1
|
Guo Y, Yang L, Yao L, Zhou C, Zhu Y, Xu C, Wang W, Song J, Zhang M, Deng Z. Carbon dots derived from Ligusticum Chuanxiong mitigate cardiac injury by disrupting the harmful oxidative stress-apoptosis cycle. J Nanobiotechnology 2025; 23:391. [PMID: 40442709 PMCID: PMC12121267 DOI: 10.1186/s12951-025-03477-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2025] [Accepted: 05/19/2025] [Indexed: 06/02/2025] Open
Abstract
BACKGROUND Myocardial ischemia-reperfusion injury (MIRI) represents a significant complication following myocardial infarction surgery, for which preventive strategies remain limited. The primary pathological characteristics of MIRI include oxidative stress and apoptosis. RESULTS This study presents the synthesis of carbon dots derived from Ligusticum Chuanxiong (LC-CDs) through the application of the hydrothermal method. The LC-CDs show strong scavenging abilities for free radicals, effectively reducing oxidative stress and preventing apoptosis, which helps combat MIRI. The findings demonstrate that LC-CDs can effectively neutralize excessive ROS within cells, thereby alleviating oxidative stress, restoring mitochondrial function, and preventing DNA damage. Concurrently, LC-CDs suppress the polarization of M1-type macrophages and reduce the secretion of pro-inflammatory cytokines. Following the in situ administration of LC-CDs into the hearts of MIRI-model rats, a significant reduction in the necrotic area of the myocardium was observed, alongside the restoration of cardiac function, with no adverse reactions reported. Moreover, similar to the pharmacological effects of Ligusticum chuanxiong, LC-CDs can also inhibit apoptosis by protecting mitochondria and suppressing the expression of apoptotic proteins (Caspase3, Caspase9, and Bax). CONCLUSIONS The intervention strategy employing LC-CDs, which targets oxidative stress and apoptosis in MIRI, holds promise as a potential model for the clinical treatment of MIRI.
Collapse
Affiliation(s)
- Yapeng Guo
- School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Lei Yang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Li Yao
- Department of Neurology, XD Group Hospital, Xi'an, Shaanxi, 710077, China
| | - Chengdong Zhou
- School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Yuanyuan Zhu
- School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Chenxi Xu
- School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Wenlong Wang
- School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Jian Song
- Institute of Cardiovascular Sciences, Guangxi Academy of Medical Sciences, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, 530021, China
| | - Mingzhen Zhang
- School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
| | - Zhichao Deng
- School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
| |
Collapse
|
2
|
Liu JY, Luo JF, Wu XY, Liu T, Wang R, Zhang Q, Liu YM, Wu H. SLP65/SLP76 Csk-interacting membrane protein promotes hepatic ischemia-reperfusion injury by activating TLR4/Erk1/2-mediated macrophages M1 polarization. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167916. [PMID: 40403937 DOI: 10.1016/j.bbadis.2025.167916] [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: 11/12/2024] [Revised: 04/03/2025] [Accepted: 05/19/2025] [Indexed: 05/24/2025]
Abstract
BACKGROUND SLP65/SLP76, Csk-interacting membrane protein (SCIMP), is a membrane protein plays a crucial role in the regulation of macrophage polarization. This study aimed to investigate the mechanism of SCIMP-mediated M1 polarization in liver macrophages and ischemia-reperfusion injury (IRI) in liver transplantation. METHODS Mice underwent orthotopic liver transplantation. In in vivo experiments, mice were divided into the Sham group, LT group, LT+Scramble group, LT+SCIMP (-) group, and LT+ERK1/2 (-) group, and SCIMP or ERK1/2 knockdown was performed using AAV-Erk1/2-RNAi-F4/80-EGFP and AAV-SCIMP-RNAi-F4/80-EGFP. In the subsequent in vitro experiments with primary cells, macrophages were divided into the Ctrl group, H/R group, H/R+Scramble group, H/R+SCIMP (-) group, and H/R+ERK1/2 (-) group, with SCIMP knockdown achieved using siRNA. Immunoprecipitation (IP) was used to detect the interaction between TLR4 and Erk1/2. Liver damage was detected by Hematoxylin and eosin (HE) staining. Polarization was detected by western blot (WB), RT-PCR, immunohistochemistry (IHC), immunofluorescence technique (IF), enzyme-linked immunosorbent assay (ELISA) and flow cytometry (FC). RESULTS Knockdown of SCIMP ameliorated hepatic IRI and liver macrophages M1 polarization. Mechanically, SCIMP promoted the interaction between Erk1/2 and TLR4 in hypoxia/reoxygenation (H/R)-induced liver macrophages, while the inhibition of Erk1/2 reduced liver macrophages M1 polarization and liver IRI. CONCLUSION SCIMP promotes hepatic ischemia-reperfusion injury by activating TLR4/Erk1/2-mediated liver macrophages M1 polarization, which might become a potential therapeutic target in clinic.
Collapse
Affiliation(s)
- Jun-Yan Liu
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, China
| | - Jie-Fu Luo
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, China
| | - Xin-Yi Wu
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, China
| | - Tao Liu
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, China
| | - Rui Wang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, China
| | - Qi Zhang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, China
| | - Yi-Ming Liu
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, China.
| | - Hao Wu
- Department of Urology Surgery, The Second Affiliated Hospital of Chongqing Medical University, China.
| |
Collapse
|
3
|
Zhao QH, Zhang YT, Wen K, Ding Q, Chen ZY, Tula D, Li JH, Zhou J, Xiao YF, Guan XH, Deng KY, Wang LF, Xin HB. Myeloid but not hepatocytic CD38 is a key driver for hepatic ischemia/reperfusion injury. Signal Transduct Target Ther 2025; 10:150. [PMID: 40341132 PMCID: PMC12062225 DOI: 10.1038/s41392-025-02233-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 03/07/2025] [Accepted: 04/03/2025] [Indexed: 05/10/2025] Open
Abstract
Hepatic ischemia-reperfusion injury (HIRI) is a critical condition that often occurs during liver transplantation and surgical liver resection. However, its mechanism has not been fully elucidated. Nicotinamide adenine dinucleotide (NAD+), functioning as a coenzyme or cofactor, is crucial for both redox and non-redox processes. In mammals, CD38 serves as the primary enzyme responsible for NAD+ degradation. In this study, we reported that the absence of CD38 markedly reduces HIRI in CD38 global knockout (CD38KO) and CD38 myeloid-specific knockout (CD38MKO) mice, but not in CD38 hepatocyte-specific knockout (CD38LKO) mice compared with the control (CD38fl/fl) mice by suppressing HIRI-induced hepatic oxidative stress, inflammatory responses, and pyroptosis. The findings were corroborated by a noticeable decrease in levels of alanine aminotransferase (ALT), aspartate transaminase (AST), and lactate dehydrogenase (LDH), along with reduced necrosis. Besides, we found that the expressions of SIRT1 and its downstream targets, p53 and PPARγ, were elevated in the liver tissues of CD38KO and CD38MKO mice compared to CD38fl/fl mice, while the acetylation levels of p53 were reduced. Furthermore, we demonstrated that myeloid CD38 deficiency not only promoted M2-type polarization and inhibited M1-type polarization of macrophages but also suppressed NLRP3-mediated pyroptosis by triggering NAD+/SIRT1 signaling in macrophages, resulting in the reduction of oxidative stress, inflammation, and pyroptosis in the liver, ultimately protecting against HIRI. This study highlights myeloid CD38 as a promising target for the prevention and treatment of HIRI clinically.
Collapse
Affiliation(s)
- Qi-Hang Zhao
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
- School of Life Science, Nanchang University, Nanchang 330031, China
| | - Ya-Ting Zhang
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Ke Wen
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Qi Ding
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Zi-Ying Chen
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
- School of Life Science, Nanchang University, Nanchang 330031, China
| | - Dilinuer Tula
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Jia-Hui Li
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Juan Zhou
- Department of Gynecology and Obstetrics, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Yun-Fei Xiao
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Xiao-Hui Guan
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Ke-Yu Deng
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, China.
- School of Life Science, Nanchang University, Nanchang 330031, China.
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang 330031, China.
| | - Ling-Fang Wang
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, China.
| | - Hong-Bo Xin
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, China.
- School of Life Science, Nanchang University, Nanchang 330031, China.
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang 330031, China.
| |
Collapse
|
4
|
Kouroumalis E, Tsomidis I, Voumvouraki A. HFE-Related Hemochromatosis May Be a Primary Kupffer Cell Disease. Biomedicines 2025; 13:683. [PMID: 40149659 PMCID: PMC11940282 DOI: 10.3390/biomedicines13030683] [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: 01/14/2025] [Revised: 02/28/2025] [Accepted: 03/08/2025] [Indexed: 03/29/2025] Open
Abstract
Iron overload can lead to increased deposition of iron and cause organ damage in the liver, the pancreas, the heart and the synovium. Iron overload disorders are due to either genetic or acquired abnormalities such as excess transfusions or chronic liver diseases. The most common genetic disease of iron deposition is classic hemochromatosis (HH) type 1, which is caused by mutations of HFE. Other rare forms of HH include type 2A with mutations at the gene hemojuvelin or type 2B with mutations in HAMP that encodes hepcidin. HH type 3, is caused by mutations of the gene that encodes transferrin receptor 2. Mutations of SLC40A1 which encodes ferroportin cause either HH type 4A or HH type 4B. In the present review, an overview of iron metabolism including absorption by enterocytes and regulation of iron by macrophages, liver sinusoidal endothelial cells (LSECs) and hepatocyte production of hepcidin is presented. Hereditary Hemochromatosis and the current pathogenetic model are analyzed. Finally, a new hypothesis based on published data was suggested. The Kupffer cell is the primary defect in HFE hemochromatosis (and possibly in types 2 and 3), while the hepcidin-relative deficiency, which is the common underlying abnormality in the three types of HH, is a secondary consequence.
Collapse
Affiliation(s)
- Elias Kouroumalis
- Department of Gastroenterology, PAGNI University Hospital, University of Crete Medical School, 71500 Heraklion, Greece
- Laboratory of Gastroenterology and Hepatology, University of Crete Medical School, 71500 Heraklion, Greece;
| | - Ioannis Tsomidis
- Laboratory of Gastroenterology and Hepatology, University of Crete Medical School, 71500 Heraklion, Greece;
| | - Argyro Voumvouraki
- 1st Department of Internal Medicine, AHEPA University Hospital, 54621 Thessaloniki, Greece;
| |
Collapse
|
5
|
Wang E, Li S, Li Y, Zhou T. HMOX1 as a potential drug target for upper and lower airway diseases: insights from multi-omics analysis. Respir Res 2025; 26:41. [PMID: 39871287 PMCID: PMC11773792 DOI: 10.1186/s12931-025-03124-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 01/20/2025] [Indexed: 01/29/2025] Open
Abstract
BACKGROUND Oxidative stress is key in inflammatory airway diseases. Heme oxygenase 1 (HMOX1) regulates oxidative stress, but its role in airway diseases needs exploration. METHODS Differentially expressed genes (DEGs) between healthy nasal mucosa and chronic rhinosinusitis with nasal polyps (CRSwNP) were identified from Gene Expression Omnibus (GEO). Candidate genes were further screened using Gene Set Enrichment Analysis (GSEA) and Random Forest (RF) algorithms. Causal inference between candidate genes and upper and lower airway diseases (CRSwNP, allergic rhinitis (AR), and asthma (AS)) was conducted using bidirectional two-sample Mendelian randomization (TwoSampleMR) analysis. Single-cell RNA sequencing (scRNA-seq) data were used to determine the cellular localization and intercellular interactions of candidate genes. Molecular docking was used to identify potential therapeutic agents. RESULTS HMOX1 expression was significantly elevated in CRSwNP. TwoSampleMR analysis indicated a negative causal relationship between HMOX1 exposure and the occurrence of upper and lower airway diseases (CRSwNP [(odds ratio (OR)/95% confidence interval (CI): 0.945/(0.893-0.999), P = 0.044], AR [OR/95% CI: 0.997/(0.994-0.999), P = 0.007], and AS [OR/95% CI: 0.935/(0.895-0.977), P = 0.003]). scRNA-seq data revealed HMOX1 localization in M2 macrophages. Molecular docking identified 15 antioxidants, including Acetylcysteine and Quercetin, that can upregulate HMOX1 expression. CONCLUSION HMOX1 may have a protective role in the pathogenesis of upper and lower airway diseases (CRSwNP, AR, and AS) by modulating oxidative stress. Antioxidants that increase HMOX1 expression could offer new therapeutic avenues for these diseases. CLINICAL TRIAL Not applicable.
Collapse
Affiliation(s)
- Enhao Wang
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Shazhou Li
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Yang Li
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, China.
| | - Tao Zhou
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China.
| |
Collapse
|
6
|
Ye Z, Xie E, Guo Z, Gao Y, Han Z, Dou K, Zheng J. Association of Liver Fibrosis Markers with Mortality Outcomes in Patients with Chronic Kidney Disease and Coronary Artery Disease: Insights from the NHANES 1999-2018 Data. Cardiorenal Med 2025; 15:153-163. [PMID: 39837280 PMCID: PMC11844702 DOI: 10.1159/000543500] [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: 10/17/2024] [Accepted: 12/17/2024] [Indexed: 01/23/2025] Open
Abstract
INTRODUCTION The objective of this research was to explore the possible link between markers of liver fibrosis and survival rates in a group of adults who have been diagnosed with both chronic kidney disease (CKD) and coronary artery disease (CAD). METHODS The National Health and Nutrition Examination Survey (NHANES) data (1999-2018) for participants with both CAD and CKD were analyzed. The fibrosis-4 index (FIB-4), Nonalcoholic Fatty Liver Score (NFS), Forns index, and aspartate aminotransferase/alanine aminotransferase (AST/ALT) ratio were identified as crucial biomarkers. All-cause and cardiovascular disease (CVD) mortality were primary outcomes, assessed using Cox models, Kaplan-Meier curves, and receiver operating characteristic (ROC) analysis. RESULTS A total of 1,192 CKD and CAD patients were included. The Cox regression analysis revealed substantial correlations between elevated FIB-4, NFS, Forns index, and AST/ALT levels and a heightened risk of all-cause (hazard ratio [HR]: 1.188, 95% confidence interval [CI]: 1.108-1.274; HR: 1.145, 95% CI: 1.069-1.227; HR: 1.142, 95% CI: 1.081-1.201; HR: 1.316, 95% CI: 1.056-1.639, respectively) and CVD mortality (HR: 1.133, 95% CI: 1.007-1.275; HR: 1.155, 95% CI: 1.024-1.303; HR: 1.208, 95% CI: 1.109-1.316 and HR: 1.636, 95% CI: 1.203-2.224, respectively). The ROC analysis indicated comparable predictive accuracy for all three biomarkers, with AST/ALT showing slightly superior performance. CONCLUSION Liver fibrosis markers, including AST/ALT, NFS, Forns index and FIB-4, are significant mortality predictors in CAD-CKD patients. The AST/ALT ratio, being easily measurable, may serve as an effective predictive tool for risk stratification in this population.
