS with or without metformin pretreatment. We found that the metformin pretreatment alleviated the lung injury and decreased the levels of TNF-a, IL-1β and IL-6 in the bronchoalveolar lavage fluid (BALF) and in lung tissues, as well as the levels of NLRP3, NLRC4 and cleaved caspase-1 associated with LPS-induced ALI in old mice. Furthermore, the in vitro study showed metformin dose-dependently suppressed NLRC4 inflammasome expression. Metformin activated AMPK by phosphorylation; thus, we investigated the role of AMPK in NLRC4 activation. The results demonstrated that the efficacy of metformin was reduced when using the AMPK pharmacological inhibitor compound C or AMPKα1 expression was knocked down in RAW 264.7 cells. In conclusion, our data indicated that metformin may inhibit NLRC4 inflammasome activation in LPS-induced ALI in old mice through AMPK signaling, and further understanding of the AMPK/NLRC4 axis may provide a novel therapeutic strategy for LPS-induced ALI in the future.
Collapse
Affiliation(s)
- Yuting He
- Department of Geriatrics, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Kan Xu
- Department of Geriatrics, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Yao Wang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Xin Chao
- Department of Geriatrics, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Bing'er Xu
- Department of Geriatrics, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Jiayu Wu
- Department of Geriatrics, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Jiping Shen
- Department of Geriatrics, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Weiying Ren
- Department of Geriatrics, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China.
| | - Yu Hu
- Department of Geriatrics, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China.
| |
Collapse
|
42
|
Wang B, Wang J, Lu D, Qi N, Liu Q. The Defensive Action of LYRM03 on LPS-Induced Acute Lung Injury by NF-κB/TLR4/NLRP3 Signals. J INVEST SURG 2019; 34:284-296. [PMID: 31274341 DOI: 10.1080/08941939.2019.1634165] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The aim of the current investigation was to study the role of 3-amino-2-hydroxy-4-phenyl-valyl-isoleucine (LYRM03) in lipopolysaccharide (LPS)-induced acute lung injury (ALI) and investigate its potential pathogenesis. An LPS-induced ALI model was produced with LPS (5 mg/kg) followed by 24 h of injury. Rats were randomly assigned to 6 groups for in vivo experiments: (1) Sham, (2) LYRM03 (20 mg/kg), (3) LPS, (4) LPS plus LYRM03 (5 mg/kg), (5) LPS plus LYRM03 (10 mg/kg), and (6) LPS plus LYRM03 (20 mg/kg). The rat alveolar macrophage cell line (NR8383) cells were divided into 6 groups for in vitro experiments: (1) Sham, (2) LYRM03 (200 μmol/L), (3) LPS (100 ng/mL), (4) LPS plus LYRM03 (50 μmol/L), (5) LPS plus LYRM03 (100 μmol/L), and (6) LPS plus LYRM03 (200 μmol/L). Further study about siRNA targeting NF-κB p65, TLR4, and NLRP3 to explore the potential mechanism of LYRM03 in the LPS-induced ALI models have been done. Therefore, LYRM03 decreased LPS-induced ALI and NR8383 activation as demonstrated through hematoxylin-eosin staining and western blot analysis in vivo and in vitro. LYRM03 ameliorated the content of protein in bronchoalveolar lavage fluid, myeloperoxidase in the lung and malondialdehyde (MDA) in serum. In addition, LYRM03 ameliorated the levels of tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and interleukin-18 (IL-18) in the serum of rats and the supernatant of NR8383 cells. Moreover, LYRM03 significantly inhibited the activities of nuclear factor kappa B (NF-κB), myeloid differentiation factor 88 (MyD88), and toll-like receptor 4 (TLR4). LYRM03 also reduced the increase in the inflammasome, including apoptosis-related speck-like protein containing CARD (ASC), and NOD-like receptor 3 (NLRP3), in LPS-stimulated rats and NR8383 cells. The extent of injury and lung injury scores in the LYRM03 (20 mg/kg) + siRNA targeting NF-κB p65, TLR4, or NLRP3 + LPS-treated rats were higher than that in the LYRM03 (20 mg/kg) + LPS-treated rats. In summary, LYRM03 conferred an intensely lung defensive action on LPS-induced ALI in vivo and in vitro, which could be associated with the abatement of TLR4-induced NLRP3/NF-κB.
