1
|
Güvenç M, Aydin T, Kutlu T, Etyemez M, İşler CT. Tomentosin mitigates the LPS induced cardiac injury by regulating Nrf-2/Nf-κβ pathway in mice. Eur J Pharmacol 2025; 996:177589. [PMID: 40187600 DOI: 10.1016/j.ejphar.2025.177589] [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: 10/28/2024] [Revised: 03/26/2025] [Accepted: 04/02/2025] [Indexed: 04/07/2025]
Abstract
Endotoxemic shock is a severe complication characterized by multiple organ failure, hypotension, and impaired tissue perfusion, all contributing to high morbidity and mortality. Recent studies have highlighted the anti-inflammatory and antioxidant properties of tomentosin. This study investigates the protective effects of tomentosin against lipopolysaccharide (LPS)-induced cardiac injury and elucidates its underlying mechanisms. Mice were pre-treated with tomentosin before the LPS administration. Subsequently, cardiac injury markers, oxidative stress parameters, inflammatory mediators, and Nrf-2/NF-κB protein expression levels were analysed. The results demonstrated that tomentosin significantly reduced Troponin and CK-MB levels, alleviated oxidative stress, and suppressed inflammatory responses. Furthermore, tomentosin inhibited NF-κB activation while enhancing Nrf-2 expression. In conclusion, our findings suggest that tomentosin exerts cardioprotective effects by modulating the Nrf-2/NF-κB pathway, positioning it as a potential therapeutic candidate for preventing LPS-induced cardiac dysfunction.
Collapse
Affiliation(s)
- Mehmet Güvenç
- Department of Physiology, Faculty of Veterinary Medicine, University of Hatay Mustafa Kemal, Hatay, 31060, Turkey.
| | - Tuba Aydin
- Department of Pharmacognosy, Faculty of Pharmacy, Ağrı İbrahim Çeçen University, Ağrı, 04100, Turkey
| | - Tuncer Kutlu
- Department of Pathology, Faculty of Veterinary Medicine, University of Hatay Mustafa Kemal, Hatay, 31060, Turkey
| | - Muhammed Etyemez
- Department of Physiology, Faculty of Veterinary Medicine, University of Kastamonu, Kastamonu, 37150, Turkey
| | - Cafer Tayer İşler
- Department of Surgery, Faculty of Veterinary Medicine, University of Hatay Mustafa Kemal, Hatay, 31060, Turkey
| |
Collapse
|
2
|
Zhang YT, Li HH, Teng F. Inhibition of Piezo1 ameliorates septic cardiomyopathy by blocking calcium-dependent PANoptosis. Eur J Pharmacol 2025; 996:177438. [PMID: 40020983 DOI: 10.1016/j.ejphar.2025.177438] [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: 10/20/2024] [Revised: 02/14/2025] [Accepted: 02/26/2025] [Indexed: 03/03/2025]
Abstract
Sepsis-induced cardiomyopathy (SIC) represents a severe and often fatal complication of sepsis, characterized by significant mortality. Despite extensive research, the underlying mechanisms remain incompletely understood. Recent studies have highlighted PANoptosis, an emerging form of programmed cell death, as a critical factor in inflammatory diseases. Piezo1, a mechanosensitive ion channel, has been implicated in various pathological conditions; however, its role in SIC and its involvement in PANoptosis require further investigation. In this study, the role of Piezo1 in SIC and calcium-dependent PANoptosis were investigated. SIC was induced in mice via cecal ligation and puncture (CLP), and the effects of Piezo1 inhibition on cardiac function, histological changes, mitochondrial function, and PANoptosis were assessed. Our results show that sepsis upregulates Piezo1 expression in cardiomyocytes through TLR4-NF-κB signaling. Pharmacological blockade of Piezo1 with its inhibitor GsMTx4 attenuated CLP-induced cardiac injury, histological damage, and mitochondrial dysfunction. Importantly, Piezo1 inhibition also significantly suppressed PANoptosis in septic hearts. In vitro experiments with Piezo1 siRNA, GsMTx4 and the calcium chelator BAPTA confirmed that inhibition of Piezo1 attenuates LPS-induced PANoptosis by limiting calcium release in cardiomyocytes after LPS treatment, linking Piezo1 to the regulation of these key events. Collectively, these findings reveal Piezo1 as a novel mechanosensor for sepsis and reveal a previously unrecognized role of Piezo1 in the activation of calcium-mediated PANoptosis in SIC. Given the ability of Piezo1 inhibition to mitigate key pathological features of SIC, targeting Piezo1 represents a promising therapeutic strategy for improving the outcomes of sepsis-related cardiac dysfunction.
Collapse
Affiliation(s)
- Yan-Ting Zhang
- Medical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Hui-Hua Li
- Medical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China.
| | - Fei Teng
- Emergency Medicine Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China.
| |
Collapse
|
3
|
Mestriner F, Dantas PB, Michelon-Barbosa J, Dugaich VF, Luis-Silva F, Ribeiro MS, Evora PR, Becari C. Methylene blue as a potential intervention in sepsis: Effects on survival and microcirculation in rat models of sepsis. Biomed Pharmacother 2025; 187:118131. [PMID: 40349555 DOI: 10.1016/j.biopha.2025.118131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 05/02/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025] Open
Abstract
Sepsis is a life-threatening condition characterized by systemic inflammation and microcirculatory dysfunction. Methylene blue (MB), a compound with known antioxidant and anti-inflammatory properties, has been proposed as a potential therapeutic agent. This study aimed to investigate the effects of MB on survival rates and the preservation of mesenteric microcirculation in a rat model of endotoxemia. A total of 36 rats underwent cecal ligation and puncture (CLP) surgery to induce varying degrees of sepsis: mild (4 perforations), moderate (10 perforations), and severe (20 perforations). Animals received intravenous treatment with either MB (4 mg/kg) or saline. Survival was monitored for ten days. Additionally, intravital microscopy was used to assess leukocyte rolling and adhesion in mesenteric vessels following lipopolysaccharide (LPS)-induced sepsis. The experimental groups included saline, LPS + saline, MB + saline, LPS + MB, and MB + LPS. MB treatment significantly improved survival in the severe sepsis group, with a 30 % survival rate at ten days (p = 0.02, 95 % CI: 0.12-0.48), whereas all animals in the severe sepsis + saline group died within nine days. No significant survival benefit was observed in the mild and moderate sepsis groups (mild sepsis: p = 0.45, 95 % CI: 0.08-0.34; moderate sepsis: p = 0.32, 95 % CI: 0.15-0.51). In the LPS-induced model, treatment with both LPS and MB significantly reduced leukocyte rolling and adhesion (p < 0.001, 95 % CI: 0.45-0.75 for rolling; p < 0.03, 95 % CI: 0.30-0.60 for adhesion), with values comparable to those of the control group. In contrast, MB alone had no effect on leukocyte rolling or adhesion.In summary, MB significantly improved survival in severe sepsis and inhibited leukocyte migration in mesenteric vessels. These findings suggest that MB may protect the microcirculation and enhance survival under severe septic conditions, representing a promising therapeutic approach for sepsis management.
Collapse
Affiliation(s)
- Fabiola Mestriner
- Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Pedro Brüch Dantas
- Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Jessyca Michelon-Barbosa
- Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Vinicius Flora Dugaich
- Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Fabio Luis-Silva
- Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil; Department of Clinical Medicine, Barao de Maua University Center, University of Ribeirão Preto, São Paulo, Brazil
| | - Mauricio S Ribeiro
- Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Paulo R Evora
- Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Christiane Becari
- Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil; Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Bauru, São Paulo, Brazil.
| |
Collapse
|
4
|
Elkhalil A, Whited A, Ghose P. SQST-1/p62-regulated SKN-1/Nrf mediates a phagocytic stress response via transcriptional activation of lyst-1/LYST. PLoS Genet 2025; 21:e1011696. [PMID: 40315422 PMCID: PMC12068719 DOI: 10.1371/journal.pgen.1011696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 05/12/2025] [Accepted: 04/19/2025] [Indexed: 05/04/2025] Open
Abstract
Cells may be intrinsically fated to die to sculpt tissues during development or to maintain homeostasis. Cells can also die in response to various stressors, injury or pathological conditions. Additionally, cells of the metazoan body are often highly specialized with distinct domains that differ both structurally and with respect to their neighbors. Specialized cells can also die, as in normal brain development or pathological states and their different regions may be eliminated via different programs. Clearance of different types of cell debris must be performed quickly and efficiently to prevent autoimmunity and secondary necrosis of neighboring cells. Moreover, all cells, including those programmed to die, may be subject to various stressors. Some largely unexplored questions include whether predestined cell elimination during development could be altered by stress, if adaptive stress responses exist and if polarized cells may need compartment-specific stress-adaptive programs. We leveraged Compartmentalized Cell Elimination (CCE) in the nematode C. elegans to explore these questions. CCE is a developmental cell death program whereby three segments of two embryonic polarized cell types are eliminated differently. We have previously employed this in vivo genetic system to uncover a cell compartment-specific, cell non-autonomous clearance function of the fusogen EFF-1 in phagosome closure during corpse internalization. Here, we introduce an adaptive response that serves to aid developmental phagocytosis as a part of CCE during stress. We employ a combination of forward and reverse genetics, CRISPR/Cas9 gene editing, stress response assays and advanced fluorescence microscopy. Specifically, we report that, under heat stress, the selective autophagy receptor SQST-1/p62 promotes the nuclear translocation of the oxidative stress-related transcription factor SKN-1/Nrf via negative regulation of WDR-23. This in turn allows SKN-1/Nrf to transcribe lyst-1/LYST (lysosomal trafficking associated gene) which subsequently promotes the phagocytic resolution of the developmentally-killed internalized cell even under stress conditions.
Collapse
Affiliation(s)
- Aladin Elkhalil
- The University of Texas at Arlington, Arlington, Texas, United States of America
| | - Alec Whited
- The University of Texas at Arlington, Arlington, Texas, United States of America
| | - Piya Ghose
- The University of Texas at Arlington, Arlington, Texas, United States of America
| |
Collapse
|
5
|
Liu H, Wang L, Zhou J. Nrf2 and its signaling pathways in sepsis and its complications: A comprehensive review of research progress. Medicine (Baltimore) 2025; 104:e42132. [PMID: 40258745 PMCID: PMC12014120 DOI: 10.1097/md.0000000000042132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 03/30/2025] [Indexed: 04/23/2025] Open
Abstract
Sepsis is a life-threatening condition characterized by organ dysfunction resulting from a dysregulated host immune response to infection. It is associated with a high incidence, intricate pathophysiological mechanisms, and rapidly progressive severity, rendering it a leading cause of mortality among patients in intensive care units. The Nuclear Factor Erythroid 2-Related Factor 2 (Nrf2) is a transcription factor pivotal for maintaining cellular redox homeostasis by regulating the expression of antioxidant and cytoprotective genes. Emerging evidence suggests that activation of the Nrf2 signaling pathway attenuates sepsis-induced inflammatory responses, oxidative stress, and organ dysfunction, thereby improving clinical outcomes. These findings underscore the potential of Nrf2 as a therapeutic target, offering a promising avenue for the development of novel interventions aimed at mitigating the complications and improving the prognosis of sepsis.
Collapse
Affiliation(s)
- Huan Liu
- Department of Emergency Internal Medicine, Jining NO.1 People’s Hospital, Jining, PR China
| | - Lei Wang
- Department of Pulmonary and Critical Care Medicine, Jining NO.1 People’s Hospital, Jining, PR China
| | - Jinhua Zhou
- Department of Pulmonary and Critical Care Medicine, Jining NO.1 People’s Hospital, Jining, PR China
| |
Collapse
|
6
|
Zhao Y, Gai C, Yu S, Song Y, Gu B, Luo Q, Wang X, Hu Q, Liu W, Liu D, Wang Z. Liposomes-Loaded miR-9-5p Alleviated Hypoxia-Ischemia-Induced Mitochondrial Oxidative Stress by Targeting ZBTB20 to Inhibiting Nrf2/Keap1 Interaction in Neonatal Mice. Antioxid Redox Signal 2025; 42:512-528. [PMID: 39869050 DOI: 10.1089/ars.2024.0640] [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] [Indexed: 01/28/2025]
Abstract
Aims: Hypoxia ischemia (HI) is a leading cause of cerebral palsy and long-term neurological sequelae in infants. Given that mitochondrial dysfunction in neurons contributes to HI brain damage, this study aimed to investigate the regulatory role of miR-9-5p in mitochondrial function following HI injury. Results: Overexpression of miR-9-5p in HI mice or H2O2-exposed PC12 cells suppressed neuronal injury, associated with increased mitochondrial copy number, normalizing mitochondrial membrane potential, improved nuclear factor-erythroid factor 2-related factor 2 (Nrf2) activation, and downregulation of Keap1. This was mediated, in part, through the ability of this miR-9-5p to bind and regulate the transcriptional activity of zinc finger and BTB domain-containing protein 20 (ZBTB20). Further study suggested that the knockdown of ZBTB20 exerts neuroprotection by inhibiting Nrf2/Keap1 interaction to promote the translocation of Nrf2 from the cytoplasm to the nucleus and the consequent expression of antioxidant proteins. Notably, the protective effects of miR-9-5p overexpression against HI-induced mitochondrial damage were reversed by the Nrf2 inhibitor ML385. Finally, the utilization of liposomes for the delivery of miR-9-5p (miR-9-5p@Lip) presents a promising therapeutic strategy for the treatment of HI injury. Innovation: miR-9-5p is a potential therapeutic agent for ischemic stroke through its modulation of the ZBTB20/Nrf2/Keap1 signaling pathway, influencing mitochondrial function and antioxidant response. Furthermore, the use of liposomal delivery for miR-9-5p offers a promising therapeutic strategy for HI injury. Conclusion: Overexpression of miR-9-5p protects against cerebral HI injury by modulating mitochondrial function through the ZBTB20/Nrf2/Keap1 signaling pathway. Antioxid. Redox Signal. 42, 512-528. [Figure: see text].