Collapse
Affiliation(s)
- Zixiang Ye
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Cardiology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Enmin Xie
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ziyu Guo
- Department of Cardiology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Yanxiang Gao
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, China
| | - Zhongwei Han
- Department of Cardiology, Yantai Muping District Hospital of Traditional Chinese Medicine, Yantai, China
| | - Kefei Dou
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jingang Zheng
- Department of Cardiology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
7
|
Feng H, Li Z, Zheng R. Bexarotene ameliorated the pulmonary inflammation and M1 polarization of alveolar macrophages induced by cigarette smoke via PPARγ/HO-1. Respir Res 2024; 25:431. [PMID: 39696251 DOI: 10.1186/s12931-024-03064-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 12/06/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Alveolar macrophages (AMs) modulate pulmonary inflammation in chronic obstructive pulmonary disease (COPD), contributing to its progression. The PPARγ/RXRα heterodimer influences AM polarization induced by cigarette smoke (CS). Although PPARγ agonists suppress CS-induced M1 macrophage polarization, the impact of RXRα agonists on this process has not been determined. This study explored the effects and mechanisms of the RXRα agonist bexarotene on macrophage polarization in a COPD mouse model. METHODS C57BL/6 mice were assigned to the control, model, bexarotene, or model + bexarotene group. The COPD model was induced by CS exposure and intraperitoneal injection of cigarette smoke extract (CSE), followed by intraperitoneal administration of bexarotene. Additionally, MH-S cells were exposed to CSE and bexarotene. Lung tissues were subjected to hematoxylin-eosin staining, and emphysema and inflammatory scores were assessed. Cytokine levels and cell differentials in bronchoalveolar lavage fluid were measured, and macrophage polarization was evaluated using immunohistochemistry, flow cytometry, and qPCR. RESULTS Bexarotene effectively reduced inflammatory scores, cytokine levels, and neutrophil counts and ameliorated emphysema and M1 polarization of AMs in COPD model mice. Furthermore, while CSE exposure reduced PPARγ expression and the transcriptional activity of AMs, bexarotene enhanced the transcriptional response of PPARγ to CSE. HO-1 was identified as a potential target of PPARγ; its levels were assessed in AMs, revealing that bexarotene mitigated the CSE-induced reduction in HO-1. Notably, the effect of bexarotene was partially inhibited by the PPARγ inhibitor. CONCLUSIONS Our results indicated that bexarotene may curb inflammation and M1 polarization in COPD through activation of the PPARγ/HO-1 pathway.
Collapse
Affiliation(s)
- Haoshen Feng
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, No.36 Sanhao Street, Shenyang, 110004, Liaoning, China
| | - Zhe Li
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, PR China
| | - Rui Zheng
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, No.36 Sanhao Street, Shenyang, 110004, Liaoning, China.
| |
Collapse
|
8
|
Li Z, Li J, Wu M, Li Z, Zhou J, Lu Y, Xu Y, Qin L, Fan Z. Redox-sensitive epigenetic activation of SUV39H1 contributes to liver ischemia-reperfusion injury. Redox Biol 2024; 78:103414. [PMID: 39603205 PMCID: PMC11635714 DOI: 10.1016/j.redox.2024.103414] [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: 09/18/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 11/29/2024] Open
Abstract
Liver ischemia-reperfusion (I/R) injury is a clinically relevant pathophysiological process that determines the effectiveness of life-saving liver transplantation, to which aberrant ROS accumulation plays a key role. In the present study we investigated the role of SUV39H1, a lysine methyltransferases, in this process focusing on regulatory mechanism and translational potential. We report that SUV39H1 expression was up-regulated in the liver tissues of mice subjected to ischemia-reperfusion and in hepatocytes exposed to hypoxia-reoxygenation (H/R) in a redox-sensitive manner. Mechanistically, coactivator associated arginine methyltransferases 1 (CARM1) mediated redox-sensitive Suv39h1 trans-activation by promoting histone H3R17 methylation. Consistently, pharmaceutical CARM1 inhibition attenuated liver I/R injury. In addition, global or hepatocyte conditional Suv39h1 KO mice were protected from liver I/R injury. RNA-seq revealed that aldehyde dehydrogenase 1 family 1a (Aldh1a1) as a novel target for SUV39H1. SUV39H1 directly bound to the Aldh1a1 promoter and repressed Aldh1a1 transcription in H/R-challenged hepatocytes. ALDH1A1 silencing abrogated the protective effects of SUV39H1 deficiency on H/R-inflicted injuries whereas ALDH1A1 over-expression mitigated liver I/R injury in mice. Importantly, administration of a small-molecule SUV39H1 inhibitor achieved similar hepatoprotective effects as SUV39H1 deletion. Finally, increased Suv39h1 expression and decreased Aldh1a1 expression were observed in liver I/R specimens in humans. In conclusion, our data uncover a regulatory role for SUV39H1 in liver I/R injury and serve as proof-of-concept that targeting the SUV39H1-ALDH1A1 axis might be considered as a reasonable approach for the intervention of liver I/R injury.
Collapse
Affiliation(s)
- Zilong Li
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing, China; Institute of Brain Science and Brain-inspired Research, Shandong First Medical University, Jinan, China.
| | - Jichen Li
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Meng Wu
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Zexin Li
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Jiawen Zhou
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Yunjie Lu
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yong Xu
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing, China; Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Translational Medicine, Nanjing Medical University, Nanjing, China; Institute of Biomedical Research, College of Agriculture and Biology, Liaocheng University, Liaocheng, China.
| | - Lei Qin
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital of Soochow University, Suzhou, China.
| | - Zhiwen Fan
- Department of Pathology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, China.
| |
Collapse
|
9
|
Consonni FM, Incerti M, Bertolotti M, Ballerini G, Garlatti V, Sica A. Heme catabolism and heme oxygenase-1-expressing myeloid cells in pathophysiology. Front Immunol 2024; 15:1433113. [PMID: 39611159 PMCID: PMC11604077 DOI: 10.3389/fimmu.2024.1433113] [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: 05/15/2024] [Accepted: 10/01/2024] [Indexed: 11/30/2024] Open
Abstract
Although the pathological significance of myeloid cell heterogeneity is still poorly understood, new evidence indicates that distinct macrophage subsets are characterized by specific metabolic programs that influence disease onset and progression. Within this scenario, distinct subsets of macrophages, endowed with high rates of heme catabolism by the stress-responsive enzyme heme oxygenase-1 (HO-1), play critical roles in physiologic and pathological conditions. Of relevance, the substrates of HO-1 activity are the heme groups that derive from cellular catabolism and are converted into carbon monoxide (CO), biliverdin and Fe2+, which together elicit anti-apoptotic, anti-inflammatory activities and control oxidative damage. While high levels of expression of HO-1 enzyme by specialized macrophage populations (erythrophagocytes) guarantee the physiological disposal of senescent red blood cells (i.e. erythrocateresis), the action of HO-1 takes on pathological significance in various diseases, and abnormal CO metabolism has been observed in cancer, hematological diseases, hypertension, heart failure, inflammation, sepsis, neurodegeneration. Modulation of heme catabolism and CO production is therefore a feasible therapeutic opportunity in various diseases. In this review we discuss the role of HO-1 in different pathological contexts (i.e. cancer, infections, cardiovascular, immune-mediated and neurodegenerative diseases) and highlight new therapeutic perspectives on the modulation of the enzymatic activity of HO-1.
Collapse
Affiliation(s)
- Francesca Maria Consonni
- Department of Pharmaceutical Sciences, University of Piemonte Orientale “A. Avogadro”, Novara, Italy
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Martina Incerti
- Department of Pharmaceutical Sciences, University of Piemonte Orientale “A. Avogadro”, Novara, Italy
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Milena Bertolotti
- Navita S.r.l., University of Eastern Piedmont A. Avogadro, Novara, Italy
| | - Giulia Ballerini
- Department of Pharmaceutical Sciences, University of Piemonte Orientale “A. Avogadro”, Novara, Italy
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Valentina Garlatti
- Department of Pharmaceutical Sciences, University of Piemonte Orientale “A. Avogadro”, Novara, Italy
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Antonio Sica
- Department of Pharmaceutical Sciences, University of Piemonte Orientale “A. Avogadro”, Novara, Italy
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| |
Collapse
|
10
|
Li M, Tan J, Zhang R, Gong X, Xie J, Liu C, Wu C, Li X. Sunitinib alleviates hepatic ischemia reperfusion injury by inhibiting the JAK2/STAT pathway and promoting the M2 polarization of macrophages. Immunopharmacol Immunotoxicol 2024:1-13. [PMID: 39155607 DOI: 10.1080/08923973.2024.2390455] [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: 12/26/2023] [Accepted: 07/22/2024] [Indexed: 08/20/2024]
Abstract
BACKGROUND Hepatic ischemia reperfusion injury (IRI) is a common liver surgery complication. This study aims to explore the effect and potential mechanism of Sunitinib - a multi-target tyrosine kinase inhibitor - on hepatic IRI. METHODS We established a hepatic IRI model using C57BL/6 mice, and integrated 40 mg/kg of Sunitinib, solely or combined with 100 μg/kg of coumermycin A1 (C-A1), in the treatment strategy. H&E staining, TUNEL assay, and detection of serum ALT and AST activities were used to assess liver damage. Further, ELISA kits and Western Blots were utilized to determine IL-1β, TNF-α, IL-6, CXCL10, and CXCL2 levels. Primary macrophages, once isolated, were cultured in vitro with either 2 nM of Sunitinib, or Sunitinib in conjunction with 1 μM of C-A1, to gauge their influence on macrophage polarization. qPCR and Western blot were conducted to examine the level of p-STAT1/STAT1, p-STAT3/STAT3, p-JAK2/JAK2, and M1/M2 polarization markers. To quantify immune cell infiltration, we applied Immunofluorescence. RESULTS Sunitinib pretreatment significantly alleviated liver injury and reduced p-STAT1/STAT1, p-STAT3/STAT3, p-JAK2/JAK2 levels. In vitro, Sunitinib treatment curbed M1 polarization induced by LPS + IFN-γ and bolstered M2 polarization triggered by IL-4. C-A1 application upregulated JAK2/STAT pathway phosphorylation and promoted LPS + IFN-γ-induced M1 polarization, which was reversed by Sunitinib treatment. In IL-4-stimulated macrophages, application of C-A1 activated the JAK2/STAT pathway and decreased M2-type macrophages, which was reversed by Sunitinib treatment either. CONCLUSION Sunitinib is capable of guiding the polarization of macrophages toward an M2-type phenotype via the inhibition of the JAK2/STAT pathway, thereby exerting a protective effect on hepatic IRI.
Collapse
Affiliation(s)
- Mingxia Li
- Department of Anesthesiology, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, China
| | - Juan Tan
- Research Associate Department of Pathology, The Xiangya Third Hospital, Central South University, Changsha, China
| | - Rongsen Zhang
- Department of Ultrasound Diagnosis, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoxiang Gong
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jun Xie
- Department of General Surgery, Hengdong County People's Hospital, Hengdong County, Hengyang, China
| | - Cong Liu
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Chenhao Wu
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiaojing Li
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
11
|
Ludwig N, Cucinelli S, Hametner S, Muckenthaler MU, Schirmer L. Iron scavenging and myeloid cell polarization. Trends Immunol 2024; 45:625-638. [PMID: 39054114 DOI: 10.1016/j.it.2024.06.006] [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: 06/05/2024] [Revised: 06/25/2024] [Accepted: 06/27/2024] [Indexed: 07/27/2024]
Abstract
Myeloid cells that populate all human organs and blood are a versatile class of innate immune cells. They are crucial for sensing and regulating processes as diverse as tissue homeostasis and inflammation and are frequently characterized by their roles in either regulating or promoting inflammation. Recent studies in cultured cells and mouse models highlight the role of iron in skewing the functional properties of myeloid cells in tissue damage and repair. Here, we review certain emerging concepts on how iron influences and determines myeloid cell polarization in the context of its uptake, storage, and metabolism, including in conditions such as multiple sclerosis (MS), sickle cell disease, and tumors.