Collapse
Affiliation(s)
- Bin Wang
- Department of Critical Care Medicine, Rizhao People Hospital, Rizhao, People's Republic of China
| | - Jiaoyue Wang
- Department of Critical Care Medicine, Rizhao People Hospital, Rizhao, People's Republic of China
| | - Daopeng Lu
- Department of Emergency, Jinan Medical Emergency Center, Jinan, People's Republic of China
| | - Na Qi
- Department of Respiratory Medicine, Hengshui People Hospital, Hengshui, People's Republic of China
| | - Qin Liu
- Department of Emergency, Jinan Medical Emergency Center, Jinan, People's Republic of China
| |
Collapse
|
43
|
Bai X, Guo A, Li Y. Protective effects of calcitonin on IL-1 stimulated chondrocytes by regulating MMPs/TIMP-1 ratio via suppression of p50-NF-κB pathway. Biosci Biotechnol Biochem 2019; 83:598-604. [PMID: 30582416 DOI: 10.1080/09168451.2018.1559718] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
ABSTRACT
The aim of this study was to investigate the effects and underlying mechanisms of calcitonin (CT) on interleukin 1 beta (IL-1β) stimulated human chondrocytes. IL-1β (5 ng/mL) was added into chondrocytes to establish osteoarthritis (OA) model in vitro. Different concentrations of CT (0.1, 0.5, 1, 5, 10 and 50 nM) were used for treating IL-1β stimulated chondrocytes. Cell viability of chondrocytes was measured by cell counting kit-8 (CCK8) method. Western blotting was performed to evaluate the expression of matrix metalloproteinases (MMP-13), tissue inhibitor of metalloproteinases 1 (TIMP-1), p50 and p38. CT inhibited MMP-13 expression and promoted TIMP-1 expression in the IL-1β stimulated human chondrocytes. The CT-mediated alteration of MMP-13/TIMP-1 ratio was partially attributed to the inactivation of the p50- nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) pathway by suppressing p50 in IL-1β stimulated chondrocytes. CT might play a protective role in IL-1β stimulated OA model via p50-NF-κB pathway.
Abbreviations: CT: calcitonin; IL-1β: interleukin-1β; MMP-13: matrix metalloproteinases-13; TIMP-1: tissue inhibitors of metalloproteinases-1.
Collapse
Affiliation(s)
- Xiaodong Bai
- Department of Orthopedics, Beijing Friendship Hospital, Capital Medical University, Beijing, P.R. China
| | - Ai Guo
- Department of Orthopedics, Beijing Friendship Hospital, Capital Medical University, Beijing, P.R. China
| | - Yadong Li
- Department of Orthopedics, Beijing Friendship Hospital, Capital Medical University, Beijing, P.R. China
| |
Collapse
|
44
|
Zhou Z, Su Y, Fa XE. Isorhynchophylline exerts anti-inflammatory and anti-oxidative activities in LPS-stimulated murine alveolar macrophages. Life Sci 2019; 223:137-145. [DOI: 10.1016/j.lfs.2019.03.017] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 02/27/2019] [Accepted: 03/07/2019] [Indexed: 01/17/2023]
|
45
|
Li N, Xu H, Ou Y, Feng Z, Zhang Q, Zhu Q, Cai Z. LPS-induced CXCR7 expression promotes gastric Cancer proliferation and migration via the TLR4/MD-2 pathway. Diagn Pathol 2019; 14:3. [PMID: 30636642 PMCID: PMC6330400 DOI: 10.1186/s13000-019-0780-x] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 01/04/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Lipopolysaccharide (LPS) from Helicobacter pylori (HP) plays an important role in gastric cancer occurrence and development. Toll-like receptor 4 (TLR4) and myeloid differential protein-2 (MD-2) are also reported to be involved in gastric cancer cell proliferation and invasion. CXC chemokine receptor 7 (CXCR7), a second receptor for CXCL12, has been detected in multiple types of tumor tissues. Nevertheless, the biological function and regulation of CXCR7 and its relationship with TLR4 and MD-2 in gastric cancer are not completely understood and therefore warrant further study. METHODS CXCR7 expression was examined in 150 gastric cancer tissues using immunohistochemistry (IHC). RT-PCR and western blotting were used to detect CXCR7 expression in several gastric cancer cell lines (SGC7901, AGS, MGC-803, MKN-45 and BGC823). shRNAs were designed using a pGPU6/GFP/Neo vector. A CCK-8 assay was used to assess cell proliferation, and transwell assays were performed