Collapse
Affiliation(s)
- Yijing Zhao
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, People's Republic of China
| | - Chengcheng Gai
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, People's Republic of China
| | - Shuwen Yu
- Department of Medical Psychology and Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, People's Republic of China
| | - Yan Song
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, People's Republic of China
| | - Bing Gu
- Department of Medical Psychology and Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, People's Republic of China
| | - Qian Luo
- Department of Medical Psychology and Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, People's Republic of China
| | - Xixi Wang
- Department of Medical Psychology and Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, People's Republic of China
| | - Quan Hu
- The Affiliated Taian City Central Hospital of Qingdao University, Taian, People's Republic of China
| | - Weiyang Liu
- Jinan Xicheng Experimental High School, Jinan, People's Republic of China
| | - Dexiang Liu
- Department of Medical Psychology and Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, People's Republic of China
| | - Zhen Wang
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, People's Republic of China
| |
Collapse
|
7
|
Qi M, Wei J, Zhang M, Jiao C, He C, Sui L, Ma S, Mao Z, Pan X, Zhu X. THE CAUSAL ASSOCIATION OF CARDIOMETABOLIC DISEASES AND SEPSIS-RELATED OUTCOMES: A MENDELIAN RANDOMIZATION AND POPULATION STUDY. Shock 2025; 63:579-586. [PMID: 39965633 DOI: 10.1097/shk.0000000000002538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2025]
Abstract
ABSTRACT Objective: The causality between cardiometabolic disease (CMD) and sepsis has remained largely unknown. To elucidate this, we conducted a Mendelian randomization (MR) and population study. Methods: First, we used univariable and multivariable MR analyses to investigate causal associations between CMD and sepsis-related outcomes. We obtained genome-wide association study summary from both the MRC Integrative Epidemiology Unit and the FinnGen consortium. Subsequently, a two-step mediation MR analysis was performed to explore mediators. Afterward, we conducted an observational study using the Medical Information Mart for Intensive Care IV database, in which multivariable logistic regression models were utilized to examine the relationship between CMD and sepsis-related outcomes. Results: In the MR study, type 2 diabetes mellitus (OR = 1.058, 95% CI = 1.017-1.100, P = 0.005), obesity (OR = 1.113, 95% CI = 1.057-1.172, P < 0.001), and heart failure (HF) (OR = 1.178, 95% CI = 1.063-1.305, P = 0.002) were independently causally related to sepsis. Obesity (OR = 1.215, 95% CI = 1.027-1.437, P = 0.023) and HF (OR = 1.494, 95% CI = 1.080-2.065, P = 0.015) also showed independent causal associations with sepsis critical care admission. Mediation MR analysis identified 23 blood metabolites potentially causally linked to sepsis ( P < 0.05), yet none mediated the relationship between CMD and sepsis. In the observational study, we found associations between sepsis and several conditions including type 2 diabetes mellitus, obesity, hypertension, stroke, HF, and hyperlipidemia after adjusting for confounding factors. Moreover, hypertension, stroke, HF, coronary artery disease, and hyperlipidemia were linked to sepsis critical care admission. Conclusion: This study has, for the first time, revealed indicative evidence of a causal relationship between CMD and sepsis through observational and genetic evidence. Taken together, clinical attention to sepsis may be warranted among patients with CMD.
Collapse
Affiliation(s)
- Mengmeng Qi
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Jin Wei
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Meng Zhang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Chucheng Jiao
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Chang He
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Liutao Sui
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Shiyin Ma
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Zhi Mao
- Department of Critical Care Medicine, The First Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Xudong Pan
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xiaoyan Zhu
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
8
|
Zheng Y, Gu N, Qiu K, Tian F, Chen L, Chen Y, Zeng L. The role and mechanism study of Cxcl14 in chronic critically ill cardiac dysfunction. Biochem Biophys Res Commun 2025; 754:151525. [PMID: 40010139 DOI: 10.1016/j.bbrc.2025.151525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 02/18/2025] [Accepted: 02/21/2025] [Indexed: 02/28/2025]
Abstract
OBJECTIVE To investigate the role and mechanism of Cxcl14 in cardiac function impairment in chronic critical illness (CCI). METHOD Clinical data from CCI patients with heart failure and uncomplicated CCI cases were collected. Serum Cxcl14 concentrations were quantified via ELISA. A CCI heart function injury mouse model (CLP model) was established, and the heart function and morphological indicators was measured. The inflammatory infiltration of myocardial tissue, the Cxcl4 expression levels and myocardial fibrosis related proteins, and the alleviating effect of anti-Cxcl14 antibody on CCI cardiac dysfunction was detected. RESULT The serum level of Cxcl14 was found to exhibit a positive correlation with N-terminal pro-brain natriuretic peptide (NT-proBNP) levels and a negative correlation with left ventricular ejection fraction (LVEF) in patients with CCI. Subsequent investigations into the role of Cxcl14 in CCI-related cardiac dysfunction revealed significant findings. In the CLP model, Cxcl14 was shown to reduce both LVEF and left ventricular intramural shortening fraction (LVFS), while simultaneously increasing the expression of α-smooth muscle actin (α-SMA) and collagen III in myocardial tissue. Additionally, Cxcl14 elevated the levels of ED-1, myeloperoxidase (MPO), tumor necrosis factor-alpha (TNF-α), and interleukin-6 (IL-6), promoting inflammatory cell infiltration in the myocardium. Administration of an anti-Cxcl14 antibody mitigated these effects, providing a potential therapeutic intervention. CONCLUSION The expression of Cxcl14 was positively associated with the onset and severity of cardiac dysfunction in CCI patients. In the CLP mouse model, Cxcl14 aggravated myocardial injury and fibrosis, while promoting the infiltration of inflammatory cells into myocardial tissue.
Collapse
Affiliation(s)
- Yongke Zheng
- Department of Rehabilitation, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, 261 Huansha Road, 310006, Hangzhou, Zhejiang, China
| | - Nanyuan Gu
- Department of Respiratory Support, Hangzhou Geriatric Hospital, 50 Jingshen Road, 310022, Hangzhou, Zhejiang, China
| | - Kai Qiu
- Department of Respiratory Support, Hangzhou Geriatric Hospital, 50 Jingshen Road, 310022, Hangzhou, Zhejiang, China
| | - Fu Tian
- Department of Respiratory Support, Hangzhou Geriatric Hospital, 50 Jingshen Road, 310022, Hangzhou, Zhejiang, China
| | - Leifei Chen
- Department of Respiratory Support, Hangzhou Geriatric Hospital, 50 Jingshen Road, 310022, Hangzhou, Zhejiang, China
| | - Yang Chen
- Department of Respiratory Support, Hangzhou Geriatric Hospital, 50 Jingshen Road, 310022, Hangzhou, Zhejiang, China
| | - Longhuan Zeng
- Department of Respiratory Support, Hangzhou Geriatric Hospital, 50 Jingshen Road, 310022, Hangzhou, Zhejiang, China.
| |
Collapse
|
9
|
Liu D, Zhang Y, Bai B, Xiong X, Zhou Q, Shi R. Integration of single-cell RNA sequencing and network pharmacology to elucidate the effect of Yantiao Formula on alleviating ALI by regulating the polarization of alveolar macrophages. JOURNAL OF ETHNOPHARMACOLOGY 2025; 343:119436. [PMID: 39914692 DOI: 10.1016/j.jep.2025.119436] [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: 08/08/2024] [Revised: 01/19/2025] [Accepted: 01/31/2025] [Indexed: 02/14/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) has a high mortality rate and often occurs in sepsis. Yantiao Formula (YTF) is used effectively in clinic but its mechanism in the treatment of ALI induced by sepsis remains unelucidated. AIM OF THE STUDY This study aims to explore the potential molecular mechanisms of YTF in the treatment of sepsis-induced ALI. MATERIALS AND METHODS Using ACQUITY UPLC I-Class, the chemical components contained in YTF were characterized. The network pharmacology approach was used to predict the components and targets of YTF for treating sepsis-induce ALI. Single-cell RNA sequencing (scRNA-seq) was used to find changes in the lung microenvironment after CLP-induced sepsis. Experimental validation was also performed in vitro and in vivo. Using molecular docking, we speculated on the potential pharmacological substances of YTF. RESULTS We detected 596 ingredients in YTF and identified 7 absorbed prototypes in serum. 1031 targets for 596 components were retrieved through TCMSP and SwissTargetPrediction databases. 365 potential targets for YTF and sepsis were identified. We observed that the targets of YTF for sepsis were significantly enriched in TNF and chemokine related pathway using GO and KEGG analysis. It was confirmed that at different time points, different doses of YTF increased the CLP-induced PaO2, reduced PaCO2 levels and W/D ratio of lung tissue. CLP- decreased survival rates was also significantly improved by YTF. YTF reversed the increase of IL-6 and IL-1β caused by CLP. Using scRNA-seq analysis, we found that changes in the proportion of cell types and the polarization state of macrophages were evident. Furthermore, the altered levels of biomarkers (M1: IL-1β, iNOS and TNF- α; M2: CD206/Mrc1 and Arg-1) provided evidence of macrophages polarization. We found that CLP-challenged group presented enhanced iNOS and IL-1β expression and YTF increased CD206 and Arg-1 expression in CLP- induced sepsis using immunohistochemical analysis. Similarly, the same results were validated in LPS- induced ALI in NR8383 cells. The material basis and potential therapeutic targets of YTF were also demonstrated using molecular docking. CONCLUSIONS YTF reduced the release of inflammatory factors and attenuated sepsis-induced ALI. The combined application of scRNA-seq, network pharmacology and molecular docking was helpful for revealing the mechanism of YTF, which was related to altering levels of M1 and M2 biomarkers to regulate macrophage polarization. The role of YTF in exerting its effects was closely relevant to the potential binding targets of its absorbed prototypes.
Collapse
Affiliation(s)
- Deng Liu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yifei Zhang
- Department of Intensive Care Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Bufan Bai
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xudong Xiong
- Department of Intensive Care Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Qianmei Zhou
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, China.
| | - Rong Shi
- Department of Intensive Care Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
10
|
Liu D, Sun S, Qiao H, Xin Q, Zhou S, Li L, Song N, Zhang L, Chen Q, Tian F, Mu X, Zhang S, Zhang J, Guo M, Wang H, Zhang XD, Zhang R. Ce 12V 6 Clusters with Multi-Enzymatic Activities for Sepsis Treatment. Adv Healthc Mater 2025; 14:e2401581. [PMID: 39129228 DOI: 10.1002/adhm.202401581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 07/22/2024] [Indexed: 08/13/2024]
Abstract
Artificial enzymes, especially nanozymes, have attracted wide attention due to their controlled catalytic activity, selectivity, and stability. The rising Cerium-based nanozymes exhibit unique SOD-like activity, and Vanadium-based nanozymes always hold excellent GPx-like activity. However, most inflammatory diseases involve polymerase biocatalytic processes that require multi-enzyme activities. The nanocomposite can fulfill multi-enzymatic activity simultaneously, but large nanoparticles (>10 nm) cannot be excreted rapidly, leading to biosafety challenges. Herein, atomically precise Ce12V6 clusters with a size of 2.19 nm are constructed. The Ce12V6 clusters show excellent glutathione peroxidase (GPx) -like activity with a significantly lower Michaelis-Menten constant (Km, 0.0125 mM versus 0.03 mM of natural counterpart) and good activities mimic superoxide dismutase (SOD) and peroxidase (POD). The Ce12V6 clusters exhibit the ability to scavenge the ROS including O2 ·- and H2O2 via the cascade reactions of multi-enzymatic activities. Further, the Ce12V6 clusters modulate the proinflammatory cytokines including tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and interleukin-1β (IL-1β) and consequently rescue the multi-organ failure in the lipopolysaccharide (LPS)-induced sepsis mouse model. With excellent biocompatibility, the Ce12V6 clusters show promise in the treatment of sepsis.
Collapse
Affiliation(s)
- Di Liu
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Si Sun
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Huanhuan Qiao
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Qi Xin
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin Third Central Hospital, Tianjin, 300170, China
| | - Sufei Zhou
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Lingxia Li
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Nan Song
- Department of Physics, School of Science, Tianjin Chengjian University, Tianjin, 300384, 18, China
| | - Lijie Zhang
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Qi Chen
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
- The First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Fangzhen Tian
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Xiaoyu Mu
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Shaofang Zhang
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Jing Zhang
- Department of Cardiology Tianjin Chest Hospital, Tianjin University, Tianjin, 300222, China
| | - Meili Guo
- Department of Physics, School of Science, Tianjin Chengjian University, Tianjin, 300384, 18, China
| | - Hao Wang
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Xiao-Dong Zhang
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
- Department of Physics and Tianjin Key Laboratory of Low Dimensional Materials Physics and Preparing Technology, School of Sciences, Tianjin University, Tianjin, 300350, China
| | - Ruiping Zhang
- The First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| |
Collapse
|
11
|
Sweet MJ, Ramnath D, Singhal A, Kapetanovic R. Inducible antibacterial responses in macrophages. Nat Rev Immunol 2025; 25:92-107. [PMID: 39294278 DOI: 10.1038/s41577-024-01080-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/05/2024] [Indexed: 09/20/2024]
Abstract
Macrophages destroy bacteria and other microorganisms through phagocytosis-coupled antimicrobial responses, such as the generation of reactive oxygen species and the delivery of hydrolytic enzymes from lysosomes to the phagosome. However, many intracellular bacteria subvert these responses, escaping to other cellular compartments to survive and/or replicate. Such bacterial subversion strategies are countered by a range of additional direct antibacterial responses that are switched on by pattern-recognition receptors and/or host-derived cytokines and other factors, often through inducible gene expression and/or metabolic reprogramming. Our understanding of these inducible antibacterial defence strategies in macrophages is rapidly evolving. In this Review, we provide an overview of the broad repertoire of antibacterial responses that can be engaged in macrophages, including LC3-associated phagocytosis, metabolic reprogramming and antimicrobial metabolites, lipid droplets, guanylate-binding proteins, antimicrobial peptides, metal ion toxicity, nutrient depletion, autophagy and nitric oxide production. We also highlight key inducers, signalling pathways and transcription factors involved in driving these different antibacterial responses. Finally, we discuss how a detailed understanding of the molecular mechanisms of antibacterial responses in macrophages might be exploited for developing host-directed therapies to combat antibiotic-resistant bacterial infections.
Collapse
Affiliation(s)
- Matthew J Sweet
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia.
| | - Divya Ramnath
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Amit Singhal
- Infectious Diseases Labs (ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Ronan Kapetanovic
- INRAE, Université de Tours, Infectiologie et Santé Publique (ISP), Nouzilly, France
| |
Collapse
|
12
|
von Stade D, Meyers M, Johnson J, Schlegel T, Romeo A, Regan D, McGilvray K. Primary Human Macrophage and Tenocyte Tendon Healing Phenotypes Changed by Exosomes Per Cell Origin. Tissue Eng Part A 2025. [PMID: 39761039 DOI: 10.1089/ten.tea.2024.0143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2025] Open
Abstract
The high failure rate of surgical repair for tendinopathies has spurred interest in adjunct therapies, including exosomes (EVs). Mesenchymal stromal cell (MSC)-derived EVs (MSCdEVs) have been of particular interest as they improve several metrics of tendon healing in animal models. However, research has shown that EVs derived from tissue-native cells, such as tenocytes, are functionally distinct and may better direct tendon healing. To this end, we investigated the differential regulation of human primary macrophage transcriptomic responses and cytokine secretion by tenocyte-derived EVs (TdEVs) compared with MSCdEVs. Compared with MSCdEVs, TdEVs upregulated TNFa-NFkB and TGFB signaling and pathways associated with osteoclast differentiation in macrophages while decreasing secretion of several pro-inflammatory cytokines. Conditioned media of these TdEV educated macrophages drove increased tenocyte migration and decreased MMP3 and MMP13 expression. In contrast, MSCdEV education of macrophages drove increased gene expression pathways related to INFa, INFg and protection against oxidative stress while increasing cytokine expression of MCP1 and IL6. These data demonstrate that EV cell source differentially impacts the function of key effector cells in tendon healing and that TdEVs, compared with MSCdEVs, promote a more favorable tendon healing phenotype within these cells.