Collapse
Affiliation(s)
- Natalie Ludwig
- Department of Neurology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany
| | - Stefania Cucinelli
- Department of Paediatric Hematology, Oncology, and Immunology, University of Heidelberg, Heidelberg, Germany; Molecular Medicine Partnership Unit (MMPU), European Molecular Biology Laboratory and University of Heidelberg, Heidelberg, Germany
| | - Simon Hametner
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria; Medical Neuroscience Cluster, Medical University of Vienna, Vienna, Austria
| | - Martina U Muckenthaler
- Department of Paediatric Hematology, Oncology, and Immunology, University of Heidelberg, Heidelberg, Germany; Molecular Medicine Partnership Unit (MMPU), European Molecular Biology Laboratory and University of Heidelberg, Heidelberg, Germany; German Centre for Cardiovascular Research (DZHK), Partner site Heidelberg/Mannheim, Heidelberg, Germany; Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany.
| | - Lucas Schirmer
- Department of Neurology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany; Mannheim Center for Translational Neuroscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Mannheim Institute for Innate Immunoscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
| |
Collapse
|
12
|
Chen M, Ji T, Liu YY, Liu WL, Yan XT, Jiang HX, Zhang ZZ, He XH. Emodin alleviates intestinal ischemia/reperfusion-induced lung injury by upregulating HO-1 expression via PI3K/AkT pathway. Surgery 2024; 176:499-510. [PMID: 38811326 DOI: 10.1016/j.surg.2024.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 03/16/2024] [Accepted: 04/06/2024] [Indexed: 05/31/2024]
Abstract
BACKGROUND Emodin, a natural anthraquinone derivative found in various Chinese medicinal herbs, has been proved to be an effective therapeutic agent in the treatment of many diseases. However, its effect on lung injury after intestinal ischemia/reperfusion injury remains unknown. This research was designed to investigate whether emodin protects against intestinal ischemia/reperfusion-induced lung injury and to elucidate the underlying molecular mechanisms in vivo and in vitro. METHODS Intestinal ischemia/reperfusion injury was induced by occluding the superior mesenteric artery in mice, and mouse lung epithelial-12 cells were subjected to oxygen-glucose deprivation and reoxygenation to establish an in vitro model. RESULTS Our data indicated that emodin treatment reduced intestinal ischemia/reperfusion-induced oxidative stress, inflammation and apoptosis in lung tissues and alleviated lung injury. However, the protective effects of emodin on intestinal ischemia/reperfusion-induced lung injury were reversed by the protein kinase B inhibitor triciribine or the heme oxygenase-1 inhibitor tin protoporphyrin IX. The protein kinase inhibitor triciribine also downregulated the expression of heme oxygenase-1. CONCLUSION In conclusion, our data suggest that emodin treatment protects against intestinal ischemia/reperfusion-induced lung injury by enhancing heme oxygenase-1 expression via activation of the PI3K/protein kinase pathway. Emodin may act as a potential therapeutic agent for the prevention and treatment of lung injury induced by intestinal ischemia/reperfusion.
Collapse
Affiliation(s)
- Meng Chen
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Hubei, China; Department of Anesthesiology, Maternal and Child Health Hospital of Hubei Province, Wuhan, Hubei, China
| | - Tuo Ji
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Hubei, China; Department of Anesthesiology, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Yin-Yin Liu
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Hubei, China
| | - Wan-Li Liu
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Hubei, China
| | - Xue-Tao Yan
- Department of Anesthesiology, Shenzhen Bao'an Maternity and Child Health Hospital, China
| | - Hai-Xing Jiang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Hubei, China
| | - Zong-Ze Zhang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Hubei, China
| | - Xiang-Hu He
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Hubei, China.
| |
Collapse
|
13
|
Duarte TL, Viveiros N, Godinho C, Duarte D. Heme (dys)homeostasis and liver disease. Front Physiol 2024; 15:1436897. [PMID: 39135705 PMCID: PMC11317413 DOI: 10.3389/fphys.2024.1436897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 07/15/2024] [Indexed: 08/15/2024] Open
Abstract
Heme is essential for a variety of proteins involved in vital physiological functions in the body, such as oxygen transport, drug metabolism, biosynthesis of steroids, signal transduction, antioxidant defense and mitochondrial respiration. However, free heme is potentially cytotoxic due to the capacity of heme iron to promote the oxidation of cellular molecules. The liver plays a central role in heme metabolism by significantly contributing to heme synthesis, heme detoxification, and recycling of heme iron. Conversely, enzymatic defects in the heme biosynthetic pathway originate multisystemic diseases (porphyrias) that are highly associated with liver damage. In addition, there is growing evidence that heme contributes to the outcomes of inflammatory, metabolic and malignant liver diseases. In this review, we summarize the contribution of the liver to heme metabolism and the association of heme dyshomeostasis with liver disease.
Collapse
Affiliation(s)
- Tiago L. Duarte
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC–Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Nicole Viveiros
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Catarina Godinho
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Faculdade de Medicina da Universidade do Porto (FMUP), Porto, Portugal
| | - Delfim Duarte
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Serviço de Hematologia e Transplantação da Medula Óssea, Instituto Português de Oncologia do Porto Francisco Gentil, E.P.E. (IPO Porto), Porto, Portugal
- Departamento de Biomedicina, Faculdade de Medicina da Universidade do Porto (FMUP), Porto, Portugal
| |
Collapse
|
14
|
Zhang J, Pei J, Yu C, Luo J, Hong Y, Hua Y, Wei G. CCR7 and CD48 as Predicted Targets in Acute Rejection Related to M1 Macrophage after Pediatric Kidney Transplantation. J Immunol Res 2024; 2024:6908968. [PMID: 38957433 PMCID: PMC11217580 DOI: 10.1155/2024/6908968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 05/28/2024] [Accepted: 06/12/2024] [Indexed: 07/04/2024] Open
Abstract
Background Kidney transplantation (KT) is the best treatment for end-stage renal disease. Although long and short-term survival rates for the graft have improved significantly with the development of immunosuppressants, acute rejection (AR) remains a major risk factor attacking the graft and patients. The innate immune response plays an important role in rejection. Therefore, our objective is to determine the biomarkers of congenital immunity associated with AR after KT and provide support for future research. Materials and Methods A differential expression genes (DEGs) analysis was performed based on the dataset GSE174020 from the NCBI gene Expression Synthesis Database (GEO) and then combined with the GSE5099 M1 macrophage-related gene identified in the Molecular Signatures Database. We then identified genes in DEGs associated with M1 macrophages defined as DEM1Gs and performed gene ontology (GO) and Kyoto Encyclopedia of Genomes (KEGG) enrichment analysis. Cibersort was used to analyze the immune cell infiltration during AR. At the same time, we used the protein-protein interaction (PPI) network and Cytoscape software to determine the key genes. Dataset, GSE14328 derived from pediatric patients, GSE138043 and GSE9493 derived from adult patients, were used to verify Hub genes. Additional verification was the rat KT model, which was used to perform HE staining, immunohistochemical staining, and Western Blot. Hub genes were searched in the HPA database to confirm their expression. Finally, we construct the interaction network of transcription factor (TF)-Hub genes and miRNA-Hub genes. Results Compared to the normal group, 366 genes were upregulated, and 423 genes were downregulated in the AR group. Then, 106 genes related to M1 macrophages were found among these genes. GO and KEGG enrichment analysis showed that these genes are mainly involved in cytokine binding, antigen binding, NK cell-mediated cytotoxicity, activation of immune receptors and immune response, and activation of the inflammatory NF-κB signaling pathway. Two Hub genes, namely CCR7 and CD48, were identified by PPI and Cytoscape analysis. They have been verified in external validation sets, originated from both pediatric patients and adult patients, and animal experiments. In the HPA database, CCR7 and CD48 are mainly expressed in T cells, B cells, macrophages, and tissues where these immune cells are distributed. In addition to immunoinfiltration, CD4+T, CD8+T, NK cells, NKT cells, and monocytes increased significantly in the AR group, which was highly consistent with the results of Hub gene screening. Finally, we predicted that 19 TFs and 32 miRNAs might interact with the Hub gene. Conclusions Through a comprehensive bioinformatic analysis, our findings may provide predictive and therapeutic targets for AR after KT.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Urology Children's Hospital of Chongqing Medical UniversityNational Clinical Research Center for Child Health and DisordersMinistry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing, China
| | - Jun Pei
- Department of Urology Children's Hospital of Chongqing Medical UniversityNational Clinical Research Center for Child Health and DisordersMinistry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing, China
| | - Chengjun Yu
- Department of Urology Children's Hospital of Chongqing Medical UniversityNational Clinical Research Center for Child Health and DisordersMinistry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing, China
| | - Jin Luo
- Department of Urology Children's Hospital of Chongqing Medical UniversityNational Clinical Research Center for Child Health and DisordersMinistry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing, China
| | - Yifan Hong
- Department of Urology Children's Hospital of Chongqing Medical UniversityNational Clinical Research Center for Child Health and DisordersMinistry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing, China
| | - Yi Hua
- Department of Urology Children's Hospital of Chongqing Medical UniversityNational Clinical Research Center for Child Health and DisordersMinistry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing, China
| | - Guanghui Wei
- Department of Urology Children's Hospital of Chongqing Medical UniversityNational Clinical Research Center for Child Health and DisordersMinistry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing, China
| |
Collapse
|
15
|
Chang C, Gupta R, Sedighian F, Louie A, Gonzalez DM, Le C, Cho JM, Park SK, Castellanos J, Ting TW, Dong TS, Arias-Jayo N, Lagishetty V, Navab M, Reddy S, Sioutas C, Hsiai T, Jacobs JP, Araujo JA. Subchronic inhalation exposure to ultrafine particulate matter alters the intestinal microbiome in various mouse models. ENVIRONMENTAL RESEARCH 2024; 248:118242. [PMID: 38242419 PMCID: PMC11737635 DOI: 10.1016/j.envres.2024.118242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 01/13/2024] [Accepted: 01/17/2024] [Indexed: 01/21/2024]
Abstract
Exposure to ultrafine particles (UFPs) has been associated with multiple adverse health effects. Inhaled UFPs could reach the gastrointestinal tract and influence the composition of the gut microbiome. We have previously shown that oral ingestion of UFPs alters the gut microbiome and promotes intestinal inflammation in hyperlipidemic Ldlr-/- mice. Particulate matter (PM)2.5 inhalation studies have also demonstrated microbiome shifts in normolipidemic C57BL/6 mice. However, it is not known whether changes in microbiome precede or follow inflammatory effects in the intestinal mucosa. We hypothesized that inhaled UFPs modulate the gut microbiome prior to the development of intestinal inflammation. We studied the effects of UFP inhalation on the gut microbiome and intestinal mucosa in two hyperlipidemic mouse models (ApoE-/- mice and Ldlr-/- mice) and normolipidemic C57BL/6 mice. Mice were exposed to PM in the ultrafine-size range by inhalation for 6 h a day, 3 times a week for 10 weeks at a concentration of 300-350 μg/m3.16S rRNA gene sequencing was performed to characterize sequential changes in the fecal microbiome during exposures, and changes in the intestinal microbiome at the end. PM exposure led to progressive differentiation of the microbiota over time, associated with increased fecal microbial richness and evenness, altered microbial composition, and differentially abundant microbes by week 10 depending on the mouse model. Cross-sectional analysis of the small intestinal microbiome at week 10 showed significant changes in α-diversity, β-diversity, and abundances of individual microbial taxa in the two hyperlipidemic models. These alterations of the intestinal microbiome were not accompanied, and therefore could not be caused, by increased intestinal inflammation as determined by histological analysis of small and large intestine, cytokine gene expression, and levels of fecal lipocalin. In conclusion, 10-week inhalation exposures to UFPs induced taxonomic changes in the microbiome of various animal models in the absence of intestinal inflammation.
Collapse
Affiliation(s)
- Candace Chang
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA; Division of Cardiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA; Department of Environmental Health Sciences, Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA, USA; Goodman-Luskin Microbiome Center, University of California Los Angeles, Los Angeles, CA, USA; Molecular Toxicology Interdepartmental Program, University of California Los Angeles, Los Angeles, CA, USA
| | - Rajat Gupta
- Division of Cardiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA; Department of Environmental Health Sciences, Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA, USA; Molecular Toxicology Interdepartmental Program, University of California Los Angeles, Los Angeles, CA, USA
| | - Farzaneh Sedighian
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Allen Louie
- Division of Cardiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA; Department of Environmental Health Sciences, Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA, USA; Molecular Toxicology Interdepartmental Program, University of California Los Angeles, Los Angeles, CA, USA
| | - David M Gonzalez
- Division of Cardiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA; Molecular Toxicology Interdepartmental Program, University of California Los Angeles, Los Angeles, CA, USA
| | - Collin Le
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Jae Min Cho
- Division of Cardiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Seul-Ki Park
- Division of Cardiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Jocelyn Castellanos
- Division of Cardiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA; Department of Environmental Health Sciences, Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA, USA
| | - To-Wei Ting
- Department of Environmental Health Sciences, Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA, USA
| | - Tien S Dong
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA; Goodman-Luskin Microbiome Center, University of California Los Angeles, Los Angeles, CA, USA; Division of Gastroenterology, Hepatology and Parenteral Nutrition, Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA, Los Angeles, CA, USA
| | - Nerea Arias-Jayo
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA; Goodman-Luskin Microbiome Center, University of California Los Angeles, Los Angeles, CA, USA
| | - Venu Lagishetty
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA; Goodman-Luskin Microbiome Center, University of California Los Angeles, Los Angeles, CA, USA
| | - Mohamad Navab
- Division of Cardiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Srinivasa Reddy
- Division of Cardiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA; Molecular Toxicology Interdepartmental Program, University of California Los Angeles, Los Angeles, CA, USA; Molecular & Medical Pharmacology, University of California Los Angeles, Los Angeles, CA, USA
| | - Constantinos Sioutas
- University of Southern California (USC) Viterbi School of Engineering, Los Angeles, CA, USA
| | - Tzung Hsiai
- Division of Cardiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA; Henry Samueli School of Engineering, University of California Los Angeles, Los Angeles, CA, USA
| | - Jonathan P Jacobs
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA; Goodman-Luskin Microbiome Center, University of California Los Angeles, Los Angeles, CA, USA; Molecular Toxicology Interdepartmental Program, University of California Los Angeles, Los Angeles, CA, USA; Division of Gastroenterology, Hepatology and Parenteral Nutrition, Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA, Los Angeles, CA, USA.
| | - Jesus A Araujo
- Division of Cardiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA; Department of Environmental Health Sciences, Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA, USA; Molecular Toxicology Interdepartmental Program, University of California Los Angeles, Los Angeles, CA, USA; Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
16
|
Berendes LS, Westhoff PS, Wittkowski H, Seelhöfer A, Varga G, Marquardt T, Park JH. Clinical and molecular analysis of a novel variant in heme oxygenase-1 deficiency: Unraveling its role in inflammation, heme metabolism, and pulmonary phenotype. Mol Genet Metab Rep 2024; 38:101038. [PMID: 38178812 PMCID: PMC10764348 DOI: 10.1016/j.ymgmr.2023.101038] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 12/08/2023] [Accepted: 12/08/2023] [Indexed: 01/06/2024] Open
Abstract
Heme oxygenase 1 (HO-1) is the pivotal catalyst for the primary and rate-determining step in heme catabolism, playing a crucial role in mitigating heme-induced oxidative damage. Pathogenic variants in the HMOX1 gene which encodes HO-1, are responsible for a severe, multisystem disease characterized by recurrent inflammatory episodes, organ failure, and an ultimately fatal course. Chronic hemolysis and abnormally low bilirubin levels are cardinal laboratory features of this disorder. In this study, we describe a patient with severe interstitial lung disease, frequent episodes of hyperinflammation non-responsive to immunosuppression, and fatal pulmonary hemorrhage. Employing exome sequencing, we identified two protein truncating variants in HMOX1, c.262_268delinsCC (p.Ala88Profs*51) and a previously unreported variant, c.55dupG (p.Glu19Glyfs*14). Functional analysis in patient-derived lymphoblastoid cells unveiled the complete absence of HO-1 protein expression and a marked reduction in cell viability upon exposure to hemin. These findings confirm the pathogenicity of the identified HMOX1 variants, further underscoring their association with severe pulmonary manifestations . This study describes the profound clinical consequences stemming from disruptions in redox metabolism.