to assess cell migration. In addition, a gastric cancer xenograft model was generated. RESULTS The LPS-TLR4-MD-2 pathway elevates CXCR7 expression in SGC7901 cells, and TLR4/MD-2-mediated increases in CXCR7 levels modulate the proliferation and migration of tumor cells. Knockdown of TLR4 and MD-2 demonstrated that both are essential for LPS-induced CXCR7 expression, which in turn is responsible for LPS-induced SGC7901 cell proliferation and migration. Moreover, higher TLR4, MD-2 and CXCR7 expression was detected in gastric cancer tissues than in paracancerous normal control tissues. The expression levels of TLR4, MD-2 and CXCR7 were closely related to gastric cancer TNM stage and lymph node metastasis. In an animal model, significant differences in CXCR7 expression in tumor masses were observed between the control group and experimental group. CONCLUSIONS The results of this study indicate that CXCR7 plays an important role in gastric cancer progression via inflammatory mechanisms, suggesting that CXCR7 could provide a basis for the development and clinical application of a targeted drug for gastric cancer.
Collapse
Affiliation(s)
- Nan Li
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China.,Department of Pathology, Bengbu Medical College, Bengbu, China
| | - Huanbai Xu
- Department of Endocrinology and Metabolism, Shanghai Jiaotong University Affiliated First People's Hospital, Shanghai, China
| | - Yurong Ou
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China.,Department of Pathology, Bengbu Medical College, Bengbu, China
| | - Zhenzhong Feng
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China.,Department of Pathology, Bengbu Medical College, Bengbu, China
| | - Qiong Zhang
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China.,Department of Pathology, Bengbu Medical College, Bengbu, China
| | - Qing Zhu
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China.,Department of Pathology, Bengbu Medical College, Bengbu, China
| | - Zhaogen Cai
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China. .,Department of Pathology, Bengbu Medical College, Bengbu, China.
| |
Collapse
|
46
|
Immanuel CN, Teng B, Dong B, Gordon EM, Kennedy JA, Luellen C, Schwingshackl A, Cormier SA, Fitzpatrick EA, Waters CM. Apoptosis signal-regulating kinase-1 promotes inflammasome priming in macrophages. Am J Physiol Lung Cell Mol Physiol 2019; 316:L418-L427. [PMID: 30628485 DOI: 10.1152/ajplung.00199.2018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
We previously showed that mice deficient in apoptosis signal-regulating kinase-1 (ASK1) were partially protected against ventilator-induced lung injury. Because ASK1 can promote both cell death and inflammation, we hypothesized that ASK1 activation regulates inflammasome-mediated inflammation. Mice deficient in ASK1 expression (ASK1-/-) exhibited significantly less inflammation and lung injury (as measured by neutrophil infiltration, IL-6, and IL-1β) in response to treatment with inhaled lipopolysaccharide (LPS) compared with wild-type (WT) mice. To determine whether this proinflammatory response was mediated by ASK1, we investigated inflammasome-mediated responses to LPS in primary macrophages and bone marrow-derived macrophages (BMDMs) from WT and ASK1-/- mice, as well as the mouse alveolar macrophage cell line MH-S. Cells were treated with LPS alone for priming or LPS followed by ATP for activation. When macrophages were stimulated with LPS followed by ATP to activate the inflammasome, we found a significant increase in secreted IL-1β from WT cells compared with ASK1-deficient cells. LPS priming stimulated an increase in NOD-like receptor 3 (NLRP3) and pro-IL-1β in WT BMDMs, but expression of NLRP3 was significantly decreased in ASK1-/- BMDMs. Subsequent ATP treatment stimulated an increase in cleaved caspase-1 and IL-1β in WT BMDMs compared with ASK1-/- BMDMs. Similarly, treatment of MH-S cells with LPS + ATP caused an increase in both cleaved caspase-1 and IL-1β that was diminished by the ASK-1 inhibitor NQDI1. These results demonstrate, for the first time, that ASK1 promotes inflammasome priming.