Collapse
Affiliation(s)
- Devin von Stade
- Orthopaedic and Bioengineering Research Laboratory, Colorado State University, Fort Collins, Colorado, USA
| | - Melinda Meyers
- Animal Reproduction and Biotechnology Laboratory, Colorado State University, Fort Collins, Colorado, USA
| | - James Johnson
- Orthopaedic and Bioengineering Research Laboratory, Colorado State University, Fort Collins, Colorado, USA
| | | | - Anthony Romeo
- Shoulder Elbow Sports Medicine, Chicago, Illinois, USA
| | - Daniel Regan
- Flint Animal Cancer Center and Dept. of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | - Kirk McGilvray
- Orthopaedic and Bioengineering Research Laboratory, Colorado State University, Fort Collins, Colorado, USA
| |
Collapse
|
13
|
Edres HA, Elmassry IH, Lebda MA, Othman SI, El-Karim DRSG, Rudayni HA, Ebied SKM, Allam AA, Hashem AE. Berberine and Cyperus rotundus extract nanoformulations protect the rats against Staphylococcus-induced mastitis via antioxidant and anti-inflammatory activities: role of MAPK signaling. Cell Biochem Biophys 2024:10.1007/s12013-024-01628-8. [PMID: 39707026 DOI: 10.1007/s12013-024-01628-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/14/2024] [Indexed: 12/23/2024]
Abstract
Berberine (BER) and Cyperus rotundus rhizomes extract (CRE) are phytochemicals characterized by broad-spectrum pharmacological activity that could tackle the side effects of conventional mastitis therapies, however, they undergo a modest bioavailability. In the current study, nanoformulations of BER and CRE chitosan hydrogel (BER/CH-NPs, CRE/CH-NPs) were investigated for their antibacterial, antioxidant, anti-inflammatory and anti-apoptotic effects against S. aureus-induced mastitis in a rat model. The experiment was conducted on 80 early lactating female albino rats allocated into 6 groups; control, mastitis, BER/CH-NPs (1 and 0.5 mg), CRE/CH-NPs (0.5 and 0.25 mg), BER/CH-NPs + CRE/CH-NPs (0.5 + 0.25 and 0.25 + 0.125 mg). The nanoparticles were given by oral gavage once every other day from day 2 to day 12 after parturition. On the 13th day, intra-mammary inoculation with 100 µl of S. aureus suspension containing 2.1 × 108 CFU/ml in all groups except the control group. The results expressed the effect of BER/CH-NPs and CRE/CH-NPs on mammary gland tissue including significantly diminished viable bacterial load as well as attenuated the levels of MPO, MDA, caspase-3 with elevating Nrf2 level, and modulating glutathione redox. Also, the nanoformulations resulted in attenuation of the mRNA expression of TLR2, NOD2, Keap-1 and MAPK signaling pathway additional to the immune reactivity of NF-κB P65 and p-ERK as well as the preservation of the regular alveolar architecture. The supplementation of the berberine and Cyperus rotundus extract nanoformulations could be a prospective protective approach against Staphylococcal mastitis via their antibacterial, antioxidant, antiapoptotic, anti-inflammatory and modulation of MAPK signaling pathway.
Collapse
Affiliation(s)
- Hanan A Edres
- Department of Biochemistry, Faculty of Veterinary Medicine, Alexandria University, Alexandria, 21944, Egypt
| | - Ingi H Elmassry
- Department of Biochemistry, Faculty of Veterinary Medicine, Alexandria University, Alexandria, 21944, Egypt
| | - Mohamed A Lebda
- Department of Biochemistry, Faculty of Veterinary Medicine, Alexandria University, Alexandria, 21944, Egypt.
| | - Sarah I Othman
- Department of Biology, college of Science, Princess Nourah bint Abdulrahman University, P.O. BOX 84428, Riyadh, 11671, Saudi Arabia
| | - Dina R S Gad El-Karim
- Department of Pathology and Clinical Pathology, Faculty of Veterinary Medicine, Alexandria University, Alexandria, 21944, Egypt
| | - Hassan A Rudayni
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University, Riyadh, 11623, Saudi Arabia
| | - Sawsan Kh M Ebied
- Bacteriology Unit, Animal Health Research Institute, Alexandria Province, Alexandria, 21944, Egypt
| | - Ahmed A Allam
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University, Riyadh, 11623, Saudi Arabia
- Department of Zoology, Faculty of Science, Beni-suef University, Beni-suef, 65211, Egypt
| | - Aml E Hashem
- Department of Biochemistry, Faculty of Veterinary Medicine, Alexandria University, Alexandria, 21944, Egypt
| |
Collapse
|
14
|
Chen Q, Qiu FS, Xie W, Yu WY, Su ZA, Qin GM, Kang YK, Jiang SL, Yu CH. Gypenoside A-loaded mPEG-PLGA nanoparticles ameliorate high-glucose-induced retinal microvasculopathy by inhibiting ferroptosis. Int J Pharm 2024; 666:124758. [PMID: 39326476 DOI: 10.1016/j.ijpharm.2024.124758] [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: 06/14/2024] [Revised: 08/30/2024] [Accepted: 09/24/2024] [Indexed: 09/28/2024]
Abstract
Diabetic retinopathy (DR) is one of the chronic microvascular complications of type 2 diabetes mellitus (T2DM), which will cause retinal detachment and blindness without ideal therapies. Gypenoside A (GPA) are the main bioactive compound from Gynostemma pentaphyllum, and have various pharmacological effects. However, it suffered from poor bioavailability and potential cardiotoxicity in the clinical application. To overcome those limitations, in this study, nearly spherical nanoparticles (GPA-NP) with a mean particle size of 140.6 ± 22.4 nm were prepared by encapsulating GPA into mPEG-PLGA. This encapsulation efficiency was 84.4 ± 6.9 %, and the drug load was 4.02 %±0.35 %. The results showed that GPA-NP displayed more prolonged GPA release and higher bioavailability in vitro than GPA. GPA-NP obviously reduced the levels of oxidative stress markers and inflammatory cytokines in both retinal tissues of DR mice and high glucose-exposed HRMEC better than GPA alone. Mechanismly, GPA blocked the Nrf2-Keap1 interaction by binding with Kelch domain of Keap1 via alkyl and hydrogen bonds. Therefore, GPA-NP exerted more potent protectivity effects against high glucose-induced retinal microvascular endothelial ferroptosis in vitro and in vivo by activating Nrf2/HO-1/GPX4 pathway. It could be a promising therapeutic agent for preventing DR.
Collapse
Affiliation(s)
- Qin Chen
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China; Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou 310003, China
| | - Fen-Sheng Qiu
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou 310018, China; Key Laboratory of Experimental Animal and Safety Evaluation, Hangzhou Medical College, Hangzhou 310013, China
| | - Wei Xie
- Key Laboratory of Experimental Animal and Safety Evaluation, Hangzhou Medical College, Hangzhou 310013, China
| | - Wen-Ying Yu
- Key Laboratory of Experimental Animal and Safety Evaluation, Hangzhou Medical College, Hangzhou 310013, China
| | - Zhao-An Su
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China; Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou 310003, China
| | - Guang-Ming Qin
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China; Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou 310003, China.
| | - You-Kun Kang
- Key Laboratory of Experimental Animal and Safety Evaluation, Hangzhou Medical College, Hangzhou 310013, China
| | - Song-Lin Jiang
- Key Laboratory of Experimental Animal and Safety Evaluation, Hangzhou Medical College, Hangzhou 310013, China
| | - Chen-Huan Yu
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou 310018, China.
| |
Collapse
|
15
|
Guo N, Xia Y, He N, Cheng H, Zhang L, Liu J. IRGM Deficiency Exacerbates Sepsis-Induced Acute Lung Injury by Inhibiting Autophagy Through the AKT/mTOR Signaling Pathway. J Inflamm Res 2024; 17:10255-10272. [PMID: 39654860 PMCID: PMC11626208 DOI: 10.2147/jir.s496687] [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: 09/17/2024] [Accepted: 11/23/2024] [Indexed: 12/12/2024] Open
Abstract
Background Sepsis is a life-threatening condition characterized by organ dysfunction due to an impaired immune response to infection. The lungs are highly susceptible to infection, often resulting in acute lung injury (ALI). The immune-related GTPase M (IRGM) and its murine homolog Irgm1 mediate autophagy and are implicated in inflammatory diseases, yet their roles in sepsis-induced ALI remain unclear. Methods We used RNA sequencing and bioinformatics to explore IRGM regulation. Sepsis-induced ALI was modeled in mice using cecal ligation and puncture (CLP). An in vitro model was created by stimulating A549 cells with lipopolysaccharide (LPS). Results In A549 cells, LPS treatment induced upregulation of IRGM expression and enhanced autophagy levels. IRGM knockdown exacerbated LPS-induced ALI, characterized by suppressed autophagy and increased apoptosis, along with significantly elevated levels of p-AKT and p-mTOR. Further investigation revealed that treatment with the AKT inhibitor MK2206 effectively reversed the autophagy inhibition caused by IRGM knockdown and reduced apoptosis. These findings suggest that the AKT/mTOR signaling pathway plays a crucial role in IRGM-mediated protection against sepsis-related ALI. Conclusion This study identifies the protective role of IRGM in sepsis-induced ALI and reveals that IRGM mitigates ALI by promoting autophagy through inhibition of the AKT/mTOR pathway. These findings provide insights into the pathogenesis of sepsis-related ALI and highlight IRGM as a potential therapeutic target.
Collapse
Affiliation(s)
- Na Guo
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu Province, People’s Republic of China
| | - Yu Xia
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu Province, People’s Republic of China
| | - Nannan He
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu Province, People’s Republic of China
| | - Huixin Cheng
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu Province, People’s Republic of China
| | - Lei Zhang
- Gansu Provincial Maternity and Child-Care Hospital (Gansu Provincial Center Hospital), Lanzhou, Gansu Province, People’s Republic of China
| | - Jian Liu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu Province, People’s Republic of China
- Gansu Provincial Maternity and Child-Care Hospital (Gansu Provincial Center Hospital), Lanzhou, Gansu Province, People’s Republic of China
| |
Collapse
|
16
|
Wang C, Wang P, Fu J, Yang Z, Du H, Zhang M, Zhang S, Shen M, Wang T, Wei K, Zhu R. Pinus massoniana pollen polysaccharides alleviate LPS-induced myocardial injury through p110β-mediated inhibition of the PI3K/AKT/NFκB pathway. Int J Biol Macromol 2024; 283:137713. [PMID: 39551317 DOI: 10.1016/j.ijbiomac.2024.137713] [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: 06/06/2024] [Revised: 11/04/2024] [Accepted: 11/14/2024] [Indexed: 11/19/2024]
Abstract
The inflammatory response is the core of the pathogenesis of lipopolysaccharide (LPS)-induced sepsis myocardial injury (SMI). Pinus massoniana pollen polysaccharide (PPPS) is a natural polymer with known biological activities, including anti-inflammatory, antioxidant, and antiviral properties. In this study, we aimed to investigate the impact of PPPS on SMI, myocardial enzyme levels, pathological changes, oxidative stress, cell apoptosis, and related signaling pathways in LPS-induced SMI models were observed by hematoxylin-eosin (HE) staining, immunohistochemical (IHC) staining, qPCR, Western blot analysis, with a particular focus on anti-inflammatory effects of PPPS. Animal experiments have shown that PPPS mitigates myocardial tissue injury and suppresses the inflammatory response. In the cellular experiments, PPPS protected H9c2 cells from LPS/adenosine triphosphate (ATP)-induced injury and inflammation. Transcriptome analysis and cardiomyocyte validation revealed that PPPS inhibited activation of the PI3K/PTEN/AKT signaling pathway. Additionally, intervention with the PI3K/PTEN/AKT signaling pathway activator counteracted the anti-inflammatory effects of PPPS. Further investigations indicated that PPPS exerts its anti-inflammatory effects in SMI by enhancing the expression of p110β protein and facilitating its interaction with PTEN, thereby inhibiting activation of the PI3K/AKT/NFκB signaling pathway. These results suggest that PPPS is a promising therapeutic agent for the treatment of SMI.
Collapse
Affiliation(s)
- Cheng Wang
- Shandong Provincial Key Laboratory of Zoonoses, College of Veterinary Medicine, Shandong Agricultural University, Tai'an, Shandong 271018, China; Shandong Academy of Chinese Medicine, Jinan, Shandong 250014, China
| | - Ping Wang
- Shandong Academy of Chinese Medicine, Jinan, Shandong 250014, China
| | - Jialei Fu
- Shandong Academy of Chinese Medicine, Jinan, Shandong 250014, China
| | - Zongtong Yang
- Shandong Academy of Chinese Medicine, Jinan, Shandong 250014, China
| | - Haitao Du
- Shandong Academy of Chinese Medicine, Jinan, Shandong 250014, China
| | - Mengru Zhang
- Shandong Academy of Chinese Medicine, Jinan, Shandong 250014, China
| | - Shuyu Zhang
- Shandong Provincial Key Laboratory of Zoonoses, College of Veterinary Medicine, Shandong Agricultural University, Tai'an, Shandong 271018, China
| | - Mingyue Shen
- Shandong Provincial Key Laboratory of Zoonoses, College of Veterinary Medicine, Shandong Agricultural University, Tai'an, Shandong 271018, China
| | - Tingting Wang
- Weifang Hospital of Traditional Chinese Medicine, Weifang, Shandong 261000, China
| | - Kai Wei
- Shandong Provincial Key Laboratory of Zoonoses, College of Veterinary Medicine, Shandong Agricultural University, Tai'an, Shandong 271018, China.
| | - Ruiliang Zhu
- Shandong Provincial Key Laboratory of Zoonoses, College of Veterinary Medicine, Shandong Agricultural University, Tai'an, Shandong 271018, China.
| |
Collapse
|
17
|
Bao-Yuan H, Shu-Ru L, Le-Xin C, Liang-Liang B, Cheng-Cheng L, Chun-Qi X, Ming-Jun L, Jia-Xin Z, En-Xin Z, Xiao-Jun Z. Shikonin ameliorated LPS-induced acute lung injury in mice via modulating MCU-mediated mitochondrial Ca 2+ and macrophage polarization. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156043. [PMID: 39366155 DOI: 10.1016/j.phymed.2024.156043] [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: 05/30/2024] [Revised: 08/28/2024] [Accepted: 09/11/2024] [Indexed: 10/06/2024]
Abstract
BACKGROUND Macrophages play a pivotal role in the development and recovery of acute lung injury (ALI), wherein their phenotypic differentiation and metabolic programming are orchestrated by mitochondria. Specifically, the mitochondrial calcium uniporter (MCU) regulates mitochondrial Ca2+ (mCa2+) uptake and may bridge the metabolic reprogramming and functional regulation of immune cells. However, the precise mechanism on macrophages remains elusive. Shikonin, a natural naphthoquinone, has demonstrated efficacy in mitigating ALI and suppressing glycolysis in macrophages, yet which mechanism remains to be fully elucidated. PURPOSE This study explored whether Shikonin ameliorated ALI via modulating MCU-mediated mCa2+ and macrophage polarization. METHODS This study firstly examined the protective effects of Shikonin on LPS-induced ALI mice, and investigated whether it is depends on macrophage by depleting macrophage using clodronate liposomes. The regulatory effect of Shikonin on macrophage polarization and mitochondrial MCU/Ca2+ signal was testified on RAW264.7 cells, and further validated by knocking-down MCU expression or by using RU360, an MCU inhibitor. Additionally, the crucial role of MCU in the therapeutic effect of Shikonin, along with its regulation on macrophage polarization was validated in mice with LPS-induced ALI under the intervention of RU360. RESULTS Shikonin alleviated LPS-induced mice ALI, down-regulated inflammatory cytokines and inhibited the pro-inflammatory polarization of macrophages. Intravenous injection of clodronate liposomes on mice abolished the protective effects of Shikonin on ALI. On RAW264.7 cells, LPS&IFN decreased the protein expression of MCU, while induced pro-inflammatory polarization and glycolytic metabolism. In contrast, Shikonin increased MCU expression, activated MCU-mediated mCa2+ signal, promoted the polarization of macrophages to anti-inflammatory M2 phenotype, and driven a metabolic shift from glycolysis to oxidative phosphorylation. Either knocking-down MCU expression or pharmacological inhibiting MCU by using RU360 mitigated the effects of Shikonin on Raw 264.7 cells. Furthermore, RU360 counteracted the ameliorative effect of Shikonin on ALI mice. CONCLUSION The current data showed that Shikonin alleviated LPS-induced mice ALI by activating mitochondrial MCU/mCa2+ signal and regulating macrophage metabolism.