Collapse
Affiliation(s)
| | | | - Helmut Wittkowski
- University of Münster, Department of Pediatric Rheumatology and Immunology, Münster, Germany
| | - Anja Seelhöfer
- University of Münster, Department of General Pediatrics, Münster, Germany
| | - Georg Varga
- University of Münster, Department of Pediatric Rheumatology and Immunology, Münster, Germany
| | - Thorsten Marquardt
- University of Münster, Department of General Pediatrics, Münster, Germany
| | - Julien H. Park
- University of Münster, Department of General Pediatrics, Münster, Germany
| |
Collapse
|
17
|
Minayoshi Y, Maeda H, Hamasaki K, Nagasaki T, Takano M, Fukuda R, Mizuta Y, Tanaka M, Sasaki Y, Otagiri M, Watanabe H, Maruyama T. Mouse Type-I Interferon-Mannosylated Albumin Fusion Protein for the Treatment of Chronic Hepatitis. Pharmaceuticals (Basel) 2024; 17:260. [PMID: 38399475 PMCID: PMC10893114 DOI: 10.3390/ph17020260] [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: 12/31/2023] [Revised: 02/03/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
Although a lot of effort has been put into creating drugs and combination therapies against chronic hepatitis, no effective treatment has been established. Type-I interferon is a promising therapeutic for chronic hepatitis due to its excellent anti-inflammatory effects through interferon receptors on hepatic macrophages. To develop a type-I IFN equipped with the ability to target hepatic macrophages through the macrophage mannose receptor, the present study designed a mouse type-I interferon-mannosylated albumin fusion protein using site-specific mutagenesis and albumin fusion technology. This fusion protein exhibited the induction of anti-inflammatory molecules, such as IL-10, IL-1Ra, and PD-1, in RAW264.7 cells, or hepatoprotective effects on carbon tetrachloride-induced chronic hepatitis mice. As expected, such biological and hepatoprotective actions were significantly superior to those of human fusion proteins. Furthermore, the repeated administration of mouse fusion protein to carbon tetrachloride-induced chronic hepatitis mice clearly suppressed the area of liver fibrosis and hepatic hydroxyproline contents, not only with a reduction in the levels of inflammatory cytokine (TNF-α) and fibrosis-related genes (TGF-β, Fibronectin, Snail, and Collagen 1α2), but also with a shift in the hepatic macrophage phenotype from inflammatory to anti-inflammatory. Therefore, type-I interferon-mannosylated albumin fusion protein has the potential as a new therapeutic agent for chronic hepatitis.
Collapse
Affiliation(s)
- Yuki Minayoshi
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-Honmachi, Chuo-ku, Kumamoto 862-0973, Japan; (Y.M.); (K.H.); (T.N.); (M.T.); (R.F.); (Y.M.); (H.W.)
| | - Hitoshi Maeda
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-Honmachi, Chuo-ku, Kumamoto 862-0973, Japan; (Y.M.); (K.H.); (T.N.); (M.T.); (R.F.); (Y.M.); (H.W.)
| | - Keisuke Hamasaki
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-Honmachi, Chuo-ku, Kumamoto 862-0973, Japan; (Y.M.); (K.H.); (T.N.); (M.T.); (R.F.); (Y.M.); (H.W.)
| | - Taisei Nagasaki
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-Honmachi, Chuo-ku, Kumamoto 862-0973, Japan; (Y.M.); (K.H.); (T.N.); (M.T.); (R.F.); (Y.M.); (H.W.)
| | - Mei Takano
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-Honmachi, Chuo-ku, Kumamoto 862-0973, Japan; (Y.M.); (K.H.); (T.N.); (M.T.); (R.F.); (Y.M.); (H.W.)
| | - Ryo Fukuda
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-Honmachi, Chuo-ku, Kumamoto 862-0973, Japan; (Y.M.); (K.H.); (T.N.); (M.T.); (R.F.); (Y.M.); (H.W.)
| | - Yuki Mizuta
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-Honmachi, Chuo-ku, Kumamoto 862-0973, Japan; (Y.M.); (K.H.); (T.N.); (M.T.); (R.F.); (Y.M.); (H.W.)
| | - Motohiko Tanaka
- Department of Gastroenterology and Hepatology, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556, Japan; (M.T.); (Y.S.)
- Public Health and Welfare Bureau, 5-1-1 Oe, Chuo-ku, Kumamoto 862-0971, Japan
| | - Yutaka Sasaki
- Department of Gastroenterology and Hepatology, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556, Japan; (M.T.); (Y.S.)
- Osaka Central Hospital, 3-3-30 Umeda, Kita-ku, Osaka 530-0001, Japan
| | - Masaki Otagiri
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto 860-0082, Japan;
- DDS Research Institute, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto 860-0082, Japan
| | - Hiroshi Watanabe
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-Honmachi, Chuo-ku, Kumamoto 862-0973, Japan; (Y.M.); (K.H.); (T.N.); (M.T.); (R.F.); (Y.M.); (H.W.)
| | - Toru Maruyama
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-Honmachi, Chuo-ku, Kumamoto 862-0973, Japan; (Y.M.); (K.H.); (T.N.); (M.T.); (R.F.); (Y.M.); (H.W.)
| |
Collapse
|
18
|
He J, Tang MY, Liu LX, Kong CX, Chen W, Wang L, Zhi SB, Sun HW, Huang YC, Chen GY, Xin HB, Deng KY. Myeloid Deletion of Cdc42 Protects Liver From Hepatic Ischemia-Reperfusion Injury via Inhibiting Macrophage-Mediated Inflammation in Mice. Cell Mol Gastroenterol Hepatol 2024; 17:965-981. [PMID: 38342302 PMCID: PMC11047801 DOI: 10.1016/j.jcmgh.2024.01.023] [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: 09/14/2023] [Revised: 01/31/2024] [Accepted: 01/31/2024] [Indexed: 02/13/2024]
Abstract
BACKGROUND & AIMS Hepatic ischemia-reperfusion injury (HIRI) often occurs in liver surgery, such as partial hepatectomy and liver transplantation, in which myeloid macrophage-mediated inflammation plays a critical role. Cell division cycle 42 (Cdc42) regulates cell migration, cytoskeleton rearrangement, and cell polarity. In this study, we explore the role of myeloid Cdc42 in HIRI. METHODS Mouse HIRI models were established with 1-hour ischemia followed by 12-hour reperfusion in myeloid Cdc42 knockout (Cdc42mye) and Cdc42flox mice. Myeloid-derived macrophages were traced with RosamTmG fluorescent reporter under LyzCre-mediated excision. The experiments for serum or hepatic enzymic activities, histologic and immunologic analysis, gene expressions, flow cytometry analysis, and cytokine antibody array were performed. RESULTS Myeloid deletion of Cdc42 significantly alleviated hepatic damages with the reduction of hepatic necrosis and inflammation, and reserved hepatic functions following HIRI in mice. Myeloid Cdc42 deficiency suppressed the infiltration of myeloid macrophages, reduced the secretion of proinflammatory cytokines, restrained M1 polarization, and promoted M2 polarization of myeloid macrophages in livers. In addition, inactivation of Cdc42 promoted M2 polarization via suppressing the phosphorylation of STAT1 and promoting phosphorylation of STAT3 and STAT6 in myeloid macrophages. Furthermore, pretreatment with Cdc42 inhibitor, ML141, also protected mice from hepatic ischemia-reperfusion injury. CONCLUSIONS Inhibition or deletion of myeloid Cdc42 protects liver from HIRI via restraining the infiltration of myeloid macrophages, suppressing proinflammatory response, and promoting M2 polarization in macrophages.
Collapse
Affiliation(s)
- Jing He
- The National Engineering Research Center for Bioengineering Drugs and Technology, Institution of Translation Medicine, Nanchang University, Nanchang, Jiangxi, PR China
| | - Meng-Yu Tang
- College of Life Science, Nanchang University, Nanchang, Jiangxi, PR China
| | - Li-Xin Liu
- The National Engineering Research Center for Bioengineering Drugs and Technology, Institution of Translation Medicine, Nanchang University, Nanchang, Jiangxi, PR China; College of Pharmacy, Nanchang University, Nanchang, Jiangxi, PR China
| | - Chen-Xian Kong
- College of Life Science, Nanchang University, Nanchang, Jiangxi, PR China
| | - Wen Chen
- College of Life Science, Nanchang University, Nanchang, Jiangxi, PR China
| | - Lu Wang
- The National Engineering Research Center for Bioengineering Drugs and Technology, Institution of Translation Medicine, Nanchang University, Nanchang, Jiangxi, PR China
| | - Shao-Bin Zhi
- The National Engineering Research Center for Bioengineering Drugs and Technology, Institution of Translation Medicine, Nanchang University, Nanchang, Jiangxi, PR China
| | - Hong-Wei Sun
- The National Engineering Research Center for Bioengineering Drugs and Technology, Institution of Translation Medicine, Nanchang University, Nanchang, Jiangxi, PR China
| | - Yu-Chun Huang
- The National Engineering Research Center for Bioengineering Drugs and Technology, Institution of Translation Medicine, Nanchang University, Nanchang, Jiangxi, PR China
| | - Guo-Yu Chen
- The First Clinical Medical College, Nanchang University, Nanchang, Jiangxi, PR China
| | - Hong-Bo Xin
- The National Engineering Research Center for Bioengineering Drugs and Technology, Institution of Translation Medicine, Nanchang University, Nanchang, Jiangxi, PR China; College of Life Science, Nanchang University, Nanchang, Jiangxi, PR China; College of Pharmacy, Nanchang University, Nanchang, Jiangxi, PR China.
| | - Ke-Yu Deng
- The National Engineering Research Center for Bioengineering Drugs and Technology, Institution of Translation Medicine, Nanchang University, Nanchang, Jiangxi, PR China; College of Life Science, Nanchang University, Nanchang, Jiangxi, PR China; College of Pharmacy, Nanchang University, Nanchang, Jiangxi, PR China.
| |
Collapse
|
19
|
Zeng J, Zhang Y, Huang C. Macrophages polarization in renal inflammation and fibrosis animal models (Review). Mol Med Rep 2024; 29:29. [PMID: 38131228 PMCID: PMC10784723 DOI: 10.3892/mmr.2023.13152] [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] [Accepted: 11/01/2023] [Indexed: 12/23/2023] Open
Abstract
Chronic kidney disease (CKD) is a significant public health concern. Renal fibrosis is the final common pathway in the progression of kidney diseases, irrespective of the initial injury. Substantial evidence underscores the pivotal role of renal inflammation in the genesis of renal fibrosis. The presence of macrophages within normal renal tissue is significantly increased within diseased renal tissue, indicative of their crucial regulatory function in inflammation and fibrosis. Macrophages manifest a high degree of heterogeneity, exhibiting distinct phenotypic and functional traits in response to diverse stimuli within the local microenvironment in various types of kidney diseases. Broadly, macrophages are categorized into two principal groups: Classically activated, designated as M1 macrophages and alternatively activated, designated as M2 macrophages. A number of experimental models are widely used to study the underlying mechanisms driving renal inflammation and fibrosis progression. The present review delineated the phenotypic and functional attributes of macrophages present in diverse induced models, analyzing their disposition in relation to M1 and M2 polarization states.
Collapse
Affiliation(s)
- Ji Zeng
- Department of Pharmacy, Ma'anshan City Hospital of Traditional Chinese Medicine, Ma'anshan, Anhui 243000, P.R. China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Yuan Zhang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Cheng Huang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| |
Collapse
|
20
|
Peng D, Huang Z, Yang H, Luo Y, Wu Z. PPM1G regulates hepatic ischemia/reperfusion injury through STING-mediated inflammatory pathways in macrophages. Immun Inflamm Dis 2024; 12:e1189. [PMID: 38372470 PMCID: PMC10875902 DOI: 10.1002/iid3.1189] [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: 03/30/2023] [Revised: 01/08/2024] [Accepted: 02/04/2024] [Indexed: 02/20/2024] Open
Abstract
BACKGROUND Ischemia/reperfusion injury (IRI) is generally unavoidable following liver transplantation. Here, we investigated the role of protein phosphatase, Mg2+ /Mn2+ dependent 1G (PPM1G) in hepatic IRI. METHODS Hepatic IRI was mimicked by employing a hypoxia/reperfusion (H/R) model in RAW 264.7 cells and a 70% warm ischemia model in C57BL/6 mice, respectively. In vitro, expression changes of tumor necrosis factor-α and interleukin were detected by quantitative real-time polymerase chain reaction (qRT-PCR), western blot analysis, and enzyme-linked immunosorbent assay. The protein expressions of PPM1G and the stimulator of interferon genes (STING) pathway components were analyzed by western blot. Interaction between PPM1G and STING was verified by coimmunoprecipitation (CO-IP). Immunofluorescence was applied for detection of p-IRF3. Flow cytometry, qRT-PCR and western blot were utilized to analyze markers of macrophage polarization. In vivo, histological analyses of mice liver were carried out by TUNEL and H&E staining. Changes in serum aminotransferases were also detected. RESULTS Following H/R intervention, a steady decline in PPM1G along with an increase in inflammatory cytokines in vitro was observed. Addition of plasmid with PPM1G sequence limited the release of inflammatory cytokines and downregulated phosphorylation of STING. CO-IP validated the interaction between PPM1G and STING. Furthermore, inhibition of PPM1G with lentivirus enhanced phosphorylation of STING and its downstream components; meanwhile, p65, p38, and Jnk were also surged to phosphorylation. Expression of INOS and CD86 was surged, while CD206, Arg-1, and IL-10 were inhibited. In vivo, PPM1G inhibition further promoted liver damage, hepatocyte apoptosis, and transaminases release. Selective inhibition of STING with C-176 partially reversed the activation of STING pathway and inflammatory cytokines in vitro. M1 markers were also suppressed by C-176. In vivo, C-176 rescued liver damage and transaminase release caused by PPM1G inhibition. CONCLUSION PPM1G suppresses hepatic IRI and macrophage M1 phenotype by repressing STING-mediated inflammatory pathways.