Collapse
Affiliation(s)
- Camille N Immanuel
- Division of Pediatric Critical Care, Department of Pediatrics, Children's Foundation Research Institute at Le Bonheur Children's Hospital, University of Tennessee Health Sciences , Memphis, Tennessee.,Department of Physiology, University of Kentucky , Lexington, Kentucky
| | - Bin Teng
- Department of Physiology, University of Tennessee Health Science Center , Memphis, Tennessee
| | - Brittany Dong
- Department of Physiology, University of Kentucky , Lexington, Kentucky
| | | | - Joseph A Kennedy
- Department of Physiology, University of Tennessee Health Science Center , Memphis, Tennessee
| | - Charlean Luellen
- Department of Physiology, University of Tennessee Health Science Center , Memphis, Tennessee
| | - Andreas Schwingshackl
- Department of Pediatrics, Mattel Children's Hospital at the University of California , Los Angeles, California
| | - Stephania A Cormier
- Department of Biological Sciences, Louisiana State University , Baton Rouge, Louisiana
| | - Elizabeth A Fitzpatrick
- Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center , Memphis, Tennessee
| | | |
Collapse
|
47
|
The utility of complement assays in clinical immunology: A comprehensive review. J Autoimmun 2018; 95:191-200. [PMID: 30391025 DOI: 10.1016/j.jaut.2018.10.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 10/17/2018] [Indexed: 12/19/2022]
Abstract
The multi-tasking organ liver, which is the major synthesis site of most serum proteins, supplies humoral components of the innate, - including proteins of the complement system; and, less intensely, also of the acquired immune system. In addition to hepatocyte origins, C1q, factor D, C3, C7 and other protein components of the complement system are produced at various body locations by monocytes/macrophages, lymphocytes, adipocytes, endometrium, enterocytes, keratinocytes and epithelial cells; but the contribution of these alternate sites to the total serum concentrations is slight. The two major exceptions are factor D, which cleaves factor B of the alternative pathway derived largely from adipocytes, and C7, derived largely from polymorphonuclear leukocytes and monocytes/macrophages. Whereas the functional meaning of the extrahepatic synthesis of factor D remains to be elucidated, the local contribution of C7 may up- or downregulate the complement attack. The liver, however, is not classified as part of the immune system but is rather seen as victim of autoimmune diseases, a point that needs apology. Recent histological and cell marker technologies now turn the hands to also conceive the liver as proactive autoimmune disease catalyst. Hosting non-hepatocytic cells, e.g. NK cells, macrophages, dendritic cells as well as T and B lymphocytes, the liver outreaches multiple sites of the immune system. Immunopharmacological follow up of liver transplant recipients teaches us on liver-based presence of ABH-glycan HLA phenotypes and complement mediated ischemia/regeneration processes. In clinical context, the adverse reactions of the complement system can now be curbed by specific drug therapy. This review extends on the involvement of the complement system in liver autoimmune diseases and should allow to direct therapeutic opportunities.