Collapse
Affiliation(s)
- Huang Bao-Yuan
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, China; The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong Province Lingnan Characteristic Hospital Preparation Transformation Engineering Technology Research Center, Guangzhou, China
| | - Lu Shu-Ru
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, China; Affiliated Dongguan Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan, China
| | - Chen Le-Xin
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Bai Liang-Liang
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Li Cheng-Cheng
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xu Chun-Qi
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Li Ming-Jun
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zeng Jia-Xin
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhang En-Xin
- Shenzhen Bao'an Authentic TCM Therapy Hospital, Shenzhen, China.
| | - Zhang Xiao-Jun
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
18
|
Tan YT, Li T, Wang RB, Liu ZK, Ma MY, Huang RZ, Mo HY, Luo SY, Lin JF, Xu RH, Ju HQ. WTAP weakens oxaliplatin chemosensitivity of colorectal cancer by preventing PANoptosis. Cancer Lett 2024; 604:217254. [PMID: 39270768 DOI: 10.1016/j.canlet.2024.217254] [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: 06/11/2024] [Revised: 08/17/2024] [Accepted: 09/09/2024] [Indexed: 09/15/2024]
Abstract
As the most abundant post-transcriptional modification in eukaryotes, N6-methyladenosine (m6A) plays a crucial role in cancer cell proliferation, invasion and chemoresistance. However, its specific effects on chemosensitivity to oxaliplatin-based regimens and the impact of these drugs on m6A methylation levels in colorectal cancer (CRC) remain largely unexplored. In this study, we demonstrated that the m6A methyltransferase Wilms tumor 1-associating protein (WTAP) weakens oxaliplatin chemosensitivity in HCT116 and DLD1 cells. Mechanistically, oxaliplatin treatment upregulated WTAP expression, preventing multiple forms of cell death simultaneously, a process known as PANoptosis, by decreasing intracellular oxidative stress through maintaining the expression of nuclear factor erythroid-2-related factor 2 (NRF2), a major antioxidant response element, in an m6A-dependent manner. In addition, high WTAP expression in CRC patients is associated with a poor prognosis and reduced benefit from standard chemotherapy by clinical data analysis of The Cancer Genome Atlas (TCGA) database and patient cohort study. These findings suggest that targeting WTAP-NRF2-PANoptosis axis could enhance the antitumor efficacy of oxaliplatin-based chemotherapy in CRC treatment.
Collapse
Affiliation(s)
- Yue-Tao Tan
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, 510060, PR China
| | - Ting Li
- Department of Gastroenterology and Urology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, 410013, PR China
| | - Ruo-Bing Wang
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Jinan University, Guangzhou, 510632, PR China
| | - Ze-Kun Liu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, 510060, PR China; Department of Radiology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Meng-Yao Ma
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Jinan University, Guangzhou, 510632, PR China
| | - Ren-Ze Huang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, 510060, PR China
| | - Hai-Yu Mo
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, 510060, PR China
| | - Shu-Yu Luo
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, 510060, PR China
| | - Jin-Fei Lin
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, 510060, PR China; Department of Clinical Laboratory, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, PR China.
| | - Rui-Hua Xu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, 510060, PR China; Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, 510060, PR China.
| | - Huai-Qiang Ju
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, 510060, PR China.
| |
Collapse
|
19
|
Chen X, Yang Z, Liao M, Zhao Q, Lu Y, Li Q, Liu S, Li S, Chen J, He Y. Ginkgo Flavone Aglycone Ameliorates Atherosclerosis via Inhibiting Endothelial Pyroptosis by Activating the Nrf2 Pathway. Phytother Res 2024; 38:5458-5473. [PMID: 39322309 DOI: 10.1002/ptr.8321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 07/26/2024] [Accepted: 08/05/2024] [Indexed: 09/27/2024]
Abstract
Natural antioxidants have been shown to be effective against atherosclerosis. Ginkgo flavone aglycone (GA) has strong antioxidant properties and can protect against endothelial damage. However, the mechanisms by which GA protects against atherosclerosis remain largely unexplored. This study hopes to find the anti-atherosclerotic mechanism of GA. ApoE-/- mice fed a high-fat diet were used for modeling atherosclerosis. The efficacy of GA on mice with atherosclerosis was evaluated based on the following indicators: Oil Red O staining, Masson staining, lipid content, and apoptosis. Transmission electron microscopy, Western blot, immunofluorescence staining, and propidium iodide staining were used to analyze the effects of GA on ox-LDL-treated human aortic endothelial cells. GA activated Nrf2 by promoting the nuclear translocation of Nrf2, thereby inhibiting endothelial pyroptosis. GA prevented endothelial pyroptosis suppressed oxidative stress, and inhibited the development of atherosclerosis in ApoE-/- mice fed high-fat diets. At the cellular level, GA suppressed ox-LDL-induced pyroptosis of HAECs by reducing reactive oxygen species (ROS) levels and inhibiting NLRP3 inflammasome. Furthermore, siRNA targeting Nrf2 or ML385, an Nrf2 inhibitor, reversed these effects. GA liberated Nrf2 from Keap1 sequestration, enhanced the nuclear translocation of Nrf2 and the transcription of downstream antioxidant proteins, reinforced the antioxidant defense system, and inhibited oxidative stress, thereby preventing endothelial cell pyroptosis, and attenuating the progression of atherosclerosis. This study indicated that GA mitigated endothelial pyroptosis by modulating Keap1/Nrf2 interactions, shedding light on the potential mechanisms underlying the protective effects of natural antioxidants against atherosclerosis.
Collapse
Affiliation(s)
- Xingyi Chen
- Clinical Trials Center, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- School of Pharmacy, Guizhou Medical University, Guiyang, China
| | - Zhuan Yang
- School of Pharmacy, Guizhou Medical University, Guiyang, China
| | - Meijuan Liao
- School of Pharmacy, Guizhou Medical University, Guiyang, China
| | - Qing Zhao
- School of Pharmacy, Guizhou Medical University, Guiyang, China
| | - Yuan Lu
- School of Pharmacy, Guizhou Medical University, Guiyang, China
| | - Qin Li
- Clinical Trials Center, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Shijing Liu
- Clinical Trials Center, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Shiliang Li
- Department of Vascular Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Jiyu Chen
- Clinical Trials Center, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- School of Pharmacy, Guizhou Medical University, Guiyang, China
| | - Yan He
- Clinical Trials Center, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- School of Pharmacy, Guizhou Medical University, Guiyang, China
| |
Collapse
|
20
|
Zhao Y, Zhou Z, Cui X, Yu Y, Yan P, Zhao W. Enhancing insight into ferroptosis mechanisms in sepsis: A genomic and pharmacological approach integrating single-cell sequencing and Mendelian randomization. Int Immunopharmacol 2024; 140:112910. [PMID: 39121604 DOI: 10.1016/j.intimp.2024.112910] [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: 05/14/2024] [Revised: 07/26/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
This research investigated the intricate relationship between ferroptosis and sepsis by utilizing advanced genomic and pharmacological methodologies. Specifically, we obtained expression quantitative trait loci (eQTLs) for 435 genes associated with ferroptosis from the eQTLGen Consortium and detected notable cis-eQTLs for 281 of these genes. Next, we conducted a detailed analysis to assess the impact of these eQTLs on susceptibility to sepsis using Mendelian randomization (MR) with data from a cohort of 10,154 sepsis patients and 452,764 controls sourced from the UK Biobank. MR analysis revealed 16 ferroptosis-related genes that exhibited significant associations with sepsis outcomes. To bolster the robustness of these findings, sensitivity analyses were performed to assess pleiotropy and heterogeneity, thus confirming the reliability of the causal inferences. Furthermore, single-cell RNA sequencing data from sepsis patients offered a detailed examination of gene expression profiles, demonstrating varying levels of ferroptosis marker expression across different cell types. Pathway enrichment analysis utilizing gene set enrichment analysis (GSEA) further revealed the key biological pathways involved in the progression of sepsis. Additionally, the use of computational molecular docking facilitated the prediction of interactions between identified genes and potential therapeutic compounds, highlighting novel drug targets. In conclusion, our integrated approach combining genomics and pharmacology offers valuable insights into the involvement of ferroptosis in sepsis, laying the groundwork for potential therapeutic strategies targeting this cell death pathway to enhance sepsis management.
Collapse
Affiliation(s)
- Yuanqi Zhao
- Department of Clinical Laboratory, School of Clinical Medicine, Dali University, Dali, China
| | - Zijian Zhou
- Department of Clinical Laboratory, School of Clinical Medicine, Dali University, Dali, China
| | - Xiuyu Cui
- Department of Clinical Laboratory, School of Clinical Medicine, Dali University, Dali, China
| | - Yiwei Yu
- Department of Clinical Laboratory, School of Clinical Medicine, Dali University, Dali, China
| | - Ping Yan
- Department of Gastroenterology, First Affiliated Hospital of Dali University, Dali, China.
| | - Weidong Zhao
- Department of Clinical Laboratory, School of Clinical Medicine, Dali University, Dali, China; Department of Clinical Laboratory, Second Infectious Disease Hospital of Yunnan Province, Dali, China; Immunology Discipline Team, School of Basic Medicine, Dali University, Dali, China.
| |
Collapse
|
21
|
Gu B, Jiang Y, Huang Z, Li H, Yu W, Li T, Liu C, Wang P, Chen J, Sun L, Tan P, Fu W, Wen J. MRG15 aggravates sepsis-related liver injury by promoting PCSK9 synthesis and secretion. Int Immunopharmacol 2024; 140:112898. [PMID: 39128417 DOI: 10.1016/j.intimp.2024.112898] [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: 03/19/2024] [Revised: 07/22/2024] [Accepted: 08/05/2024] [Indexed: 08/13/2024]
Abstract
OBJECTIVE Disorders of lipid oxidation play an important role in organ damage, and lipid metabolites are associated with inflammation and coagulation dysfunction in sepsis. However, the specific molecular mechanism by which lipid metabolism-related proteins regulate sepsis is still unclear. The aim of this study is to investigate the role of mortality factor 4-like protein 1 (MORF4L1, also called MRG15), a hepatic lipid metabolism related gene, in sepsis-induced liver injury. METHODS In the mouse sepsis models established by cecal ligation and puncture (CLP) and lipopolysaccharide (LPS), the impact of pretreatment with the MRG15 inhibitor argatroban on sepsis-related liver injury was investigated. In the LPS-induced hepatocyte sepsis cell model, the effects of MRG15 overexpression or knockdown on hepatic inflammation and lipid metabolism were studied. Additionally, in a co-culture system of hepatocytes and macrophages, the influence of MRG15 knockdown in hepatocytes on the synthesis and secretion of inflammation-related protein PCSK9 as well as its effect on macrophage activation were examined. RESULTS Studies have shown that MRG15 expression was increased in septicemia mice and positively correlated with lipid metabolism and inflammation. However, knockdown of MRG15 ameliorates sepsis-induced hepatocyte injury. Increased MRG15 in LPS-stimulated hepatocytes promotes PCSK9 synthesis and secretion, which induces macrophage M1 polarization and exacerbates the inflammatory response. Agatroban, an inhibitor of MRG15, ameliorates sepsis-induced liver injury in mice by inhibiting MRG15-induced lipid metabolism disorders and inflammatory responses. CONCLUSIONS In sepsis, increased MRG15 expression in hepatocytes leads to disturbed hepatic lipid metabolism and induces macrophage M1 polarization by secreting PCSK9, ultimately exacerbating liver injury.
Collapse
Affiliation(s)
- Boyuan Gu
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Yu Jiang
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Zhiwei Huang
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Han Li
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Wenhao Yu
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Tongxi Li
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Chen Liu
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Pengru Wang
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Jiatong Chen
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Lei Sun
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Peng Tan
- Metabolic Hepatobiliary and Pancreatic Diseases Key Laboratory of Luzhou City, Academician (Expert) Workstation of Sichuan Province, Luzhou 646000, China
| | - Wenguang Fu
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China; Metabolic Hepatobiliary and Pancreatic Diseases Key Laboratory of Luzhou City, Academician (Expert) Workstation of Sichuan Province, Luzhou 646000, China.
| | - Jian Wen
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China; Metabolic Hepatobiliary and Pancreatic Diseases Key Laboratory of Luzhou City, Academician (Expert) Workstation of Sichuan Province, Luzhou 646000, China.
| |
Collapse
|
22
|
Jian W, Ma H, Hu Y, Zhang Q, Xu J, Jiang J, Zhu G, Gong Y. Oltipraz attenuated cerebral ischemia-reperfusion injury through inhibiting the oxidative stress and ferroptosis in mice. Int Immunopharmacol 2024; 140:112800. [PMID: 39096875 DOI: 10.1016/j.intimp.2024.112800] [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: 04/30/2024] [Revised: 07/11/2024] [Accepted: 07/23/2024] [Indexed: 08/05/2024]
Abstract
Oltipraz (OPZ) is a synthetic dithiolethione and is considered a novel activator of nuclear factor E2-related factor 2 (Nrf2). Increasing evidence indicates that Nrf2 protects against cerebral ischemia/reperfusion (I/R) injury by antagonizing ferroptosis and lipid peroxidation. However, the protective effects of OPZ on cerebral I/R injury remain to be elucidated. We investigated the in vitro and in vivo neuroprotective effects of OPZ. Mice were subjected to middle cerebral artery occlusion/reperfusion (MCAO/R) to construct an in vivo model and PC12 cells were exposed to oxygen and glucose deprivation/reoxygenation (OGD/R) to establish an in vitro model. OPZ administration reduced the infarct volume and brain water content, and alleviated the neurological deficit of MCAO/R mice. Moreover, OPZ ameliorated MCAO/R-induced oxidative stress by decreasing the levels of 4-HNE and MDA and increasing the activities of SOD and GSH. We also found that OPZ ameliorated MCAO/R-induced ferroptosis by increasing SLC7A11 and GPX4 protein expression and downregulating ACSL4 protein expression. Similarly, the in vitro results revealed that OGD/R-induced oxidative stress and ferroptosis. Finally, mechanistic analysis revealed that OPZ significantly upregulated the Nrf2 expression and Nrf2 knockout (Nrf2 KO) abolished the OPZ-mediated protective effects. Taken together, these findings demonstrate that OPZ ameliorates cerebral I/R injury by suppressing the oxidative stress and ferroptosis.