Collapse
Affiliation(s)
- Dadi Peng
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Zuotian Huang
- Department of Hepatobiliary Pancreatic Tumor CenterChongqing University Cancer HospitalChongqingChina
| | - Hang Yang
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Yunhai Luo
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Zhongjun Wu
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| |
Collapse
|
21
|
Tian J, Fu W, Xie Z, Zhao Y, Yang H, Zhao J. Methionine enkephalin (MENK) protected macrophages from ferroptosis by downregulating HMOX1 and ferritin. Proteome Sci 2024; 22:2. [PMID: 38245706 PMCID: PMC10799539 DOI: 10.1186/s12953-024-00228-x] [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: 05/02/2023] [Accepted: 01/06/2024] [Indexed: 01/22/2024] Open
Abstract
OBJECTIVE The aim of this work was to investigate the immunological effect of MENK by analyzing the protein spectrum and bioinformatics of macrophage RAW264.7, and to explore the relationship between macrophage and ferroptosis. RESULT We employed proteomic analysis to identify differentially expressed proteins (DEPs) between macrophages and macrophages intervened by MENK. A total of 208 DEPs were identified. Among these, 96 proteins had upregulated expression and 112 proteins had downregulated expression. Proteomic analysis revealed a significant enrichment of DEPs associated with iron metabolism. The identification of hub genes was conducted using KEGG pathway diagrams and protein-protein interaction (PPI) analysis. The hub genes identified in this study include HMOX1 and Ferritin (FTH and FTL). A correlation was established between HMOX1, FTH, and FTL in the GO and KEGG databases. The results of PCR, WB and immunofluorescence showed that MENK downregulated the level of HMOX1 and FTH. CONCLUSION MENK had the potential to become an adjuvant chemotherapy drug by regulating iron metabolism in macrophages, reducing levels of HMOX1 and ferritin. We proposed an innovative research direction on the therapeutic potential of MENK, focusing on the relationship between ferroptosis and macrophage activity.
Collapse
Affiliation(s)
- Jing Tian
- Department of Immunology, School of Basic Medical Science, Jinzhou Medical University, Jinzhou, Liaoning, 121000, China.
| | - Wenrui Fu
- Graduate College, Jinzhou Medical University, Jinzhou, Liaoning, 121000, China
| | - Zifeng Xie
- First Clinical Medical College, Jinzhou Medical University, Jinzhou, Liaoning, 121000, China
| | - Yuanlong Zhao
- First Clinical Medical College, Jinzhou Medical University, Jinzhou, Liaoning, 121000, China
| | - Haochen Yang
- First Clinical Medical College, Jinzhou Medical University, Jinzhou, Liaoning, 121000, China
| | - Jiafan Zhao
- First Clinical Medical College, Jinzhou Medical University, Jinzhou, Liaoning, 121000, China
| |
Collapse
|
22
|
Chullo G, Panisello-Rosello A, Marquez N, Colmenero J, Brunet M, Pera M, Rosello-Catafau J, Bataller R, García-Valdecasas JC, Fundora Y. Focusing on Ischemic Reperfusion Injury in the New Era of Dynamic Machine Perfusion in Liver Transplantation. Int J Mol Sci 2024; 25:1117. [PMID: 38256190 PMCID: PMC10816079 DOI: 10.3390/ijms25021117] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/04/2024] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
Liver transplantation is the most effective treatment for end-stage liver disease. Transplant indications have been progressively increasing, with a huge discrepancy between the supply and demand of optimal organs. In this context, the use of extended criteria donor grafts has gained importance, even though these grafts are more susceptible to ischemic reperfusion injury (IRI). Hepatic IRI is an inherent and inevitable consequence of all liver transplants; it involves ischemia-mediated cellular damage exacerbated upon reperfusion and its severity directly affects graft function and post-transplant complications. Strategies for organ preservation have been constantly improving since they first emerged. The current gold standard for preservation is perfusion solutions and static cold storage. However, novel approaches that allow extended preservation times, organ evaluation, and their treatment, which could increase the number of viable organs for transplantation, are currently under investigation. This review discusses the mechanisms associated with IRI, describes existing strategies for liver preservation, and emphasizes novel developments and challenges for effective organ preservation and optimization.
Collapse
Affiliation(s)
- Gabriela Chullo
- Service of Digestive, Hepato-Pancreatico-Biliary and Liver Transplant Surgery, Institut Clínic de Malalties Digestives i Metabòliques (ICMDM), Hospital Clinic of Barcelona, 08036 Barcelona, Spain; (G.C.); (M.P.); (J.C.G.-V.)
- Institut d’Investigacions Biomediques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain; (J.C.); (M.B.); (R.B.)
| | - Arnau Panisello-Rosello
- Institut d’Investigacions Biomediques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain; (J.C.); (M.B.); (R.B.)
| | - Noel Marquez
- Hepato-Pancreatico-Biliary and Liver Transplant Surgery, Institut Clínic de Malalties Digestives i Metabòliques (ICMDM), Hospital Clinic of Barcelona, 08036 Barcelona, Spain;
| | - Jordi Colmenero
- Institut d’Investigacions Biomediques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain; (J.C.); (M.B.); (R.B.)
- Liver Transplant Unit, Service of Hepatology, Institut Clínic de Malalties Digestives i Metabòliques (ICMDM), Hospital Clinic of Barcelona, 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades hepaticas y digestives (CIBERehd), University of Barcelona, 08036 Barcelona, Spain
| | - Merce Brunet
- Institut d’Investigacions Biomediques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain; (J.C.); (M.B.); (R.B.)
- Centro de Investigación Biomédica en Red de Enfermedades hepaticas y digestives (CIBERehd), University of Barcelona, 08036 Barcelona, Spain
- Pharmacology and Toxicology Laboratory, Biochemistry and Molecular Genetics Department, Biomedical Diagnostic Center, Hospital Clinic of Barcelona, 08036 Barcelona, Spain
| | - Miguel Pera
- Service of Digestive, Hepato-Pancreatico-Biliary and Liver Transplant Surgery, Institut Clínic de Malalties Digestives i Metabòliques (ICMDM), Hospital Clinic of Barcelona, 08036 Barcelona, Spain; (G.C.); (M.P.); (J.C.G.-V.)
- Institut d’Investigacions Biomediques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain; (J.C.); (M.B.); (R.B.)
| | - Joan Rosello-Catafau
- Experimental Pathology, Institut d’Investigacions Biomèdiques de Barcelona-Consejo Superior de Investigaciones Científicas (IBB-CSIC), 08036 Barcelona, Spain;
| | - Ramon Bataller
- Institut d’Investigacions Biomediques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain; (J.C.); (M.B.); (R.B.)
- Liver Transplant Unit, Service of Hepatology, Institut Clínic de Malalties Digestives i Metabòliques (ICMDM), Hospital Clinic of Barcelona, 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades hepaticas y digestives (CIBERehd), University of Barcelona, 08036 Barcelona, Spain
| | - Juan Carlos García-Valdecasas
- Service of Digestive, Hepato-Pancreatico-Biliary and Liver Transplant Surgery, Institut Clínic de Malalties Digestives i Metabòliques (ICMDM), Hospital Clinic of Barcelona, 08036 Barcelona, Spain; (G.C.); (M.P.); (J.C.G.-V.)
- Institut d’Investigacions Biomediques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain; (J.C.); (M.B.); (R.B.)
| | - Yiliam Fundora
- Service of Digestive, Hepato-Pancreatico-Biliary and Liver Transplant Surgery, Institut Clínic de Malalties Digestives i Metabòliques (ICMDM), Hospital Clinic of Barcelona, 08036 Barcelona, Spain; (G.C.); (M.P.); (J.C.G.-V.)
- Institut d’Investigacions Biomediques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain; (J.C.); (M.B.); (R.B.)
| |
Collapse
|
23
|
Wang Y, Liu Z, Song S, Wang J, Jin C, Jia L, Ma Y, Yuan T, Cai Z, Xiang M. IRF5 governs macrophage adventitial infiltration to fuel abdominal aortic aneurysm formation. JCI Insight 2024; 9:e171488. [PMID: 38175709 PMCID: PMC11143966 DOI: 10.1172/jci.insight.171488] [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: 04/14/2023] [Accepted: 12/26/2023] [Indexed: 01/05/2024] Open
Abstract
Abdominal aortic aneurysm (AAA) is a chronic inflammatory disease characterized by the expansion of the aortic wall. One of the most significant features is the infiltration of macrophages in the adventitia, which drives vasculature remodeling. The role of macrophage-derived interferon regulatory factor 5 (IRF5) in macrophage infiltration and AAA formation remains unknown. RNA sequencing of AAA adventitia identified Irf5 as the top significantly increased transcription factor that is predominantly expressed in macrophages. Global and myeloid cell-specific deficiency of Irf5 reduced AAA progression, with a marked reduction in macrophage infiltration. Further cellular investigations indicated that IRF5 promotes macrophage migration by direct regulation of downstream phosphoinositide 3-kinase γ (PI3Kγ, Pik3cg). Pik3cg ablation hindered AAA progression, and myeloid cell-specific salvage of Pik3cg restored AAA progression and macrophage infiltration derived from Irf5 deficiency. Finally, we found that IRF5 and PI3Kγ expression in the adventitia is significantly increased in patients with AAA. These findings reveal that the IRF5-dependent regulation of PI3Kγ is essential for AAA formation.
Collapse
Affiliation(s)
- Yidong Wang
- Department of Cardiology, State Key Laboratory of Transvascular Implantation Devices, Provincial Key Laboratory of Cardiovascular Research, and
| | - Zhenjie Liu
- Department of Vascular Surgery, The second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Shen Song
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianfang Wang
- Department of Cardiology, State Key Laboratory of Transvascular Implantation Devices, Provincial Key Laboratory of Cardiovascular Research, and
| | - Chunna Jin
- Department of Cardiology, State Key Laboratory of Transvascular Implantation Devices, Provincial Key Laboratory of Cardiovascular Research, and
| | - Liangliang Jia
- Department of Cardiology, State Key Laboratory of Transvascular Implantation Devices, Provincial Key Laboratory of Cardiovascular Research, and
| | - Yuankun Ma
- Department of Cardiology, State Key Laboratory of Transvascular Implantation Devices, Provincial Key Laboratory of Cardiovascular Research, and
| | - Tan Yuan
- Department of Cardiology, State Key Laboratory of Transvascular Implantation Devices, Provincial Key Laboratory of Cardiovascular Research, and
| | - Zhejun Cai
- Department of Cardiology, State Key Laboratory of Transvascular Implantation Devices, Provincial Key Laboratory of Cardiovascular Research, and
| | - Meixiang Xiang
- Department of Cardiology, State Key Laboratory of Transvascular Implantation Devices, Provincial Key Laboratory of Cardiovascular Research, and
| |
Collapse
|
24
|
Li Y, Ma JQ, Wang CC, Zhou J, Sun YD, Wei XL, Zhao ZQ. Ferroptosis: A potential target of macrophages in plaque vulnerability. Open Life Sci 2023; 18:20220722. [PMID: 37791060 PMCID: PMC10543703 DOI: 10.1515/biol-2022-0722] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 08/04/2023] [Accepted: 08/16/2023] [Indexed: 10/05/2023] Open
Abstract
Plaque vulnerability has been the subject of several recent studies aimed at reducing the risk of stroke and carotid artery stenosis. Atherosclerotic plaque development is a complex process involving inflammation mediated by macrophages. Plaques become more vulnerable when the equilibrium between macrophage recruitment and clearance is disturbed. Lipoperoxides, which are affected by iron levels in cells, are responsible for the cell death seen in ferroptosis. Ferroptosis results from lipoperoxide-induced mitochondrial membrane toxicity. Atherosclerosis in ApoE(-/-) mice is reduced when ferroptosis is inhibited and iron intake is limited. Single-cell sequencing revealed that a ferroptosis-related gene was substantially expressed in atherosclerosis-modeled macrophages. Since ferroptosis can be regulated, it offers hope as a non-invasive method of treating carotid plaque. In this study, we discuss the role of ferroptosis in atherosclerotic plaque vulnerability, including its mechanism, regulation, and potential future research directions.