Collapse
|
48
|
Loss H, Aschenbach JR, Ebner F, Tedin K, Lodemann U. Effects of a pathogenic ETEC strain and a probiotic Enterococcus faecium strain on the inflammasome response in porcine dendritic cells. Vet Immunol Immunopathol 2018; 203:78-87. [DOI: 10.1016/j.vetimm.2018.08.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 07/18/2018] [Accepted: 08/12/2018] [Indexed: 01/27/2023]
|
49
|
Targeting the NLRP3 Inflammasome-Related Pathways via Tianeptine Treatment-Suppressed Microglia Polarization to the M1 Phenotype in Lipopolysaccharide-Stimulated Cultures. Int J Mol Sci 2018; 19:ijms19071965. [PMID: 29976873 PMCID: PMC6073715 DOI: 10.3390/ijms19071965] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 06/29/2018] [Accepted: 07/03/2018] [Indexed: 12/28/2022] Open
Abstract
An increasing body of evidence postulates that microglia are the main mediators of inflammation-related disorders, including depression. Since activated microglia produce a wide range of pro- and anti-inflammatory factors, the modulation of M1/M2 microglial polarization by antidepressants may be crucial in the treatment of depression. The current paper aimed to investigate the impact of tianeptine on the microglia’s viability/death parameters, and on M1/M2 microglial activation in response to lipopolysaccharide (LPS) stimulation. Furthermore, the molecular mechanisms via which tianeptine affected the LPS-evoked changes were investigated. The results revealed that tianeptine had partially protective effects on the changes in microglia viability/death evoked by LPS. Tianeptine attenuated microglia activation by decreasing the expression of cluster of differentiation 40 (CD40), and major histocompatibility complex class II (MHC II) markers, as well as the release of pro-inflammatory factors: interleukin (IL)-1β, IL-18, IL-6, tumor necrosis factor alpha (TNF-α), and chemokine CC motif ligand 2 (CCL2), and the production of nitric oxide and reactive oxygen species. In contrast, we did not observe an impact of tianeptine on M2 microglia measured by IL-4, IL-10, TGF-β, and insulin-like growth factor 1 (IGF-1) expression. Moreover, we demonstrated an inhibitory effect of tianeptine on the LPS-induced activation of the nucleotide-binding oligomerization domain-like (NOD-like) receptor pyrin-containing 3 inflammasome (NLRP3) inflammasome subunits, NLRP3 and caspase-1, as well as the ability of tianeptine to reduce Toll-like receptor 4 (TLR4) levels, as well as the phosphorylation of extracellular signal-related kinases 1 and 2 (ERK1/2) and of the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). Collectively, we demonstrated that tianeptine has protective properties and inhibits M1 polarization, thus attenuating the production of inflammatory mediators. Moreover, we found that M1 microglia suppression may be related to the NLRP3 inflammasome and TLR4 signaling. These findings suggest that a better understanding of the multifaceted mechanisms of tianeptine action on microglia may increase the effectiveness of therapy, where inflammation is a central hallmark.
Collapse
|
50
|
Specific Surface Modifications of Silica Nanoparticles Diminish Inflammasome Activation and In Vivo Expression of Selected Inflammatory Genes. NANOMATERIALS 2017; 7:nano7110355. [PMID: 29084176 PMCID: PMC5707572 DOI: 10.3390/nano7110355] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 10/22/2017] [Accepted: 10/23/2017] [Indexed: 02/05/2023]
Abstract
Silica (SiO2) nanoparticles (NPs) usage includes, but is not limited to, industrial and biomedical applications. Toxic effects of SiO2 NPs have been explored either in vitro or in vivo, assessing different surface modifications to reduce their harmful effects. Here, murine bone marrow-derived dendritic (BMDC) and a mouse model of mild allergic inflammation were used to study inflammasome activation and lung inflammation. Our results showed that SiO2 plain NPs induced NACHT, LRR and PYD domains-containing protein 3 (NLRP3) inflammasome activation, increasing interleukin (IL)-1β release in vitro, and, to a lesser extent, in vivo. In addition, SiO2 plain NPs triggered a pulmonary inflammatory milieu in both non-sensitized (NS) and sensitized (S) mice, by inducing the expression of key inflammatory cytokines and chemokines. Electron microscopy showed that SiO2 NPs were mostly localized in alveolar macrophages, within vesicles and/or in phagolysosomes. Both the in vitro and the in vivo effects of SiO NPs were attenuated by coating NPs with phosphonate or amino groups, whereas PEGylation, although it mitigated inflammasome activation in vitro, was not a successful coating strategy in vivo. These findings highlight that multiple assays are required to determine the effect of surface modifications in limiting NPs inflammatory potential. Taken together, these data are obtained by comparing in vitro and in vivo effects of SiO2 NPs suggest the use of amino and phosphonate coating of silica NPs for commercial purposes and targeted applications, as they significantly reduce their proinflammatory potential.
Collapse
|