Collapse
Affiliation(s)
- Wenting Jian
- The First College of Clinical Medical Science, China Three Gorges University, China; Institute of Anesthesia and Critical Care Medicine, China Three Gorges University, China; Yichang Central People's Hospital, Hubei, China
| | - Huigai Ma
- The First College of Clinical Medical Science, China Three Gorges University, China; Yichang Central People's Hospital, Hubei, China
| | - Yang Hu
- The First College of Clinical Medical Science, China Three Gorges University, China; Institute of Anesthesia and Critical Care Medicine, China Three Gorges University, China; Yichang Central People's Hospital, Hubei, China
| | - Qingyun Zhang
- The First College of Clinical Medical Science, China Three Gorges University, China; Institute of Anesthesia and Critical Care Medicine, China Three Gorges University, China; Yichang Central People's Hospital, Hubei, China
| | - Jinfei Xu
- The First College of Clinical Medical Science, China Three Gorges University, China; Institute of Anesthesia and Critical Care Medicine, China Three Gorges University, China; Yichang Central People's Hospital, Hubei, China
| | - Jingjing Jiang
- The First College of Clinical Medical Science, China Three Gorges University, China; Institute of Anesthesia and Critical Care Medicine, China Three Gorges University, China; Yichang Central People's Hospital, Hubei, China
| | - Guohong Zhu
- The First College of Clinical Medical Science, China Three Gorges University, China; Yichang Central People's Hospital, Hubei, China
| | - Yuan Gong
- The First College of Clinical Medical Science, China Three Gorges University, China; Institute of Anesthesia and Critical Care Medicine, China Three Gorges University, China; Yichang Central People's Hospital, Hubei, China.
| |
Collapse
|
23
|
Müller J, Hartwig C, Sonntag M, Bitzer L, Adelmann C, Vainshtein Y, Glanz K, Decker SO, Brenner T, Weber GF, von Haeseler A, Sohn K. A novel approach for in vivo DNA footprinting using short double-stranded cell-free DNA from plasma. Genome Res 2024; 34:1185-1195. [PMID: 39271293 PMCID: PMC11444180 DOI: 10.1101/gr.279326.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 08/12/2024] [Indexed: 09/15/2024]
Abstract
Here, we present a method for enrichment of double-stranded cfDNA with an average length of ∼40 bp from cfDNA for high-throughput DNA sequencing. This class of cfDNA is enriched at gene promoters and binding sites of transcription factors or structural DNA-binding proteins, so that a genome-wide DNA footprint is directly captured from liquid biopsies. In short double-stranded cfDNA from healthy individuals, we find significant enrichment of 203 transcription factor motifs. Additionally, short double-stranded cfDNA signals at specific genomic regions correlate negatively with DNA methylation, positively with H3K4me3 histone modifications and gene transcription. The diagnostic potential of short double-stranded cell-free DNA (cfDNA) in blood plasma has not yet been recognized. When comparing short double-stranded cfDNA from patient samples of pancreatic ductal adenocarcinoma with colorectal carcinoma or septic with postoperative controls, we identify 136 and 241 differentially enriched loci, respectively. Using these differentially enriched loci, the disease types can be clearly distinguished by principal component analysis, demonstrating the diagnostic potential of short double-stranded cfDNA signals as a new class of biomarkers for liquid biopsies.
Collapse
Affiliation(s)
- Jan Müller
- Innovation Field In-vitro Diagnostics, Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, 70569 Stuttgart, Germany
- Max Perutz Labs, Vienna Biocenter Campus, 1030 Vienna, Austria
- University of Vienna, Max Perutz Labs, Department of Structural and Computational Biology, Center of Integrative Bioinformatics Vienna, 1030 Vienna, Austria
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, 1030 Vienna, Austria
| | - Christina Hartwig
- Innovation Field In-vitro Diagnostics, Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, 70569 Stuttgart, Germany
- Institute for Interfacial Engineering and Plasma Technology, University of Stuttgart, 70569 Stuttgart, Germany
| | - Mirko Sonntag
- Innovation Field In-vitro Diagnostics, Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, 70569 Stuttgart, Germany
- Interfaculty Graduate School of Infection Biology and Microbiology, Eberhard Karls University Tübingen, 72074 Tübingen, Germany
| | - Lisa Bitzer
- Innovation Field In-vitro Diagnostics, Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, 70569 Stuttgart, Germany
| | - Christopher Adelmann
- Innovation Field In-vitro Diagnostics, Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, 70569 Stuttgart, Germany
| | - Yevhen Vainshtein
- Innovation Field In-vitro Diagnostics, Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, 70569 Stuttgart, Germany
| | - Karolina Glanz
- Innovation Field In-vitro Diagnostics, Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, 70569 Stuttgart, Germany
| | - Sebastian O Decker
- Heidelberg University, Medical Faculty Heidelberg, Department of Anesthesiology, 69120 Heidelberg, Germany
| | - Thorsten Brenner
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany
| | - Georg F Weber
- Department of Surgery, Friedrich-Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Arndt von Haeseler
- Center of Integrative Bioinformatics Vienna, Max Perutz Labs, University of Vienna and Medical University of Vienna, Vienna BioCenter, 1030 Vienna, Austria
- University of Vienna, Faculty of Computer Science Bioinformatics and Computational Biology, 1090 Vienna, Austria
| | - Kai Sohn
- Innovation Field In-vitro Diagnostics, Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, 70569 Stuttgart, Germany;
| |
Collapse
|
24
|
Zhao Z, Wei G, Wang L, Jiang Y, Zhang X, Fang L, Du G, Kong L. Pretreatment with Dan-Shen-Yin granules alleviates ethanol-induced gastric mucosal damage in rats by inhibiting oxidative stress and apoptosis via Akt/Nrf2 signaling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155866. [PMID: 39053247 DOI: 10.1016/j.phymed.2024.155866] [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: 11/27/2023] [Revised: 07/04/2024] [Accepted: 07/08/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND Gastric ulcer (GU) is a common gastrointestinal disease with high morbidity that may be caused by various pathogenic factors. Dan-Shen-Yin (DSY), a traditional prescription, improves myocardial and gastrointestinal functions; however, its effect on GU and the underlying mechanisms requires further research. PURPOSE We aimed to evaluate the pharmacodynamics of DSY granules in GU using three different animal models and explore their potential mechanisms. METHODS DSY granules were manufactured and subjected to quality control by high-performance liquid chromatography (HPLC). Three GU models were established using ethanol, aspirin, or water immersion restraint combined with aspirin and examined using the Guth method and hematoxylin and eosin (H&E) staining. The effects of DSY granules on gastric mucosal glycoproteins and the release of defensive and aggressive factors in ethanol-induced GU were measured using periodic acid-Schiff (PAS) staining and ELISA. TUNEL staining and detection of apoptosis-related proteins were used to evaluate the role of DSY granules on apoptosis. Potential mechanisms were predicted using network pharmacology, molecular docking, and western blot to verify the related targets and pathways. RESULTS DSY granules were prepared for the first time and quality control standard was established. Pharmacodynamic evaluation indicated that DSY granules significantly reduced the GU index and gastric mucosal injury in the three GU models, and the GU inhibition rate of DSY granules was superior to omeprazole in ethanol-induced GU model (60.32 % vs. 21.96 %). Further studies in ethanol-induced GU model revealed that DSY granules increased the levels of the defensive factors (PGE2, NO, SOD, CAT, TAOC, and GSH) and decreased the levels of aggressive factors (MDA, TNF-α, and IL-1β), thereby inhibiting oxidative stress and inflammation, attenuating gastric mucosal injury. Moreover, the results of TUNEL staining and western blot showed that DSY granules suppressed apoptosis by reducing the ratios of Bax/Bcl-2 and cleaved-Caspase-3/Caspase-3. In addition, the results of network pharmacology and molecular docking suggested that the mechanisms of DSY granules against GU may be related to the Akt-related signaling pathway. Further study confirmed that DSY granules significantly reduced the ratio of p-Akt/Akt and promoted the expression of Nrf2 and NQO1, protecting the gastric mucosa. CONCLUSIONS Our results indicated that DSY granules had protective effects on GU caused by different mechanisms, especially ethanol-induced GU. DSY granules alleviated gastric mucosal damage by inhibiting oxidative stress, inflammation, and apoptosis, which may be associated with the regulation of Akt/Nrf2 signaling pathway. Therefore, DSY granules may be a promising drug for the treatment of GU.
Collapse
Affiliation(s)
- Ziyuan Zhao
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Centre for Pharmaceutical Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Guangyi Wei
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Centre for Pharmaceutical Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Longrui Wang
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Centre for Pharmaceutical Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Yinru Jiang
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Centre for Pharmaceutical Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Xinyi Zhang
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Centre for Pharmaceutical Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Lianhua Fang
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Centre for Pharmaceutical Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| | - Guanhua Du
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Centre for Pharmaceutical Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| | - Linglei Kong
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Centre for Pharmaceutical Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|
25
|
Zhuang M, Zhang X, Ji J, Zhang H, Shen L, Zhu Y, Liu X. Exosomal circ-0100519 promotes breast cancer progression via inducing M2 macrophage polarisation by USP7/NRF2 axis. Clin Transl Med 2024; 14:e1763. [PMID: 39107958 PMCID: PMC11303452 DOI: 10.1002/ctm2.1763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 06/29/2024] [Accepted: 07/02/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND Breast cancer (BC) is one of the most prevalent malignant tumours that threatens women health worldwide. It has been reported that circular RNAs (circRNAs) play an important role in regulating tumour progression and tumour microenvironment (TME) remodelling. METHODS Differentially expression characteristics and immune correlations of circRNAs in BC were verified using high-throughput sequencing and bioinformatic analysis. Exosomes were characterised by nanoparticle transmission electron microscopy and tracking analysis. The biological function of circ-0100519 in BC development was demonstrated both in vitro and in vivo. Western blotting, RNA pull-down, RNA immunoprecipitation, flow cytometry, and luciferase reporter were conducted to investigate the underlying mechanism. RESULTS Circ-0100519 was significant abundant in BC tumour tissues and related to poor prognosis. It can be encapsulated into secreted exosomes, thereby promoting BC cell invasion and metastasis via inducing M2-like macrophages polarisation.Mechanistically, circ-0100519 acted as a scaffold to enhance the interaction between the deubiquitinating enzyme ubiquitin-specific protease 7 (USP7) and nuclear factor-like 2 (NRF2) in macrophages, inducing the USP7-mediated deubiquitination of NRF2. Additionally, HIF-1α could function as an upstream effector to enhance circ-0100519 transcription. CONCLUSIONS Our study revealed that exosomal circ-0100519 is a potential biomarker for BC diagnosis and prognosis, and the HIF-1α inhibitor PX-478 may provide a therapeutic target for BC.
Collapse
Affiliation(s)
- Minyu Zhuang
- Breast Disease CenterThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsuP.R. China
| | - Xiaoqiang Zhang
- Department of Breast SurgeryCancer Hospital of the University of Chinese Academy of Science (Zhejiang Cancer Hospital)HangzhouChina
| | - Jie Ji
- Breast Disease CenterThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsuP.R. China
| | - Hongfei Zhang
- Department of Ultrasound in Medicine, Second Affiliated HospitalZhejiang University School of MedicineZhejiangChina
| | - Li Shen
- Department of General SurgeryThe Fourth Affiliated Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjingJiangsuChina
| | - Yanhui Zhu
- Breast Disease CenterThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsuP.R. China
| | - Xiaoan Liu
- Breast Disease CenterThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsuP.R. China
| |
Collapse
|
26
|
Ting KKY. Revisiting the role of hypoxia-inducible factors and nuclear factor erythroid 2-related factor 2 in regulating macrophage inflammation and metabolism. Front Cell Infect Microbiol 2024; 14:1403915. [PMID: 39119289 PMCID: PMC11306205 DOI: 10.3389/fcimb.2024.1403915] [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: 03/20/2024] [Accepted: 07/11/2024] [Indexed: 08/10/2024] Open
Abstract
The recent birth of the immunometabolism field has comprehensively demonstrated how the rewiring of intracellular metabolism is critical for supporting the effector functions of many immune cell types, such as myeloid cells. Among all, the transcriptional regulation mediated by Hypoxia-Inducible Factors (HIFs) and Nuclear factor erythroid 2-related factor 2 (NRF2) have been consistently shown to play critical roles in regulating the glycolytic metabolism, redox homeostasis and inflammatory responses of macrophages (Mφs). Although both of these transcription factors were first discovered back in the 1990s, new advances in understanding their function and regulations have been continuously made in the context of immunometabolism. Therefore, this review attempts to summarize the traditionally and newly identified functions of these transcription factors, including their roles in orchestrating the key events that take place during glycolytic reprogramming in activated myeloid cells, as well as their roles in mediating Mφ inflammatory responses in various bacterial infection models.
Collapse
Affiliation(s)
- Kenneth K. Y. Ting
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| |
Collapse
|
27
|
Bajgai B, Suri M, Singh H, Hanifa M, Bhatti JS, Randhawa PK, Bali A. Naringin: A flavanone with a multifaceted target against sepsis-associated organ injuries. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155707. [PMID: 38788393 DOI: 10.1016/j.phymed.2024.155707] [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: 01/31/2024] [Revised: 04/16/2024] [Accepted: 05/02/2024] [Indexed: 05/26/2024]
Abstract
BACKGROUND Sepsis causes multiple organ dysfunctions and raises mortality and morbidity rates through a dysregulated host response to infection. Despite the growing research interest over the last few years, no satisfactory treatment exists. Naringin, a naturally occurring bioflavonoid with vast therapeutic potential in citrus fruits and Chinese herbs, has received much attention for treating sepsis-associated multiple organ dysfunctions. PURPOSE The review describes preclinical evidence of naringin from 2011 to 2024, particularly emphasizing the mechanism of action mediated by naringin against sepsis-associated specific injuries. The combination therapy, safety profile, drug interactions, recent advancements in formulation, and future perspectives of naringin are also discussed. METHODS In vivo and in vitro studies focusing on the potential role of naringin and its mechanism of action against sepsis-associated organ injuries were identified and summarised in the present manuscript, which includes contributions from 2011 to 2024. All the articles were extracted from the Medline database using PubMed, Science Direct, and Web of Science with relevant keywords. RESULTS Research findings revealed that naringin modulates many signaling cascades, such as Rho/ROCK and PPAR/STAT1, PIP3/AKT and KEAP1/Nrf2, and IkB/NF-kB and MAPK/Nrf2/HO-1, to potentially protect against sepsis-induced intestinal, cardiac, and lung injury, respectively. Furthermore, naringin treatment exhibits anti-inflammatory, anti-apoptotic, and antioxidant action against sepsis harm, highlighting naringin's promising effects in septic settings. Naringin could be employed as a treatment against sepsis, based on studies on combination therapy, synergistic effects, and toxicological investigation that show no reported severe side effects. CONCLUSION Naringin might be a promising therapeutic approach for preventing sepsis-induced multiple organ failure. Naringin should be used alongside other therapeutic therapies with caution despite its great therapeutic potential and lower toxicity. Nonetheless, clinical studies are required to comprehend the therapeutic benefits of naringin against sepsis.