Collapse
Affiliation(s)
- Yu Li
- Department of Vascular Surgery, Changhai Hospital, The PLA Naval Medical University, 168 Changhai Road, Shanghai200433, China
| | - Ji-Qing Ma
- Department of Vascular Surgery, Changhai Hospital, The PLA Naval Medical University, 168 Changhai Road, Shanghai200433, China
| | - Chao-Chen Wang
- Department of Vascular Surgery, Changhai Hospital, The PLA Naval Medical University, 168 Changhai Road, Shanghai200433, China
| | - Jian Zhou
- Department of Vascular Surgery, Changhai Hospital, The PLA Naval Medical University, 168 Changhai Road, Shanghai200433, China
| | - Yu-Dong Sun
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University,
Nanjing201411, China
| | - Xiao-Long Wei
- Department of Vascular Surgery, Changhai Hospital, The PLA Naval Medical University, 168 Changhai Road, Shanghai200433, China
| | - Zhi-Qing Zhao
- Department of Vascular Surgery, Changhai Hospital, The PLA Naval Medical University, 168 Changhai Road, Shanghai200433, China
| |
Collapse
|
25
|
Cao Q, Wang R, Niu Z, Chen T, Azmi F, Read SA, Chen J, Lee VW, Zhou C, Julovi S, Huang Q, Wang YM, Starkey MR, Zheng G, Alexander SI, George J, Wang Y, Harris DC. Type 2 innate lymphoid cells are protective against hepatic ischaemia/reperfusion injury. JHEP Rep 2023; 5:100837. [PMID: 37691688 PMCID: PMC10482753 DOI: 10.1016/j.jhepr.2023.100837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 06/16/2023] [Accepted: 06/18/2023] [Indexed: 09/12/2023] Open
Abstract
BACKGROUND AND AIMS Although type 2 innate lymphoid cells (ILC2s) were originally found to be liver-resident lymphocytes, the role and importance of ILC2 in liver injury remains poorly understood. In the current study, we sought to determine whether ILC2 is an important regulator of hepatic ischaemia/reperfusion injury (IRI). METHODS ILC2-deficient mice (ICOS-T or NSG) and genetically modified ILC2s were used to investigate the role of ILC2s in murine hepatic IRI. Interactions between ILC2s and eosinophils or macrophages were studied in coculture. The role of human ILC2s was assessed in an immunocompromised mouse model of hepatic IRI. RESULTS Administration of IL-33 prevented hepatic IRI in association with reduction of neutrophil infiltration and inflammatory mediators in the liver. IL-33-treated mice had elevated numbers of ILC2s, eosinophils, and regulatory T cells. Eosinophils, but not regulatory T cells, were required for IL-33-mediated hepatoprotection in IRI mice. Depletion of ILC2s substantially abolished the protective effect of IL-33 in hepatic IRI, indicating that ILC2s play critical roles in IL-33-mediated liver protection. Adoptive transfer of ex vivo-expanded ILC2s improved liver function and attenuated histologic damage in mice subjected to IRI. Mechanistic studies combining genetic and adoptive transfer approaches identified a protective role of ILC2s through promoting IL-13-dependent induction of anti-inflammatory macrophages and IL-5-dependent elevation of eosinophils in IRI. Furthermore, in vivo expansion of human ILC2s by IL-33 or transfer of ex vivo-expanded human ILC2s ameliorated hepatic IRI in an immunocompromised mouse model of hepatic IRI. CONCLUSIONS This study provides insight into the mechanisms of ILC2-mediated liver protection that could serve as therapeutic targets to treat acute liver injury. IMPACT AND IMPLICATIONS We report that type 2 innate lymphoid cells (ILC2s) are important regulators in a mouse model of liver ischaemia/reperfusion injury (IRI). Through manipulation of macrophage and eosinophil phenotypes, ILC2s mitigate liver inflammation and injury during liver IRI. We propose that ILC2s have the potential to serve as a therapeutic tool for protecting against acute liver injury and lay the foundation for translation of ILC2 therapy to human liver disease.
Collapse
Affiliation(s)
- Qi Cao
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia
| | - Ruifeng Wang
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia
- Department of Nephrology, The Second Hospital of Anhui Medical University, Hefei, China
| | - Zhiguo Niu
- Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
| | - Titi Chen
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia
| | - Farhana Azmi
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia
| | - Scott A. Read
- Storr Liver Centre, Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia
| | - Jianwei Chen
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia
| | - Vincent W.S. Lee
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia
| | - Chunze Zhou
- Department of Interventional Radiology, The First Affiliated Hospital of University of Science and Technology of China, Hefei, China
| | - Sohel Julovi
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia
| | - Qingsong Huang
- Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
| | - Yuan Min Wang
- Centre for Kidney Research, Children’s Hospital at Westmead, Sydney, NSW, Australia
| | - Malcolm R. Starkey
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Guoping Zheng
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia
| | - Stephen I. Alexander
- Centre for Kidney Research, Children’s Hospital at Westmead, Sydney, NSW, Australia
| | - Jacob George
- Storr Liver Centre, Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia
| | - Yiping Wang
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia
| | - David C.H. Harris
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
26
|
Liu H, Zhang L, Liu Z, Lin J, He X, Wu S, Qin Y, Zhao C, Guo Y, Lin F. Galectin-3 as TREM2 upstream factor contributes to lung ischemia-reperfusion injury by regulating macrophage polarization. iScience 2023; 26:107496. [PMID: 37636061 PMCID: PMC10448077 DOI: 10.1016/j.isci.2023.107496] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 05/31/2023] [Accepted: 07/25/2023] [Indexed: 08/29/2023] Open
Abstract
Lung ischemia-reperfusion injury (LIRI) is a complex "aseptic" inflammatory response, macrophage play a pivotal role in the pathogenesis of LIRI. Galectin-3 (Gal3), a lectin implicated inflammation, has received limited attention in LIRI. Studies have reported Gal3 as a ligand for triggering receptor expressed on myeloid cell 2 (TREM2) in macrophages in Alzheimer's disease. Hence, we established LIRI C57BL/6 mice model and hypoxia/glucose deprivation and reoxygenation (OGD/R) model to investigate the relationship among Gal3, TREM2, and macrophage polarization. Our result demonstrated inhibition of Gal3 significantly reduced M1-type macrophage polarization while markedly increased M2-type in LIRI. In addition, we observed colocalization of Gal3 and TREM2 in macrophages, inhibition of Gal3 could recover the downregulation of TREM2 induced by LIRI while promoting TREM2 expression could attenuate lung injury in LIRI. In summary, our findings suggest Gal3 as an upstream factor of TREM2, play a crucial role in LIRI by regulating macrophage polarization.
Collapse
Affiliation(s)
- Hao Liu
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, China
- Guangxi Clinical Research Center for Anesthesiology(GK AD22035214), Nanning, Guangxi 530021, China
- Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning, Guangxi 530021, China
- Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, Guangxi 530021, China
| | - Lu Zhang
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, China
- Guangxi Clinical Research Center for Anesthesiology(GK AD22035214), Nanning, Guangxi 530021, China
- Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning, Guangxi 530021, China
- Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, Guangxi 530021, China
| | - Zhen Liu
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, China
- Guangxi Clinical Research Center for Anesthesiology(GK AD22035214), Nanning, Guangxi 530021, China
- Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning, Guangxi 530021, China
- Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, Guangxi 530021, China
| | - Jinyuan Lin
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, China
- Guangxi Clinical Research Center for Anesthesiology(GK AD22035214), Nanning, Guangxi 530021, China
- Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning, Guangxi 530021, China
- Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, Guangxi 530021, China
| | - Xiaojing He
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, China
- Guangxi Clinical Research Center for Anesthesiology(GK AD22035214), Nanning, Guangxi 530021, China
- Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning, Guangxi 530021, China
- Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, Guangxi 530021, China
| | - Siyi Wu
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, China
- Guangxi Clinical Research Center for Anesthesiology(GK AD22035214), Nanning, Guangxi 530021, China
- Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning, Guangxi 530021, China
- Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, Guangxi 530021, China
| | - Yi Qin
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, China
- Guangxi Clinical Research Center for Anesthesiology(GK AD22035214), Nanning, Guangxi 530021, China
- Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning, Guangxi 530021, China
- Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, Guangxi 530021, China
| | - Chen Zhao
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, China
- Guangxi Clinical Research Center for Anesthesiology(GK AD22035214), Nanning, Guangxi 530021, China
- Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning, Guangxi 530021, China
- Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, Guangxi 530021, China
| | - Youyuan Guo
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, China
- Guangxi Clinical Research Center for Anesthesiology(GK AD22035214), Nanning, Guangxi 530021, China
- Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning, Guangxi 530021, China
- Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, Guangxi 530021, China
| | - Fei Lin
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, China
- Guangxi Clinical Research Center for Anesthesiology(GK AD22035214), Nanning, Guangxi 530021, China
- Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning, Guangxi 530021, China
- Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, Guangxi 530021, China
| |
Collapse
|
27
|
Menon N, Kishen A. Nociceptor-Macrophage Interactions in Apical Periodontitis: How Biomolecules Link Inflammation with Pain. Biomolecules 2023; 13:1193. [PMID: 37627258 PMCID: PMC10452348 DOI: 10.3390/biom13081193] [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: 06/20/2023] [Revised: 07/20/2023] [Accepted: 07/24/2023] [Indexed: 08/27/2023] Open
Abstract
Periradicular tissues have a rich supply of peripheral afferent neurons, also known as nociceptive neurons, originating from the trigeminal nerve. While their primary function is to relay pain signals to the brain, these are known to be involved in modulating innate and adaptive immunity by initiating neurogenic inflammation (NI). Studies have investigated neuroanatomy and measured the levels of biomolecules such as cytokines and neuropeptides in human saliva, gingival crevicular fluid, or blood/serum samples in apical periodontitis (AP) to validate the possible role of trigeminal nociceptors in inflammation and tissue regeneration. However, the contributions of nociceptors and the mechanisms involved in the neuro-immune interactions in AP are not fully understood. This narrative review addresses the complex biomolecular interactions of trigeminal nociceptors with macrophages, the effector cells of the innate immune system, in the clinical manifestations of AP.
Collapse
Affiliation(s)
| | - Anil Kishen
- Dental Research Institute, Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada;
| |
Collapse
|
28
|
Du S, Zhang X, Jia Y, Peng P, Kong Q, Jiang S, Li Y, Li C, Ding Z, Liu L. Hepatocyte HSPA12A inhibits macrophage chemotaxis and activation to attenuate liver ischemia/reperfusion injury via suppressing glycolysis-mediated HMGB1 lactylation and secretion of hepatocytes. Theranostics 2023; 13:3856-3871. [PMID: 37441587 PMCID: PMC10334822 DOI: 10.7150/thno.82607] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 05/26/2023] [Indexed: 07/15/2023] Open
Abstract
Rationale: Liver ischemia-reperfusion (LI/R) injury is characterized by two interconnected phases: local ischemia that causes hepatic cell damage to release damage-associated molecular pattern (DAMPs), and DAMPs that recruit immune cells to elicit inflammatory cascade for further injury of hepatocytes. High-mobility group box 1 (HMGB1) is a representative DAMP. Studies in macrophages demonstrated that HMGB1 is secreted after lactylation during sepsis. However, whether lactylation mediates HMGB1 secretion from hepatocytes after LI/R is known. Heat shock protein A12A (HSPA12A) is an atypical member of HSP70 family. Methods: Gene expression was examined by microarray analysis and immunoblotting. The hepatic injury was analyzed using released ALT and AST activities assays. Hepatic macrophage chemotaxis was evaluated by Transwell chemotaxis assays. Inflammatory mediators were evaluated by immunoblotting. HMGB1 secretion was examined in exosomes or serum. HMGB1 lactylation was determined using immunoprecipitation and immunoblotting. Results: Here, we report that LI/R decreased HSPA12A expression in hepatocytes, while hepatocyte-specific HSPA12A overexpression attenuated LI/R-induced hepatic dysfunction and mortality of mice. We also noticed that hepatocyte HSPA12A overexpression suppressed macrophage chemotaxis to LI/R-exposed livers in vivo and to hypoxia/reoxygenation (H/R)-exposed hepatocytes in vitro. The LI/R-increased serum HMGB1 levels of mice and the H/R-increased HMGB1 lactylation and secretion levels of hepatocytes were also inhibited by hepatocyte HSPA12A overexpression. By contrast, HSPA12A knockout in hepatocytes promoted not only H/R-induced HMGB1 lactylation and secretion of hepatocytes but also the effects of H/R-hepatocytes on macrophage chemotaxis and inflammatory activation, while all these deleterious effects of HSPA12A knockout were reversed following hepatocyte HMGB1 knockdown. Further molecular analyses showed that HSPA12A overexpression reduced glycolysis-generated lactate, thus decreasing HMGB1 lactylation and secretion from hepatocytes, thereby inhibiting not only macrophage chemotaxis but also the subsequent inflammatory cascade, which ultimately protecting against LI/R injury. Conclusion: Taken together, these findings suggest that hepatocyte HSPA12A is a novel regulator that protects livers from LI/R injury by suppressing glycolysis-mediated HMGB1 lactylation and secretion from hepatocytes to inhibit macrophage chemotaxis and inflammatory activation. Therefore, targeting hepatocyte HSPA12A may have therapeutic potential in the management of LI/R injury in patients.
Collapse
Affiliation(s)
- Shuya Du
- Department of Geriatrics, Jiangsu Provincial Key Laboratory of Geriatrics, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xiaojin Zhang
- Department of Geriatrics, Jiangsu Provincial Key Laboratory of Geriatrics, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Yunxiao Jia
- Department of Geriatrics, Jiangsu Provincial Key Laboratory of Geriatrics, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Peipei Peng
- Department of Anesthesiology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Qiuyue Kong
- Department of Anesthesiology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Surong Jiang
- Department of Geriatrics, Jiangsu Provincial Key Laboratory of Geriatrics, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Yuehua Li
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 210029, China
| | - Chuanfu Li
- Departments of Surgery, East Tennessee State University, Johnson City, TN 37614, USA
| | - Zhengnian Ding
- Department of Anesthesiology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Li Liu
- Department of Geriatrics, Jiangsu Provincial Key Laboratory of Geriatrics, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
29
|
Seika P, Janikova M, Asokan S, Janovicova L, Csizmadia E, O’Connell M, Robson SC, Glickman J, Wegiel B. Free heme exacerbates colonic injury induced by anti-cancer therapy. Front Immunol 2023; 14:1184105. [PMID: 37342339 PMCID: PMC10277564 DOI: 10.3389/fimmu.2023.1184105] [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: 03/11/2023] [Accepted: 05/22/2023] [Indexed: 06/22/2023] Open
Abstract
Gastrointestinal inflammation and bleeding are commonly induced by cancer radiotherapy and chemotherapy but mechanisms are unclear. We demonstrated an increased number of infiltrating heme oxygenase-1 positive (HO-1+) macrophages (Mø, CD68+) and the levels of hemopexin (Hx) in human colonic biopsies from patients treated with radiation or chemoradiation versus non-irradiated controls or in the ischemic intestine compared to matched normal tissues. The presence of rectal bleeding in these patients was also correlated with higher HO-1+ cell infiltration. To functionally assess the role of free heme released in the gut, we employed myeloid-specific HO-1 knockout (LysM-Cre : Hmox1flfl), hemopexin knockout (Hx-/-) and control mice. Using LysM-Cre : Hmox1flfl conditional knockout (KO) mice, we showed that a deficiency of HO-1 in myeloid cells led to high levels of DNA damage and proliferation in colonic epithelial cells in response to phenylhydrazine (PHZ)-induced hemolysis. We found higher levels of free heme in plasma, epithelial DNA damage, inflammation, and low epithelial cell proliferation in Hx-/- mice after PHZ treatment compared to wild-type mice. Colonic damage was partially attenuated by recombinant Hx administration. Deficiency in Hx or Hmox1 did not alter the response to doxorubicin. Interestingly, the lack of Hx augmented abdominal radiation-mediated hemolysis and DNA damage in the colon. Mechanistically, we found an altered growth of human colonic epithelial cells (HCoEpiC) treated with heme, corresponding to an increase in Hmox1 mRNA levels and heme:G-quadruplex complexes-regulated genes such as c-MYC, CCNF, and HDAC6. Heme-treated HCoEpiC cells exhibited growth advantage in the absence or presence of doxorubicin, in contrast to poor survival of heme-stimulated RAW247.6 Mø. In summary, our data indicate that accumulation of heme in the colon following hemolysis and/or exposure to genotoxic stress amplifies DNA damage, abnormal proliferation of epithelial cells, and inflammation as a potential etiology for gastrointestinal syndrome (GIS).
Collapse
Affiliation(s)
- Philippa Seika
- Department of Surgery, Division of Surgical Sciences, Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Monika Janikova
- Department of Surgery, Division of Surgical Sciences, Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Sahana Asokan
- Department of Surgery, Division of Surgical Sciences, Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
- Division of Microbiome and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Lubica Janovicova
- Department of Surgery, Division of Surgical Sciences, Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Eva Csizmadia
- Department of Surgery, Division of Surgical Sciences, Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Mckenzie O’Connell
- Department of Surgery, Division of Surgical Sciences, Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Simon C. Robson
- Department of Anesthesia, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Jonathan Glickman
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Barbara Wegiel
- Department of Surgery, Division of Surgical Sciences, Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
30
|
Liu H, Zhu Y, Ng KTP, Lo CM, Man K. The Landscape of Aberrant Alternative Splicing Events in Steatotic Liver Graft Post Transplantation via Transcriptome-Wide Analysis. Int J Mol Sci 2023; 24:ijms24098216. [PMID: 37175922 PMCID: PMC10179559 DOI: 10.3390/ijms24098216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/28/2023] [Accepted: 04/03/2023] [Indexed: 05/15/2023] Open
Abstract
The application of steatotic liver graft has been increased significantly due to the severe donor shortage and prevalence of non-alcoholic fatty liver disease. However, steatotic donor livers are vulnerable to acute phase inflammatory injury, which may result in cancer recurrence. Alternative splicing events (ASEs) are critical for diverse transcriptional variants in hepatocellular carcinoma (HCC). Here, we aimed to depict the landscape of ASEs, as well as to identify the differential ASEs in steatotic liver graft and their association with tumor recurrence after transplantation. The overall portrait of intragraft transcripts and ASEs were elucidated through RNA sequencing with the liver graft biopsies from patients and rat transplant models. Various differential ASEs were identified in steatotic liver grafts. CYP2E1, ADH1A, CYP2C8, ADH1C, and HGD, as corresponding genes to the common pathways involved differential ASEs in human and rats, were significantly associated with HCC patients' survival. The differential ASEs related RNA-binding proteins (RBPs) were enriched in metabolic pathways. The altered immune cell distribution, particularly macrophages and neutrophils, were perturbated by differential ASEs. The cancer hallmarks were enriched in steatotic liver grafts and closely associated with differential ASEs. Our work identified the differential ASE network with metabolic RBPs, immune cell distribution, and cancer hallmarks in steatotic liver grafts. We verified the link between steatotic liver graft injury and tumor recurrence at post-transcriptional level, offered new evidence to explore metabolism and immune responses, and provided the potential prognostic and therapeutic markers for tumor recurrence.
Collapse
Affiliation(s)
- Hui Liu
- Department of Surgery, School of Clinical Medicine, HKU-SZH & LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yueqin Zhu
- Department of Surgery, School of Clinical Medicine, HKU-SZH & LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Kevin Tak-Pan Ng
- Department of Surgery, School of Clinical Medicine, HKU-SZH & LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Chung-Mau Lo
- Department of Surgery, School of Clinical Medicine, HKU-SZH & LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Kwan Man
- Department of Surgery, School of Clinical Medicine, HKU-SZH & LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
31
|
Ito Y, Hosono K, Amano H. Responses of hepatic sinusoidal cells to liver ischemia–reperfusion injury. Front Cell Dev Biol 2023; 11:1171317. [PMID: 37082623 PMCID: PMC10112669 DOI: 10.3389/fcell.2023.1171317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 03/27/2023] [Indexed: 04/07/2023] Open
Abstract
The liver displays a remarkable regenerative capacity in response to acute liver injury. In addition to the proliferation of hepatocytes during liver regeneration, non-parenchymal cells, including liver macrophages, liver sinusoidal endothelial cells (LSECs), and hepatic stellate cells (HSCs) play critical roles in liver repair and regeneration. Liver ischemia–reperfusion injury (IRI) is a major cause of increased liver damage during liver resection, transplantation, and trauma. Impaired liver repair increases postoperative morbidity and mortality of patients who underwent liver surgery. Successful liver repair and regeneration after liver IRI requires coordinated interplay and synergic actions between hepatic resident cells and recruited cell components. However, the underlying mechanisms of liver repair after liver IRI are not well understood. Recent technological advances have revealed the heterogeneity of each liver cell component in the steady state and diseased livers. In this review, we describe the progress in the biology of liver non-parenchymal cells obtained from novel technological advances. We address the functional role of each cell component in response to liver IRI and the interactions between diverse immune repertoires and non-hematopoietic cell populations during the course of liver repair after liver IRI. We also discuss how these findings can help in the design of novel therapeutic approaches. Growing insights into the cellular interactions during liver IRI would enhance the pathology of liver IRI understanding comprehensively and further develop the strategies for improvement of liver repair.
Collapse
|
32
|
Zhang T, Fang Q, Liu P, Wang P, Feng C, Wang J. Heme oxygenase 1 overexpression induces immune evasion of acute myeloid leukemia against natural killer cells by inhibiting CD48. J Transl Med 2022; 20:394. [PMID: 36058936 PMCID: PMC9441067 DOI: 10.1186/s12967-022-03589-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 08/13/2022] [Indexed: 11/29/2022] Open
Abstract
Background Acute myeloid leukemia (AML) is the most common type of acute leukemia in adults. Given the high relapse rate, more effective treatments are needed to improve clinical outcomes. We previously demonstrated that heme oxygenase 1 (HO1) is overexpressed in AML, while the functional roles of HO1 remain unclear. Methods Bioinformatics analysis and flow cytometry were conducted to assess the association between HO1 levels and immune cells or immune checkpoint/ligand molecules in AML patients. Primary natural killer (NK) cells were purified and subsequently co-cultured in vitro with transduced AML cells to determine the effects of HO1 expression on NK cell functions. AML mice models were established to investigate the effects of HO1 expression on cytotoxic effects of NK cells in vivo. The molecular mechanism was studied by flow cytometry, quantitative real-time PCR (qRT-PCR), western blotting, and immunoprecipitation. Results Bioinformatics analysis indicated a correlation between HO1 expression and the AML immune microenvironment. The present study findings indicated that HO1 specifically downregulates the expression of CD48, a ligand of the NK cell-activating receptor 2B4, thus decreasing the cytotoxic effect of NK cells. HO1 overexpression promoted tumor growth and inhibited the cytotoxic effect of NK cells in the AML mice model. Mechanistic investigations found that HO1 directly interacted with Sirt1 and increased its expression and deacetylase activity. With the overexpression of HO1, increased Sirt1 in AML cells enabled histone H3K27 deacetylation to suppress CD48 transcription and expression. Administration of Sirt1 inhibitor restored the expression of CD48. Conclusions Collectively, HO1 promotes NK cell dysfunction in AML. Therefore, restoring NK cell function by inhibiting HO1 activity is a potential immunotherapeutic approach against AML. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-022-03589-z.
Collapse
Affiliation(s)
- Tianzhuo Zhang
- Department of Clinical Medical School, Guizhou Medical University, Guiyang, 550004, China.,Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China.,Department of Guizhou Province Hematopoietic Stem Cell Transplantation Center and Key Laboratory of Hematological Disease Diagnostic and Treatment Centre, Guiyang, 550004, China
| | - Qin Fang
- Department of Pharmacy, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Ping Liu
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China.,Department of Guizhou Province Hematopoietic Stem Cell Transplantation Center and Key Laboratory of Hematological Disease Diagnostic and Treatment Centre, Guiyang, 550004, China
| | - Ping Wang
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China.,Department of Guizhou Province Hematopoietic Stem Cell Transplantation Center and Key Laboratory of Hematological Disease Diagnostic and Treatment Centre, Guiyang, 550004, China
| | - Cheng Feng
- Department of Clinical Medical School, Guizhou Medical University, Guiyang, 550004, China.,Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China.,Department of Guizhou Province Hematopoietic Stem Cell Transplantation Center and Key Laboratory of Hematological Disease Diagnostic and Treatment Centre, Guiyang, 550004, China
| | - Jishi Wang
- Department of Clinical Medical School, Guizhou Medical University, Guiyang, 550004, China. .,Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China. .,Department of Guizhou Province Hematopoietic Stem Cell Transplantation Center and Key Laboratory of Hematological Disease Diagnostic and Treatment Centre, Guiyang, 550004, China.
| |
Collapse
|
33
|
Frick K, Beller EA, Kalisvaart M, Dutkowski P, Schüpbach RA, Klinzing S. Procalcitonin in early allograft dysfunction after orthotopic liver transplantation: a retrospective single centre study. BMC Gastroenterol 2022; 22:404. [PMID: 36045337 PMCID: PMC9429388 DOI: 10.1186/s12876-022-02486-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 08/16/2022] [Indexed: 11/11/2022] Open
Abstract
Background Ischemia–reperfusion injury (IRI) is the pathophysiological hallmark of hepatic dysfunction after orthotopic liver transplantation (OLT). Related to IRI, early allograft dysfunction (EAD) after OLT affects short- and long-term outcome. During inflammatory states, the liver seems to be the main source of procalcitonin (PCT), which has been shown to increase independently of bacterial infection. This study investigates the association of PCT, IRI and EAD as well as the predictive value of PCT during the first postoperative week in terms of short- and long-term outcome after OLT. Methods Patients ≥ 18 years undergoing OLT between January 2016 and April 2020 at the University Hospital of Zurich were eligible for this retrospective study. Patients with incomplete PCT data on postoperative days (POD) 1 + 2 or combined liver-kidney transplantation were excluded. The PCT course during the first postoperative week, its association with EAD, defined by the criteria of Olthoff, and IRI, defined as aminotransferase level > 2000 IU/L within 2 PODs, were analysed. Finally, 90-day as well as 12-month graft and patient survival were assessed. Results Of 234 patients undergoing OLT, 110 patients were included. Overall, EAD and IRI patients had significantly higher median PCT values on POD 2 [31.3 (9.7–53.8) mcg/l vs. 11.1 (5.3–25.0) mcg/l; p < 0.001 and 27.7 (9.7–51.9) mcg/l vs. 11.5 (5.5–25.2) mcg/l; p < 0.001] and impaired 90-day graft survival (79.2% vs. 95.2%; p = 0.01 and 80.4% vs. 93.8%; p = 0.033). IRI patients with PCT < 15 mcg/l on POD 2 had reduced 90-day graft and patient survival (57.9% vs. 93.8%; p = 0.001 and 68.4% vs. 93.8%; p = 0.008) as well as impaired 12-month graft and patient survival (57.9% vs. 96.3%; p = 0.001 and 68.4% vs. 96.3%; p = 0.008), while the outcome of IRI patients with PCT > 15 mcg/l on POD 2 was comparable to that of patients without IRI/EAD. Conclusion Generally, PCT is increased in the early postoperative phase after OLT. Patients with EAD and IRI have a significantly increased PCT maximum on POD 2, and impaired 90-day graft survival. PCT measurement may have potential as an additional outcome predictor in the early phase after OLT, as in our subanalysis of IRI patients, PCT values < 15 mcg/l were associated with impaired outcome. Supplementary Information The online version contains supplementary material available at 10.1186/s12876-022-02486-5.
Collapse
|
34
|
Canesin G, Feldbrügge L, Wei G, Janovicova L, Janikova M, Csizmadia E, Ariffin J, Hedblom A, Herbert ZT, Robson SC, Celec P, Swanson KD, Nasser I, Popov YV, Wegiel B. Heme oxygenase-1 mitigates liver injury and fibrosis via modulation of LNX1/Notch1 pathway in myeloid cells. iScience 2022; 25:104983. [PMID: 36093061 PMCID: PMC9450142 DOI: 10.1016/j.isci.2022.104983] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 07/01/2022] [Accepted: 08/16/2022] [Indexed: 01/12/2023] Open
Abstract
Activation of resident macrophages (Mϕ) and hepatic stellate cells is a key event in chronic liver injury. Mice with heme oxygenase-1 (HO-1; Hmox1)-deficient Mϕ (LysM-Cre:Hmox1 flfl ) exhibit increased inflammation, periportal ductular reaction, and liver fibrosis following bile duct ligation (BDL)-induced liver injury and increased pericellular fibrosis in NASH model. RiboTag-based RNA-sequencing profiling of hepatic HO-1-deficient Mϕ revealed dysregulation of multiple genes involved in lipid and amino acid metabolism, regulation of oxidative stress, and extracellular matrix turnover. Among these genes, ligand of numb-protein X1 (LNX1) expression is strongly suppressed in HO-1-deficient Mϕ. Importantly, HO-1 and LNX1 were expressed by hepatic Mϕ in human biliary and nonbiliary end-stage cirrhosis. We found that Notch1 expression, a downstream target of LNX1, was increased in LysM-Cre:Hmox1 flfl mice. In HO-1-deficient Mϕ treated with heme, transient overexpression of LNX1 drives M2-like Mϕ polarization. In summary, we identified LNX1/Notch1 pathway as a downstream target of HO-1 in liver fibrosis.