Collapse
Affiliation(s)
- Bivek Bajgai
- Laboratory of Neuroendocrinology, Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India
| | - Manisha Suri
- Laboratory of Neuroendocrinology, Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India
| | - Harshita Singh
- Laboratory of Neuroendocrinology, Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India
| | - Mohd Hanifa
- Laboratory of Neuroendocrinology, Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India
| | - Jasvinder Singh Bhatti
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Ghudda, Bathinda, India
| | - Puneet Kaur Randhawa
- Department of Pharmaceutical Sciences, Amritsar Group of Colleges, Amritsar, Punjab, 143001, India; Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, 32827, USA
| | - Anjana Bali
- Laboratory of Neuroendocrinology, Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India.
| |
Collapse
|
28
|
Chen Z, Yao H, Encarnacion AM, Jeong J, Choi Y, Park S, Lee S, Lee T. Novel Inhibitor of Keap1-Nrf2 Protein-Protein Interaction Attenuates Osteoclastogenesis In Vitro and Prevents OVX-Induced Bone Loss In Vivo. Antioxidants (Basel) 2024; 13:850. [PMID: 39061918 PMCID: PMC11273523 DOI: 10.3390/antiox13070850] [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: 06/24/2024] [Revised: 07/10/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Keap1 interacts with Nrf2 by assisting in its ubiquitination and subsequent proteolysis. By preventing ROS accumulation during RANKL-induced osteoclastogenesis, Nrf2 activation can prevent the differentiation of osteoclasts. Additionally, inhibiting the Keap1-Nrf2 PPI can be an effective strategy for triggering Nrf2 to regulate oxidative stress. Structure-based virtual screening was performed to discover a potentially novel Keap1-Nrf2 PPI inhibitor wherein KCB-F06 was identified. The inhibitory effects of KCB-F06 on osteoclastogenesis were investigated in vitro through TRAP staining and bone resorption assays. An ovariectomy-induced osteoporosis mouse model was applied to evaluate KCB-F06's therapeutic effects in vivo. Lastly, the underlying mechanisms were explored using real-time PCR, Western blotting, and co-IP assays. KCB-F06 was discovered as a novel Keap1-Nrf2 PPI inhibitor. As a result, the expression of antioxidants (HO-1 and NQO1) was suppressed, hence reducing ROS accumulation during osteoclastogenesis. Subsequently, this caused the inactivation of RANKL-induced IKB/NF-kB signaling. This eventually led to the downregulation of osteoclast-specific proteins including NFATc1, which is an essential transcription factor for osteoclastogenesis. These results demonstrated that Nrf2 activation in osteoclasts is a valuable tool for osteoclastic bone loss management. In addition, KCB-F06 presents as an alternative candidate for treating osteoclast-related bone diseases and as a novel small molecule that can serve as a model for further Keap1-NRF2 PPI inhibitor development.
Collapse
Affiliation(s)
- Zhihao Chen
- Department of Oral Biochemistry, Dental Science Research Institute, School of Dentistry, Chonnam National University, Gwangju 61186, Republic of Korea; (Z.C.); (S.P.)
| | - Hongyuan Yao
- Department of Interdisciplinary Program of Biomedical Engineering, School of Dentistry, Chonnam National University, Gwangju 61186, Republic of Korea; (H.Y.); (A.M.E.); (J.J.)
| | - Alessandra Marie Encarnacion
- Department of Interdisciplinary Program of Biomedical Engineering, School of Dentistry, Chonnam National University, Gwangju 61186, Republic of Korea; (H.Y.); (A.M.E.); (J.J.)
| | - Jujin Jeong
- Department of Interdisciplinary Program of Biomedical Engineering, School of Dentistry, Chonnam National University, Gwangju 61186, Republic of Korea; (H.Y.); (A.M.E.); (J.J.)
| | - Yunju Choi
- Department of Dental Bioscience, School of Dentistry, Chonnam National University, Gwangju 61186, Republic of Korea;
| | - Sangwook Park
- Department of Oral Biochemistry, Dental Science Research Institute, School of Dentistry, Chonnam National University, Gwangju 61186, Republic of Korea; (Z.C.); (S.P.)
| | - Sunwoo Lee
- Department of Chemistry, Chonnam National University, Gwangju 61186, Republic of Korea;
| | - Taehoon Lee
- Department of Oral Biochemistry, Dental Science Research Institute, School of Dentistry, Chonnam National University, Gwangju 61186, Republic of Korea; (Z.C.); (S.P.)
| |
Collapse
|
29
|
Lei X, Liu X, Yu J, Li K, Xia L, Su S, Lin P, Zhang D, Li Y. 3-methyladenine ameliorates acute lung injury by inhibiting oxidative damage and apoptosis. Heliyon 2024; 10:e33996. [PMID: 39055838 PMCID: PMC11269838 DOI: 10.1016/j.heliyon.2024.e33996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/28/2024] Open
Abstract
Background Acute lung injury (ALI) is a condition characterized by inflammation and oxidative damage. 3-methyladenine (3-MA) has great potential for regulating apoptosis, but its regulatory role in ALI is unknown. Methods Lipopolysaccharide (LPS)-treated mice and tert-butyl hydroperoxide (TBHP)-treated bronchial epithelial cells were used to simulate in vivo and in vitro ALI models, respectively. In vivo, lung injury was assessed by histopathological analysis and lung injury scoring. The total cell count, protein content, and inflammatory factors in bronchoalveolar lavage fluid (BALF) were examined. The level of apoptosis in lung tissue was assessed through TUNEL staining. In the vitro ALI model, cell viability and levels of reactive oxygen species and apoptosis were assessed. Results 3-MA pretreatment ameliorated lung injury, including intra-alveolar hemorrhage and inflammatory cell accumulation, both in vitro and in vivo. 3-MA pretreatment also decreased inflammatory factor levels in the BALF. 3-MA pretreatment alleviated oxidative damage, decreased reactive oxygen species levels, and attenuated morphological changes. TUNEL and Annexin V-FITC/PI staining revealed that pretreatment with 3-MA reduced the level of apoptosis. 3-MA pretreatment significantly decreased the expression of caspase-3 and Bax but increased the expression of Bcl-2 in ALI. Mechanistically, 3-MA pretreatment also affected the PKCα/NOX4 and Nrf2 pathways, which decreased the level of apoptosis in ALI. Conclusions 3-MA pretreatment inhibited inflammation and oxidative damage in ALI and inhibited apoptosis to mitigate ALI in part by inhibiting the PKCα/NOX4 pathway and activating the Nrf2 pathway. Based on these results, 3-MA might be a viable medication to treat with ALI.
Collapse
Affiliation(s)
- Xiong Lei
- The Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiling Liu
- The Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jia Yu
- The Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Kai Li
- The First School of Medicine, School of Information and Engineering, Wenzhou Medical University, Wenzhou, China
| | - Lijing Xia
- The Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shanshan Su
- The Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Pengcheng Lin
- The Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Dan Zhang
- The Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yuping Li
- The Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
30
|
Ying Q, Rong J, Hong M, Heng Z, Zhang Z, Xu Y. The emerging role of adaptor proteins in regulating innate immunity of sepsis. Pharmacol Res 2024; 205:107223. [PMID: 38797359 DOI: 10.1016/j.phrs.2024.107223] [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: 12/06/2023] [Revised: 05/18/2024] [Accepted: 05/19/2024] [Indexed: 05/29/2024]
Abstract
Sepsis is a life-threatening syndrome caused by a dysregulated immune response. A large number of adaptor proteins have been found to play a pivotal role in sepsis via protein-protein interactions, thus participating in inflammatory cascades, leading to the generation of numerous inflammatory cytokines, as well as oxidative stress and regulated cell death. Although available strategies for the diagnosis and management of sepsis have improved, effective and specific treatments are lacking. This review focuses on the emerging role of adaptor proteins in regulating the innate immunity of sepsis and evaluates the potential value of adaptor protein-associated therapeutic strategy for sepsis.
Collapse
Affiliation(s)
- Qiaoyu Ying
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Jiabing Rong
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Min Hong
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Zetao Heng
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Zhaocai Zhang
- Department of Intensive Care Unit, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China.
| | - Yinchuan Xu
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China.
| |
Collapse
|
31
|
Chen J, Ding W, Zhang Z, Li Q, Wang M, Feng J, Zhang W, Cao L, Ji X, Nie S, Sun Z. Shenfu injection targets the PI3K-AKT pathway to regulate autophagy and apoptosis in acute respiratory distress syndrome caused by sepsis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 129:155627. [PMID: 38696924 DOI: 10.1016/j.phymed.2024.155627] [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: 10/28/2023] [Revised: 03/30/2024] [Accepted: 04/09/2024] [Indexed: 05/04/2024]
Abstract
BACKGROUND Sepsis is a life-threatening organ dysfunction caused by an exaggerated response to infection. In the lungs, one of the most susceptible organs, this can manifest as acute respiratory distress syndrome (ARDS). Shenfu (SF) injection is a prominent traditional Chinese medicine used to treat sepsis. However, the exact mechanism of its action has rarely been reported in the literature. PURPOSE In the present study, we detected the protective effect of SF injection on sepsis-induced ARDS and explored its underlying mechanism. METHODS We investigated the potential targets and regulatory mechanisms of SF injections using a combination of network pharmacology and RNA sequencing. This study was conducted both in vivo and in vitro using a mouse model of ARDS and lipopolysaccharide (LPS)-stimulated MLE-12 cells, respectively. RESULTS The results showed that SF injection could effectively inhibit inflammation, oxidative stress, and apoptosis to alleviate LPS-induced ARDS. SF inhibited the PI3K-AKT pathway, which controls autophagy and apoptosis. Subsequently, MLE-12 cells were treated with 3-methyladenine to assess its effects on autophagy and apoptosis. Additional experiments were conducted by adding rapamycin, an mTOR antagonist, or SC79, an AKT agonist, to investigate the effects of SF injection on autophagy, apoptosis, and the PI3K-AKT pathway. CONCLUSION Overall, we found that SF administration could enhance autophagic activity, reduce apoptosis, suppress inflammatory responses and oxidative stress, and inhibit the PI3K-AKT pathway, thus ameliorating sepsis-induced ARDS.
Collapse
Affiliation(s)
- Juan Chen
- Department of Emergency Medicine, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210002, PR China; Department of Emergency Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, PR China; Department of Emergency Medicine, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, Jiangsu Province 221000, PR China
| | - Weichao Ding
- Department of Emergency Medicine, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210002, PR China; Department of Emergency Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, PR China; Department of Emergency Medicine, the Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, PR China
| | - Zhe Zhang
- Department of Emergency Medicine, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210002, PR China; Department of Medical Oncology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, PR China
| | - Quan Li
- Department of Emergency Medicine, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210002, PR China
| | - Mengmeng Wang
- Department of Emergency Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, PR China
| | - Jing Feng
- Department of Emergency Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, PR China
| | - Wei Zhang
- Department of Emergency Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, PR China
| | - Liping Cao
- Department of Emergency Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, PR China
| | - Xiaohang Ji
- Department of Emergency Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, PR China
| | - Shinan Nie
- Department of Emergency Medicine, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210002, PR China; Department of Emergency Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, PR China.
| | - Zhaorui Sun
- Department of Emergency Medicine, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210002, PR China; Department of Emergency Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, PR China.
| |
Collapse
|
32
|
Zhou P, Yang L, Li R, Yin Y, Xie G, Liu X, Shi L, Tao K, Zhang P. IRG1/itaconate alleviates acute liver injury in septic mice by suppressing NLRP3 expression and its mediated macrophage pyroptosis via regulation of the Nrf2 pathway. Int Immunopharmacol 2024; 135:112277. [PMID: 38788445 DOI: 10.1016/j.intimp.2024.112277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 05/13/2024] [Accepted: 05/14/2024] [Indexed: 05/26/2024]
Abstract
Sepsis, a systemic inflammatory response triggered by infection, has a considerably high mortality rate. However, effective prevention and intervention measures against sepsis remain insufficient. Therefore, this study aimed to investigate the mechanisms underlying the protective properties of immune response gene-1 (IRG1) and 4-Octyl itaconate (OI) during acute liver damage in mice with sepsis. A sepsis mouse model was established to compare wild-type and IRG1-/- groups. The impact of IRG1/Itaconate on pro- and anti-inflammatory cytokines was evaluated using J774A.1 cells. IRG1/Itaconate substantially reduced pro-inflammatory cytokines and increased the release of anti-inflammatory cytokines. It reduced pathological damage to liver tissues, preserved normal liver function, decreased the release of reactive oxygen species (ROS) and LDH, and enhanced the GSH/GSSG ratio. Moreover, IRG1 and itaconic acid activated the Nrf2 signaling pathway, regulating the expression of its downstream antioxidative stress-related proteins. Additionally, they inhibited the activity of NLRP3 inflammatory vesicles to suppress the expression of macrophage-associated pyroptosis signaling molecules. Our findings demonstrate that IRG1/OI inhibits NLRP3 inflammatory vesicle activation and macrophage pyroptosis by modulating the Nrf2 signaling pathway, thereby attenuating acute liver injury in mice with sepsis. These findings could facilitate the clinical application of IRG1/Itaconate to prevent sepsis-induced acute liver injury.
Collapse
Affiliation(s)
- Pei Zhou
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022,China
| | - Lei Yang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022,China
| | - Ruidong Li
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022,China
| | - Yuping Yin
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022,China
| | - Gengchen Xie
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022,China
| | - Xinghua Liu
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022,China
| | - Liang Shi
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022,China
| | - Kaixiong Tao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022,China.
| | - Peng Zhang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022,China.
| |
Collapse
|
33
|
Chen Y, Chen S, Zhang J, Hu X, Li N, Liu Z, Huang L, Yu J, Zhang Y, Lin X, Xu Z, Fang Y, Chen Z, Guo Y, Chen B. Electroacupuncture pre-treatment exerts a protective effect on LPS-induced cardiomyopathy in mice through the delivery of miR-381 via exosomes. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167208. [PMID: 38701956 DOI: 10.1016/j.bbadis.2024.167208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 04/25/2024] [Accepted: 04/26/2024] [Indexed: 05/06/2024]
Abstract
OBJECTIVE This study aims to investigate the cardiac protective effects and molecular mechanisms of electroacupuncture (EA) pre-treatment in lipopolysaccharide (LPS)-Induced Cardiomyopathy. METHODS AND RESULTS Pre-treatment with EA was performed 30 min before intraperitoneal injection of LPS. Cardiac function changes in mice of the EA + LPS group were observed using electrocardiography, echocardiography, and enzyme linked immunosorbent assay (ELISA) and compared with the LPS group. The results demonstrated that EA pre-treatment significantly improved the survival rate of septic mice, alleviated the severity of endotoxemia, and exhibited notable cardiac protective effects. These effects were characterized by a reduction in ST-segment elevation on electrocardiography, an increase in ejection fraction (EF) and fraction shortening (FS) on echocardiography and a decrease in the expression of serum cardiac troponin I (cTn-I) levels. Serum exosomes obtained after EA pre-treatment were extracted and administered to septic mice, revealing significant cardiac protective effects of EA-derived exosomes. Furthermore, the antagonism of circulating exosomes in mice markedly suppressed the cardiac protective effects conferred by EA pre-treatment. Analysis of serum exosomes using quantitative reverse transcription-polymerase chain reaction (qRT-PCR) revealed a significant upregulation of miR-381 expression after EA pre-treatment. Inhibition or overexpression of miR-381 through serotype 9 adeno-associated virus (AAV9)-mediated gene delivery demonstrated that overexpression of miR-381 exerted a cardiac protective effect, while inhibition of miR-381 significantly attenuated the cardiac protective effects conferred by EA pre-treatment. CONCLUSIONS Our research findings have revealed a novel endogenous cardiac protection mechanism, wherein circulating exosomes derived from EA pre-treatment mitigate LPS-induced cardiac dysfunction via miR-381.
Collapse
Affiliation(s)
- Yong Chen
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin 300102, China
| | - Shuangli Chen
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jingyu Zhang
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xiyou Hu
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Ningcen Li
- Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 500515, China
| | - Zhen Liu
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Lihong Huang
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jianbo Yu
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin 300102, China
| | - Yuan Zhang
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin 300102, China
| | - Xiaowei Lin
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zhifang Xu
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yuxin Fang
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zelin Chen
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| | - Yi Guo
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China; School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Bo Chen
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| |
Collapse
|
34
|
Meng S, Liu J, Wang Z, Fan Y, Pei S, Wang E, Song Y, Cui Y, Xie K. Inhibition of Golgi stress alleviates sepsis-induced cardiomyopathy by reducing inflammation and apoptosis. Int Immunopharmacol 2024; 133:112103. [PMID: 38648713 DOI: 10.1016/j.intimp.2024.112103] [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: 03/31/2024] [Revised: 04/13/2024] [Accepted: 04/15/2024] [Indexed: 04/25/2024]
Abstract
BACKGROUND Sepsis is often accompanied by multiple organ dysfunction, in which the incidence of cardiac injury is about 60%, and is closely related to high mortality. Recent studies have shown that Golgi stress is involved in liver injury, kidney injury, and lung injury in sepsis. However, whether it is one of the key mechanisms of sepsis-induced cardiomyopathy (SIC) is still unclear. The aim of this study is to investigate whether Golgi stress mediates SIC and the specific mechanism. METHODS Sepsis model of male C57BL/6J mice was established by cecal ligation and puncture. To observe the effect of Golgi stress on SIC, mice were injected with Golgi stimulant (Brefeldin A) or Golgi inhibitor (Glutathione), respectively. The 7-day survival rate of mice were recorded, and myocardial injury indicators including cardiac function, myocardial enzymes, myocardial pathological tissue score, myocardial inflammatory factors, and apoptosis were detected. The morphology of Golgi was observed by immunofluorescence, and the Golgi stress indices including GM-130, GOLPH3 and Goligin97 were detected by WB and qPCR. RESULTS After CLP, the cardiac function of mice was impaired and the levels of myocardial enzymes were significantly increased. Golgi stress was accompanied by increased myocardial inflammation and apoptosis. Moreover, the expressions of morphological proteins GM-130 and Golgin97 were decreased, and the expression of stress protein GOLPH3 was increased. In addition, Brefeldin A increased 7-day mortality and the above indicators in mice. The use of glutathione improves all of the above indicators. CONCLUSION Golgi stress mediates SIC, and the inhibition of Golgi stress can improve SIC by inhibiting apoptosis and inflammation.
Collapse
Affiliation(s)
- Shuqi Meng
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China; Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Jianfeng Liu
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Zhiwei Wang
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yan Fan
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Shuaijie Pei
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China; Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Enquan Wang
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yu Song
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yan Cui
- Department of Pathogen Biology, School of Basic Medical Science, Tianjin Medical University, Tianjin 300070, China.
| | - Keliang Xie
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China; Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China.
| |
Collapse
|
35
|
Liu C, Chang Z, Chen K, Xue Q, Shu B, Wei Z, Zhou X, Guo L, Zhang Y, Pan Y, Cao Q, Liang H, Sun Q, Zhang X. A mitochondrion-targeted cyanine agent for NIR-II fluorescence-guided surgery combined with intraoperative photothermal therapy to reduce prostate cancer recurrence. J Nanobiotechnology 2024; 22:224. [PMID: 38702709 PMCID: PMC11069140 DOI: 10.1186/s12951-024-02477-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 04/14/2024] [Indexed: 05/06/2024] Open
Abstract
Poorly identified tumor boundaries and nontargeted therapies lead to the high recurrence rates and poor quality of life of prostate cancer patients. Near-infrared-II (NIR-II) fluorescence imaging provides certain advantages, including high resolution and the sensitive detection of tumor boundaries. Herein, a cyanine agent (CY7-4) with significantly greater tumor affinity and blood circulation time than indocyanine green was screened. By binding albumin, the absorbance of CY7-4 in an aqueous solution showed no effects from aggregation, with a peak absorbance at 830 nm and a strong fluorescence emission tail beyond 1000 nm. Due to its extended circulation time (half-life of 2.5 h) and high affinity for tumor cells, this fluorophore was used for primary and metastatic tumor diagnosis and continuous monitoring. Moreover, a high tumor signal-to-noise ratio (up to ~ 10) and excellent preferential mitochondrial accumulation ensured the efficacy of this molecule for photothermal therapy. Therefore, we integrated NIR-II fluorescence-guided surgery and intraoperative photothermal therapy to overcome the shortcomings of a single treatment modality. A significant reduction in recurrence and an improved survival rate were observed, indicating that the concept of intraoperative combination therapy has potential for the precise clinical treatment of prostate cancer.
Collapse
Affiliation(s)
- Chenchen Liu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology & Center for Biomedical Optics and Molecular Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Science, Shenzhen, 518055, China
| | - Zong Chang
- Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology & Center for Biomedical Optics and Molecular Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Science, Shenzhen, 518055, China
| | - Kailei Chen
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qiang Xue
- Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology & Center for Biomedical Optics and Molecular Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Science, Shenzhen, 518055, China
| | - Bingxin Shu
- Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology & Center for Biomedical Optics and Molecular Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Science, Shenzhen, 518055, China
| | - Zhihao Wei
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xuan Zhou
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Like Guo
- Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology & Center for Biomedical Optics and Molecular Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Science, Shenzhen, 518055, China
| | - Yuling Zhang
- Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology & Center for Biomedical Optics and Molecular Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Science, Shenzhen, 518055, China
| | - Yingying Pan
- Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology & Center for Biomedical Optics and Molecular Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Science, Shenzhen, 518055, China
| | - Qi Cao
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Huageng Liang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qinchao Sun
- Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology & Center for Biomedical Optics and Molecular Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Science, Shenzhen, 518055, China.
| | - Xiaoping Zhang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Institute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
36
|
Gao M, Cai Q, Bian Y, Wang Z, Xu L, Peng J. Protective effect of esculentoside A against myocardial infarction via targeting C-X-C motif chemokine receptor 2. Biomed Pharmacother 2024; 174:116529. [PMID: 38569275 DOI: 10.1016/j.biopha.2024.116529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 03/18/2024] [Accepted: 03/29/2024] [Indexed: 04/05/2024] Open
Abstract
Myocardial infarction (MI) is the primary cause of cardiac mortality. Esculentoside A (EsA), a triterpenoid saponin, has anti-inflammatory and antioxidant activities. However, its effect on MI remains unknown. In this study, the protective effect and mechanisms of EsA against MI were investigated. EsA significantly alleviated hypoxia-induced HL-1 cell injury, including increasing cell viability, inhibiting reactive oxygen species (ROS) production, mitochondrial membrane potential (MMP) and lactate dehydrogenase (LDH) leakage. In mouse MI model by left coronary artery (LAD) ligating, EsA obviously restored serum levels of creatine kinase isoenzymes (CK-MB), cardiac troponin I (cTnI), superoxide dismutase (SOD) and malondialdehyde (MDA). In addition, the cardioprotective effect of EsA was further confirmed by infarct size, electrocardiogram and echocardiography. Mechanistically, the targeted binding relationship between EsA and C-X-C motif chemokine receptor 2 (CXCR2) was predicted by molecular docking and dynamics, and validated by small molecule pull-down and surface plasmon resonance tests. EsA inhibited CXCR2 level both in vitro and in vivo, correspondingly alleviated oxidative stress by suppressing NOX1 and NOX2 and relieved inflammation through inhibiting p65 and p-p65. It demonstrated that EsA could play a cardioprotective role by targeting CXCR2. However, the effect of EsA against MI was abolished in combination with CXCR2 overexpression both in vitro and in vivo. This study revealed that EsA showed excellent cardioprotective activities by targeting CXCR2 to alleviate oxidative stress and inflammation in MI. EsA may function as a novel CXCR2 inhibitor and a potent candidate for the prevention and intervention of MI in the future.
Collapse
Affiliation(s)
- Meng Gao
- Institute of Intergrative Medicine, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China; College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Qing Cai
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Yehua Bian
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Zhuoya Wang
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Lina Xu
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China.
| | - Jinyong Peng
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China; College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China; Department of Traditional Chinese Medicine Pharmacology, School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; Hubei Shizhen Laboratory, Wuhan 430065, China.
| |
Collapse
|
37
|
Han T, Tang H, Lin C, Yan D, Zhou Z, Yang Y, Cai L, Zhu J, Gao B, Si Y, Fu W, Tai Z, Tang X, Guo D. Costunolide mitigates inflammation and promotes extracellualr matrix integrity of thoracic aortic dissection by inhibiting NF-κB signaling. Int Immunopharmacol 2024; 131:111784. [PMID: 38493694 DOI: 10.1016/j.intimp.2024.111784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 03/19/2024]
Abstract
BACKGROUND Thoracic aortic dissection (TAD) is one of the most fatal cardiovascular diseases. One of its important pathological characteristics is the local inflammatory response. Many studies have found that Macrophage polarization plays an extremely critical role in the inflammatory progression and tissue remodeling of TAD. Costunolide (CTD) has an improving effect on oxidative stress and inflammation in the body. However, whether it can promote the integrity of extracellular matrix in Aortic dissection and its mechanism are still unclear. METHODS The male C57BL/6J mice were used to construct an animal model of TAD with β-aminopropionitrile (BAPN) (100 mg/kg/day, lasting for 28 days), and then CTD (10 mg/kg or 100 mg/kg) was injected intraperitoneally for 28 days to check the survival rate, TAD incidence, aortic morphology and other indicators of the mice. Using hematoxylin-eosin (HE), Masson, Elastin van Gieson (EVG) staining, immunofluorescence (IF), and immunohistochemical staining, the study aimed to determine the therapeutic effects of CTD on an animal model with BAPN-induced TAD. To enhance the examination of the regulatory mechanism of CTD, we conducted transcriptome sequencing on arterial tissues of mice in both the BAPN group and the BAPN + CTD100 group. Next, ANG II were used to construct TAD model in vascular smooth muscle cells (VMSCs). The effects of CTD on the proliferation, migration, invasion, and apoptosis of ANG II-induced cells are to be detected. The expression of MMP2, MMP9, P65, and p-P65 in each group will be examined using Western blot. Finally, the overexpression of IκB kinaseβ (IKKβ) will be established in VMSCs cells to further explore the protective function of CTD. RESULTS The result showed that CTD significantly inhibited BAPN induced mortality and TAD incidence in the animal model, improved aortic vascular morphology, promoted the integrity of extracellular matrix in TAD, reduced tissue inflammation, reduced the accumulation of M1 macrophage, promoted M2 macrophage polarization, and reduced the expression of NF-κB pathway related proteins. Mechanistically, CTD significantly weakened the proliferation, migration, invasion, and apoptosis. p-P65 protein expression of TAD cells were induced by ANG II and IKK-β. CONCLUSION CTD has the potential to alleviate inflammation, VSMC apoptosis, MMP2/9 levels, and enhance extracellular matrix integrity in TAD by inhibiting the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Tonglei Han
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200030, China
| | - Hanfei Tang
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200030, China
| | - Changpo Lin
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200030, China
| | - Dong Yan
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200030, China
| | - Zhenyu Zhou
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200030, China
| | - Yimin Yang
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200030, China
| | - Liang Cai
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200030, China
| | - Jiaqi Zhu
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200030, China
| | - Bin Gao
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200030, China
| | - Yi Si
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200030, China
| | - Weiguo Fu
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200030, China
| | - Zongguang Tai
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, 1278 Baode Road, Jing'an District, Shanghai 200040, China.
| | - Xiao Tang
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200030, China.
| | - Daqiao Guo
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200030, China.
| |
Collapse
|
38
|
Lu H. Inflammatory liver diseases and susceptibility to sepsis. Clin Sci (Lond) 2024; 138:435-487. [PMID: 38571396 DOI: 10.1042/cs20230522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 01/09/2024] [Accepted: 03/12/2024] [Indexed: 04/05/2024]
Abstract
Patients with inflammatory liver diseases, particularly alcohol-associated liver disease and metabolic dysfunction-associated fatty liver disease (MAFLD), have higher incidence of infections and mortality rate due to sepsis. The current focus in the development of drugs for MAFLD is the resolution of non-alcoholic steatohepatitis and prevention of progression to cirrhosis. In patients with cirrhosis or alcoholic hepatitis, sepsis is a major cause of death. As the metabolic center and a key immune tissue, liver is the guardian, modifier, and target of sepsis. Septic patients with liver dysfunction have the highest mortality rate compared with other organ dysfunctions. In addition to maintaining metabolic homeostasis, the liver produces and secretes hepatokines and acute phase proteins (APPs) essential in tissue protection, immunomodulation, and coagulation. Inflammatory liver diseases cause profound metabolic disorder and impairment of energy metabolism, liver regeneration, and production/secretion of APPs and hepatokines. Herein, the author reviews the roles of (1) disorders in the metabolism of glucose, fatty acids, ketone bodies, and amino acids as well as the clearance of ammonia and lactate in the pathogenesis of inflammatory liver diseases and sepsis; (2) cytokines/chemokines in inflammatory liver diseases and sepsis; (3) APPs and hepatokines in the protection against tissue injury and infections; and (4) major nuclear receptors/signaling pathways underlying the metabolic disorders and tissue injuries as well as the major drug targets for inflammatory liver diseases and sepsis. Approaches that focus on the liver dysfunction and regeneration will not only treat inflammatory liver diseases but also prevent the development of severe infections and sepsis.
Collapse
Affiliation(s)
- Hong Lu
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY 13210, U.S.A
| |
Collapse
|
39
|
Cai F, Li D, Zhou K, Zhang W, Yang Y. Tiliroside attenuates acute kidney injury by inhibiting ferroptosis through the disruption of NRF2-KEAP1 interaction. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 126:155407. [PMID: 38340577 DOI: 10.1016/j.phymed.2024.155407] [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: 09/21/2023] [Revised: 11/24/2023] [Accepted: 02/01/2024] [Indexed: 02/12/2024]
Abstract
BACKGROUND Ferroptosis, an iron-dependent process that regulates cell death. Emerging evidences suggest that ferroptosis induces acute kidney injury (AKI) progression, and inhibiting ferroptosis provides an effect strategy for AKI treatment. The disruption of the NRF2-KEAP1 protein to protein interaction (PPI) induces NRF2 activation, which provides a promising strategy that can identify new ferroptosis inhibitors. A previous study revealed that tiliroside, a glycosidic flavonoid extracted from Edgeworthia chrysantha Lindl (buds), has anti-neuroinflammatory and neuroprotective effects via NRF2 activation. However, the mechanism through which tiliroside activates NRF2 is unknown, and it remains unclear whether it has protective effects against AKI. PURPOSE To investigate whether tiliroside has protective effects against AKI in mice and the associated mechanisms. METHODS Possible tiliroside substrates were analyzed using molecular docking. Cisplatin- and ischemia-reperfusion injury (IRI)-induced AKI mouse models and HK2 cells model were constructed to evaluate the protective effects of tiliroside. CRISPR/Cas9 mediated NRF2 knockout HK2 cells were used to verify whether NRF2 mediates tiliroside protective effects. RESULTS In vivo, our results showed that tiliroside treatment preserved kidney functions in AKI mice models, as showed by lower levels of serum creatinine (SCr), blood urea nitrogen (BUN), and renal injury markers, including neutrophil gelatinase-associated lipocalin (NGAL) and kidney injury molecule 1 (KIM1), compared with the mice in control groups. In vitro, tiliroside treatment greatly ameliorated cisplatin-induced ferroptosis through NRF2 activation in cultured HK2 cells, as evidenced by the protective effects of tiliroside being greatly blunted after the knockout of NRF2 in HK2 cells. Mechanistic studies indicated that tiliroside promoted NRF2/GPX4 pathway activation and ferroptosis inhibition, perhaps via the disruption of the NRF2-KEAP1 PPI. CONCLUSION Together, our results demonstrate that tiliroside may serve as a NRF2-KEAP1 PPI inhibitor and prevents ferroptosis-induced AKI, indicating its potential for clinical AKI treatment.
Collapse
Affiliation(s)
- Fangfang Cai
- School of Biopharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Dangran Li
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, 210023, China
| | - Kaiqian Zhou
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing 210008, China
| | - Wen Zhang
- Department of Nephrology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine
| | - Yunwen Yang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing 210008, China.
| |
Collapse
|
40
|
He J, Zhou S, Wang J, Sun B, Ni D, Wu J, Peng X. Anti-inflammatory and anti-oxidative electrospun nanofiber membrane promotes diabetic wound healing via macrophage modulation. J Nanobiotechnology 2024; 22:116. [PMID: 38493156 PMCID: PMC10943854 DOI: 10.1186/s12951-024-02385-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 03/07/2024] [Indexed: 03/18/2024] Open
Abstract
BACKGROUND In the inflammatory milieu of diabetic chronic wounds, macrophages undergo substantial metabolic reprogramming and play a pivotal role in orchestrating immune responses. Itaconic acid, primarily synthesized by inflammatory macrophages as a byproduct in the tricarboxylic acid cycle, has recently gained increasing attention as an immunomodulator. This study aims to assess the immunomodulatory capacity of an itaconic acid derivative, 4-Octyl itaconate (OI), which was covalently conjugated to electrospun nanofibers and investigated through in vitro studies and a full-thickness wound model of diabetic mice. RESULTS OI was feasibly conjugated onto chitosan (CS), which was then grafted to electrospun polycaprolactone/gelatin (PG) nanofibers to obtain P/G-CS-OI membranes. The P/G-CS-OI membrane exhibited good mechanical strength, compliance, and biocompatibility. In addition, the sustained OI release endowed the nanofiber membrane with great antioxidative and anti-inflammatory activities as revealed in in vitro and in vivo studies. Specifically, the P/G-CS-OI membrane activated nuclear factor-erythroid-2-related factor 2 (NRF2) by alkylating Kelch-like ECH-associated protein 1 (KEAP1). This antioxidative response modulates macrophage polarization, leading to mitigated inflammatory responses, enhanced angiogenesis, and recovered re-epithelization, finally contributing to improved healing of mouse diabetic wounds. CONCLUSIONS The P/G-CS-OI nanofiber membrane shows good capacity in macrophage modulation and might be promising for diabetic chronic wound treatment.
Collapse
Affiliation(s)
- Jibing He
- Department of Orthopedics, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, P. R. China
| | - Shasha Zhou
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, P. R. China
| | - Jiaxing Wang
- Department of Orthopedics, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, P. R. China
| | - Binbin Sun
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, P. R. China
| | - Dalong Ni
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P. R. China.
| | - Jinglei Wu
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, P. R. China.
| | - Xiaochun Peng
- Department of Orthopedics, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, P. R. China.
| |
Collapse
|
41
|
Su W, Yin Y, Cheng Y, Yu S, Hu R, Zhang H, Hu J, Ren R, Zhang Y, Zhao J, Wang A, Lyu Z, Mu Y, Gao J. The phenotype and related gene expressions of macrophages in adipose tissue of T2D mice following MSCs infusion. Immunobiology 2024; 229:152788. [PMID: 38309141 DOI: 10.1016/j.imbio.2024.152788] [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: 10/25/2023] [Revised: 12/28/2023] [Accepted: 01/24/2024] [Indexed: 02/05/2024]
Abstract
BACKGROUND Infusion of mesenchymal stem cells (MSCs) induces polarization of M2 macrophages in adipose tissue of type 2 diabetes (T2D) mice. Studies have shown that M2 macrophages were divided into four sub-phenotypes (M2a, M2b, M2c and M2d) with different functions, and manuscripts have also confirmed that macrophages co-cultured with MSCs were not matched with known four phenotype macrophages. Therefore, our study explored the phenotype and related gene expressions of macrophages in the adipose tissue of T2D mice with/without MSCs infusion. METHODS We induced a T2D mouse model by using high-fat diets and streptozotocin (STZ) injection. The mice were divided into three groups: the control group, the T2D group, and the MSCs group. MSCs were systemically injected once a week for 6 weeks. The phenotype of macrophages in adipose tissue was detected via flow cytometric analysis. We also investigated the gene expression of macrophages in different groups via SMART-RNA-sequencing and quantitative real-time reverse transcriptase polymerase chain reaction (qRT-PCR). RESULTS The present study found that the macrophages of adipose tissue in the MSCs group were polarized to the M2 phenotype mixed with four sub-phenotypes. Besides, M2a and M2c held a dominant position, while M2b and M2d (tumor-associated macrophages, TAMs) exhibited a decreasing trend after infusion of MSCs. Moreover, the MSCs group did not appear to express higher levels of tumor-associated, inflammation-associated, or fibrosis-associated genes in comparison to the T2D group. CONCLUSION The present results unveiled that the macrophage phenotype was inclined to be present in a hybridity state of four M2 sub-phenotypes and the genes related to tumor-promoting, pro-inflammation and pro-fibrosis were not increased after MSCs injection.
Collapse
Affiliation(s)
- Wanlu Su
- School of Medicine, Nankai University, No. 94 Weijin Road, Tianjin 300071, China; Department of Endocrinology, Chinese People's Liberation Army General Hospital, No. 28 Fuxing Road, Beijing 100853, China
| | - Yaqi Yin
- Department of Endocrinology, Chinese People's Liberation Army General Hospital, No. 28 Fuxing Road, Beijing 100853, China
| | - Yu Cheng
- Department of Endocrinology, Chinese People's Liberation Army General Hospital, No. 28 Fuxing Road, Beijing 100853, China
| | - Songyan Yu
- Department of Endocrinology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Ruofan Hu
- Department of Endocrinology, Chinese People's Liberation Army General Hospital, No. 28 Fuxing Road, Beijing 100853, China
| | - Haixia Zhang
- Department of Endocrinology, Chinese People's Liberation Army General Hospital, No. 28 Fuxing Road, Beijing 100853, China
| | - Jia Hu
- Department of Endocrinology, Chinese People's Liberation Army General Hospital, No. 28 Fuxing Road, Beijing 100853, China
| | - Rui Ren
- Department of Endocrinology, Chinese People's Liberation Army General Hospital, No. 28 Fuxing Road, Beijing 100853, China
| | - Yue Zhang
- Department of Endocrinology, Chinese People's Liberation Army General Hospital, No. 28 Fuxing Road, Beijing 100853, China
| | - Jian Zhao
- Department of Endocrinology, Chinese People's Liberation Army General Hospital, No. 28 Fuxing Road, Beijing 100853, China
| | - Anning Wang
- Department of Endocrinology, Chinese People's Liberation Army General Hospital, No. 28 Fuxing Road, Beijing 100853, China
| | - Zhaohui Lyu
- Department of Endocrinology, Chinese People's Liberation Army General Hospital, No. 28 Fuxing Road, Beijing 100853, China.
| | - Yiming Mu
- School of Medicine, Nankai University, No. 94 Weijin Road, Tianjin 300071, China; Department of Endocrinology, Chinese People's Liberation Army General Hospital, No. 28 Fuxing Road, Beijing 100853, China.
| | - Jieqing Gao
- Department of Endocrinology, Beijing Rehabilitation Hospital of Capital Medical University, Beijing, China.
| |
Collapse
|
42
|
Li X, Wu Y, Yang Y, Wu Y, Yu X, Hu W. Omaveloxolone ameliorates isoproterenol-induced pathological cardiac hypertrophy in mice. Free Radic Res 2024; 58:57-68. [PMID: 38145457 DOI: 10.1080/10715762.2023.2299359] [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: 05/30/2023] [Accepted: 12/19/2023] [Indexed: 12/26/2023]
Abstract
Nuclear factor erythroid 2-related factor 2 (Nrf2) is an important transcriptional regulator that plays a protective role against various cardiovascular diseases. Omaveloxolone is a newly discovered potent activator of Nrf2 that has a variety of cytoprotective functions. However, the potential role of omaveloxolone in the process of pathological cardiac hypertrophy and heart failure are still unknown. In this study, an isoproterenol (ISO)-induced pathological cardiac hypertrophy model was established to investigate the protective effect of omaveloxolone in vivo and in vitro. Our study first confirmed that omaveloxolone administration improved ISO-induced pathological cardiac hypertrophy in mice and neonatal cardiomyocytes. Omaveloxolone administration also diminished ISO-induced cardiac oxidative stress, inflammation and cardiomyocyte apoptosis. In addition, omaveloxolone administration activated the Nrf2 signaling pathway, and Nrf2 knockdown almost completely abolished the cardioprotective effect of omaveloxolone, indicated that the cardioprotective effect of omaveloxolone was directly related to the activation of the Nrf2 signaling. In summary, our study identified that omaveloxolone may be a promising therapeutic agent to mitigate pathological cardiac hypertrophy.
Collapse
Affiliation(s)
- Xianchao Li
- Health Science Center, Yangtze University, Huanggang Central Hospital of Yangtze University, Huanggang, China
| | - Yang Wu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yunzhao Yang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yaohua Wu
- Health Science Center, Yangtze University, Huanggang Central Hospital of Yangtze University, Huanggang, China
| | - Xi Yu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wenjuan Hu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
43
|
Pfefferlé M, Vallelian F. Transcription Factor NRF2 in Shaping Myeloid Cell Differentiation and Function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1459:159-195. [PMID: 39017844 DOI: 10.1007/978-3-031-62731-6_8] [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: 07/18/2024]
Abstract
NFE2-related factor 2 (NRF2) is a master transcription factor (TF) that coordinates key cellular homeostatic processes including antioxidative responses, autophagy, proteostasis, and metabolism. The emerging evidence underscores its significant role in modulating inflammatory and immune processes. This chapter delves into the role of NRF2 in myeloid cell differentiation and function and its implication in myeloid cell-driven diseases. In macrophages, NRF2 modulates cytokine production, phagocytosis, pathogen clearance, and metabolic adaptations. In dendritic cells (DCs), it affects maturation, cytokine production, and antigen presentation capabilities, while in neutrophils, NRF2 is involved in activation, migration, cytokine production, and NETosis. The discussion extends to how NRF2's regulatory actions pertain to a wide array of diseases, such as sepsis, various infectious diseases, cancer, wound healing, atherosclerosis, hemolytic conditions, pulmonary disorders, hemorrhagic events, and autoimmune diseases. The activation of NRF2 typically reduces inflammation, thereby modifying disease outcomes. This highlights the therapeutic potential of NRF2 modulation in treating myeloid cell-driven pathologies.
Collapse
Affiliation(s)
- Marc Pfefferlé
- Department of Internal Medicine, Spital Limmattal, Schlieren, Switzerland
| | - Florence Vallelian
- Department of Internal Medicine, University of Zurich and University Hospital of Zurich, Zurich, Switzerland.
| |
Collapse
|
44
|
Qiao Y, Wotring JW, Zheng Y, Zhang CJ, Zhang Y, Jiang X, Pretto CD, Eyunni S, Parolia A, He T, Cheng C, Cao X, Wang R, Su F, Ellison SJ, Wang Y, Qin J, Yan H, Zhou Q, Ma L, Sexton JZ, Chinnaiyan AM. Proxalutamide reduces SARS-CoV-2 infection and associated inflammatory response. Proc Natl Acad Sci U S A 2023; 120:e2221809120. [PMID: 37459541 PMCID: PMC10372636 DOI: 10.1073/pnas.2221809120] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 06/12/2023] [Indexed: 07/20/2023] Open
Abstract
Early in the COVID-19 pandemic, data suggested that males had a higher risk of developing severe disease and that androgen deprivation therapy might be associated with protection. Combined with the fact that TMPRSS2 (transmembrane serine protease 2), a host entry factor for the SARS-CoV-2 virus, was a well-known androgen-regulated gene, this led to an upsurge of research investigating androgen receptor (AR)-targeting drugs. Proxalutamide, an AR antagonist, was shown in initial clinical studies to benefit COVID-19 patients; however, further validation is needed as one study was retracted. Due to continued interest in proxalutamide, which is in phase 3 trials, we examined its ability to impact SARS-CoV-2 infection and downstream inflammatory responses. Proxalutamide exerted similar effects as enzalutamide, an AR antagonist prescribed for advanced prostate cancer, in decreasing AR signaling and expression of TMPRSS2 and angiotensin-converting enzyme 2 (ACE2), the SARS-CoV-2 receptor. However, proxalutamide led to degradation of AR protein, which was not observed with enzalutamide. Proxalutamide inhibited SARS-CoV-2 infection with an IC50 value of 97 nM, compared to 281 nM for enzalutamide. Importantly, proxalutamide inhibited infection by multiple SARS-CoV-2 variants and synergized with remdesivir. Proxalutamide protected against cell death in response to tumor necrosis factor alpha and interferon gamma, and overall survival of mice was increased with proxalutamide treatment prior to cytokine exposure. Mechanistically, we found that proxalutamide increased levels of NRF2, an essential transcription factor that mediates antioxidant responses, and decreased lung inflammation. These data provide compelling evidence that proxalutamide can prevent SARS-CoV-2 infection and cytokine-induced lung damage, suggesting that promising clinical data may emerge from ongoing phase 3 trials.
Collapse
Affiliation(s)
- Yuanyuan Qiao
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
- Department of Pathology, University of Michigan, Ann Arbor, MI48109
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI48109
| | - Jesse W. Wotring
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI48109
| | - Yang Zheng
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
| | - Charles J. Zhang
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI48109
| | - Yuping Zhang
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
- Department of Pathology, University of Michigan, Ann Arbor, MI48109
| | - Xia Jiang
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
| | - Carla D. Pretto
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI48109
| | - Sanjana Eyunni
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
| | - Abhijit Parolia
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
| | - Tongchen He
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
| | - Caleb Cheng
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
| | - Xuhong Cao
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
| | - Rui Wang
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
| | - Fengyun Su
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
| | - Stephanie J. Ellison
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
| | - Yini Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing102206, China
| | - Jun Qin
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing102206, China
| | - Honghua Yan
- Kintor Pharmaceutical Limited, Suzhou Industrial Park, Suzhuo215123, China
| | - Qianxiang Zhou
- Kintor Pharmaceutical Limited, Suzhou Industrial Park, Suzhuo215123, China
| | - Liandong Ma
- Kintor Pharmaceutical Limited, Suzhou Industrial Park, Suzhuo215123, China
| | - Jonathan Z. Sexton
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI48109
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI48109
- Center for Drug Repurposing, University of Michigan, Ann Arbor, MI48109
- Michigan Institute for Clinical and Health Research, University of Michigan, Ann Arbor, MI48109
- Department of Pharmacology, University of Michigan, Ann Arbor, MI48109
| | - Arul M. Chinnaiyan
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
- Department of Pathology, University of Michigan, Ann Arbor, MI48109
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI48109
- HHMI, University of Michigan, Ann Arbor, MI48109
- Department of Urology, University of Michigan, Ann Arbor, MI48109
| |
Collapse
|