Collapse
Affiliation(s)
- Giacomo Canesin
- Department of Surgery, Division of Surgical Sciences, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Linda Feldbrügge
- Charité – Universitätsmedizin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Surgery, Campus Charité Mitte and Campus Virchow-Klinikum, 13353 Berlin, Germany,Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Guangyan Wei
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA,Department of Radiation Oncology, First Affiliated Hospital, Sun Yat-sen University, 510080 Guangzhou, China
| | - Lubica Janovicova
- Department of Surgery, Division of Surgical Sciences, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA,Institute of Molecular Biomedicine, Comenius University in Bratislava, 811 08 Bratislava, Slovakia
| | - Monika Janikova
- Department of Surgery, Division of Surgical Sciences, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA,Institute of Molecular Biomedicine, Comenius University in Bratislava, 811 08 Bratislava, Slovakia
| | - Eva Csizmadia
- Department of Surgery, Division of Surgical Sciences, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Juliana Ariffin
- Department of Surgery, Division of Surgical Sciences, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Andreas Hedblom
- Department of Surgery, Division of Surgical Sciences, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Zachary T. Herbert
- Molecular Biology Core Facilities, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Simon C. Robson
- Department of Anesthesia, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Peter Celec
- Institute of Molecular Biomedicine, Comenius University in Bratislava, 811 08 Bratislava, Slovakia
| | - Kenneth D. Swanson
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Imad Nasser
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Yury V. Popov
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA,Corresponding author
| | - Barbara Wegiel
- Department of Surgery, Division of Surgical Sciences, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA,Corresponding author
| |
Collapse
|
35
|
Kervella D, Le Bas-Bernardet S, Bruneau S, Blancho G. Protection of transplants against antibody-mediated injuries: from xenotransplantation to allogeneic transplantation, mechanisms and therapeutic insights. Front Immunol 2022; 13:932242. [PMID: 35990687 PMCID: PMC9389360 DOI: 10.3389/fimmu.2022.932242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 07/07/2022] [Indexed: 11/17/2022] Open
Abstract
Long-term allograft survival in allotransplantation, especially in kidney and heart transplantation, is mainly limited by the occurrence of antibody-mediated rejection due to anti-Human Leukocyte Antigen antibodies. These types of rejection are difficult to handle and chronic endothelial damages are often irreversible. In the settings of ABO-incompatible transplantation and xenotransplantation, the presence of antibodies targeting graft antigens is not always associated with rejection. This resistance to antibodies toxicity seems to associate changes in endothelial cells phenotype and modification of the immune response. We describe here these mechanisms with a special focus on endothelial cells resistance to antibodies. Endothelial protection against anti-HLA antibodies has been described in vitro and in animal models, but do not seem to be a common feature in immunized allograft recipients. Complement regulation and anti-apoptotic molecules expression appear to be common features in all these settings. Lastly, pharmacological interventions that may promote endothelial cell protection against donor specific antibodies will be described.
Collapse
Affiliation(s)
- Delphine Kervella
- CHU Nantes, Nantes Université, Néphrologie et Immunologie Clinique, Institut Transplantation Urologie Néphrologie (ITUN), Nantes, France
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, ITUN, Nantes, France
| | - Stéphanie Le Bas-Bernardet
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, ITUN, Nantes, France
| | - Sarah Bruneau
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, ITUN, Nantes, France
| | - Gilles Blancho
- CHU Nantes, Nantes Université, Néphrologie et Immunologie Clinique, Institut Transplantation Urologie Néphrologie (ITUN), Nantes, France
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, ITUN, Nantes, France
- *Correspondence: Gilles Blancho,
| |
Collapse
|
36
|
Hang Z, Wei J, Zheng M, Gui Z, Chen H, Sun L, Fei S, Han Z, Tao J, Wang Z, Tan R, Gu M. Iguratimod Attenuates Macrophage Polarization and Antibody-Mediated Rejection After Renal Transplant by Regulating KLF4. Front Pharmacol 2022; 13:865363. [PMID: 35614941 PMCID: PMC9125033 DOI: 10.3389/fphar.2022.865363] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 04/11/2022] [Indexed: 11/13/2022] Open
Abstract
Background: This study aimed to explore the effect and mechanism of iguratimod (IGT) on M1 macrophage polarization and antibody-mediated rejection (ABMR) after renal transplant.Methods: Bioinformatics analysis was performed using three public databases derived from the GEO database. Sprague–Dawley (SD) rats were pre-sensitized with donors of Wistar rats in skin transplantation and a rat renal transplant ABMR model was established from the donors to skin pre-sensitized recipients. Subsequently, IGT was treated on the ABMR model. Routine staining and immunofluorescence (IF) staining were performed to observe the pathological changes in each group and flow cytometry was performed to detect the changes of DSA titers in peripheral blood. In addition, bone-marrow-derived macrophage (BMDM) was extracted and interfered with IGT to explore the effect of IGT in vivo. PCR, IF staining, and Western blot were used to detect the expression of related genes and proteins.Results: Bioinformatics analysis revealed that several immune cells were significantly infiltrated in the ABMR allograft, while M1 macrophage was noticed with the most significance. Results of IF staining and PCR proved the findings of the bioinformatics analysis. Based on this, IGT was observed to significantly attenuate the degree of peritubular capillary vasculitis and arteriolitis in the rat renal transplant ABMR model, whereas it decreases the expression of C4d and reduces the titer of DSA. Results in vitro suggested that M1 macrophage-related transcripts and proteins were significantly reduced by the treatment of IGT in a dose- and time-dependent manner. Furthermore, IGT intervention could remarkably decrease the expression of KLF4.Conclusion: Polarization of M1 macrophages may aggravate ABMR after renal transplant by promoting DSA-mediated endothelial cell injury, and IGT may attenuate the pathogenesis of ABMR by targeting KLF4.
Collapse
Affiliation(s)
- Zhou Hang
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jintao Wei
- Department of Emergency Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ming Zheng
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zeping Gui
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hao Chen
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Li Sun
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Shuang Fei
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhijian Han
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jun Tao
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zijie Wang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Zijie Wang, ; Min Gu, ; Ruoyun Tan,
| | - Ruoyun Tan
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Zijie Wang, ; Min Gu, ; Ruoyun Tan,
| | - Min Gu
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Zijie Wang, ; Min Gu, ; Ruoyun Tan,
| |
Collapse
|
37
|
Recruitment of monocytes primed to express heme oxygenase-1 ameliorates pathological lung inflammation in cystic fibrosis. Exp Mol Med 2022; 54:639-652. [PMID: 35581352 PMCID: PMC9166813 DOI: 10.1038/s12276-022-00770-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 02/07/2022] [Indexed: 12/04/2022] Open
Abstract
Overwhelming neutrophilic inflammation is a leading cause of lung damage in many pulmonary diseases, including cystic fibrosis (CF). The heme oxygenase-1 (HO-1)/carbon monoxide (CO) pathway mediates the resolution of inflammation and is defective in CF-affected macrophages (MΦs). Here, we provide evidence that systemic administration of PP-007, a CO releasing/O2 transfer agent, induces the expression of HO-1 in a myeloid differentiation factor 88 (MyD88) and phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT)-dependent manner. It also rescues the reduced HO-1 levels in CF-affected cells induced in response to lipopolysaccharides (LPS) or Pseudomonas aeruginosa (PA). Treatment of CF and muco-obstructive lung disease mouse models with a single clinically relevant dose of PP-007 leads to effective resolution of lung neutrophilia and to decreased levels of proinflammatory cytokines in response to LPS. Using HO-1 conditional knockout mice, we show that the beneficial effect of PP-007 is due to the priming of circulating monocytes trafficking to the lungs in response to infection to express high levels of HO-1. Finally, we show that PP-007 does not compromise the clearance of PA in the setting of chronic airway infection. Overall, we reveal the mechanism of action of PP-007 responsible for the immunomodulatory function observed in clinical trials for a wide range of diseases and demonstrate the potential use of PP-007 in controlling neutrophilic pulmonary inflammation by promoting the expression of HO-1 in monocytes/macrophages. The activity of an enzyme that is significantly reduced in cystic fibrosis (CF) could be boosted by an existing drug, reducing lung inflammation and associated tissue damage. Chronic inflammation in CF is currently treated using long-term corticosteroids which may leave patients immuno-suppressed, or high-dose ibuprofen, which is not well tolerated. Scientists hope to find alternative therapies targeting chronic inflammation. Emanuela Bruscia, Caterina Di Pietro (Yale University, New Haven, USA) and co-workers examined the mechanisms of action of the first-in-class drug PP-007 (Prolong Pharmaceuticals®) and assessed its potential for controlling inflammation in CF. Patients with CF have reduced expression of the heme oxygenase-1 enzyme in immune cells called monocytes. In CF mouse models, treatment with PP-007 boosted the expression of this enzyme in circulating monocytes. The treatment reduced levels of proinflammatory proteins and associated lung damage.
Collapse
|
38
|
Hecht JL, Janikova M, Choudhury R, Liu F, Canesin G, Janovicova L, Csizmadia E, Jorgensen EM, Esselen KM, Celec P, Swanson KD, Wegiel B. Labile Heme and Heme Oxygenase-1 Maintain Tumor-Permissive Niche for Endometriosis-Associated Ovarian Cancer. Cancers (Basel) 2022; 14:2242. [PMID: 35565370 PMCID: PMC9105072 DOI: 10.3390/cancers14092242] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/12/2022] [Accepted: 04/26/2022] [Indexed: 01/12/2023] Open
Abstract
Endometriosis, a painful gynecological condition accompanied by inflammation in women of reproductive age, is associated with an increased risk of ovarian cancer. We evaluated the role of peritoneal heme accumulated during menstrual cycling, as well as peritoneal and lesional macrophage phenotype, in promoting an oncogenic microenvironment. We quantified the heme-degrading enzyme, heme oxygenase-1 (HO-1, encoded by Hmox1) in normal peritoneum, endometriotic lesions and endometriosis-associated ovarian cancer (EAOC) of clear cell type (OCCC). HO-1 was expressed primarily in macrophages and increased in endometrioma and OCCC tissues relative to endometriosis and controls. Further, we compared cytokine expression profiles in peritoneal macrophages (PM) and peripheral blood mononuclear cells (PBMC) in women with endometriosis versus controls as a measure of a tumor-promoting environment in the peritoneum. We found elevated levels of HO-1 along with IL-10 and the pro-inflammatory cytokines (IL-1β, IL-16, IFNγ) in PM but not in PBMC from endometriosis patients. Using LysM-Cre:Hmox1flfl conditional knockout mice, we show that a deficiency of HO-1 in macrophages led to the suppression of growth of ID8 ovarian tumors implanted into the peritoneum. The restriction of ID8 ovarian tumor growth was associated with an increased number of Mac3+ macrophage and B cells in LysM-Cre:Hmox1flfl mice compared to controls. Functional experiments in ovarian cancer cell lines show that HO-1 is induced by heme. Low levels of exogenous heme promoted ovarian cancer colony growth in soft agar. Higher doses of heme led to slower cancer cell colony growth in soft agar and the induction of HO-1. These data suggest that perturbation of heme metabolism within the endometriotic niche and in cancer cells themselves may be an important factor that influences tumor initiation and growth.
Collapse
Affiliation(s)
- Jonathan L. Hecht
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Ave, Boston, MA 02215, USA;
| | - Monika Janikova
- Department of Surgery, Division of Surgical Sciences, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; (M.J.); (R.C.); (G.C.); (L.J.); (E.C.)
- Faculty of Medicine, Institute of Molecular Biomedicine, Comenius University in Bratislava, 814 99 Bratislava, Slovakia;
| | - Reeham Choudhury
- Department of Surgery, Division of Surgical Sciences, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; (M.J.); (R.C.); (G.C.); (L.J.); (E.C.)
| | - Fong Liu
- Department of OB/GYN, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA; (F.L.); (E.M.J.); (K.M.E.)
- Greater Baltimore Medical Center, 6569 Charles Street, Towson, MD 21204, USA
| | - Giacomo Canesin
- Department of Surgery, Division of Surgical Sciences, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; (M.J.); (R.C.); (G.C.); (L.J.); (E.C.)
- Vor Biopharma, 100 Cambridgepark Dr, Suite 400, Cambridge, MA 02140, USA
| | - Lubica Janovicova
- Department of Surgery, Division of Surgical Sciences, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; (M.J.); (R.C.); (G.C.); (L.J.); (E.C.)
- Faculty of Medicine, Institute of Molecular Biomedicine, Comenius University in Bratislava, 814 99 Bratislava, Slovakia;
| | - Eva Csizmadia
- Department of Surgery, Division of Surgical Sciences, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; (M.J.); (R.C.); (G.C.); (L.J.); (E.C.)
| | - Elisa M. Jorgensen
- Department of OB/GYN, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA; (F.L.); (E.M.J.); (K.M.E.)
| | - Katharine M. Esselen
- Department of OB/GYN, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA; (F.L.); (E.M.J.); (K.M.E.)
| | - Peter Celec
- Faculty of Medicine, Institute of Molecular Biomedicine, Comenius University in Bratislava, 814 99 Bratislava, Slovakia;
| | - Kenneth D. Swanson
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA;
| | - Barbara Wegiel
- Department of Surgery, Division of Surgical Sciences, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; (M.J.); (R.C.); (G.C.); (L.J.); (E.C.)
| |
Collapse
|
39
|
Zhu L, Lian W, Yao Z, Yang X, Wang Z, Lai Y, Xu S, Zhao B, Liu K. Integrated Analysis of Ferroptosis and Immunity-Related Genes Associated with Intestinal Ischemia/Reperfusion Injury. J Inflamm Res 2022; 15:2397-2411. [PMID: 35444445 PMCID: PMC9015787 DOI: 10.2147/jir.s351990] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 04/01/2022] [Indexed: 11/23/2022] Open
Abstract
PURPOSE Intestinal ischemia/reperfusion (I/R) injury is an unresolved clinical challenge due to its high prevalence, difficulty in diagnosis, and lack of clinically effective therapeutic agents. Ferroptosis is a novel form of cell-regulated death that has been shown to play a role in various I/R models and has been shown to be immune-related. Further unraveling the molecular mechanisms associated with ferroptosis and immunity in intestinal I/R injury may lead to the discovery of potentially effective drugs. METHODS We obtained differentially expressed mRNAs (DEGs) in mouse intestinal tissues following intestinal I/R injury or sham surgery. Then, we extracted ferroptosis-related DEGs (FRGs) and immune-related DEGs (IRGs) from the DEGs. In addition, we performed functional analysis of FRGs and IRGs. Next, we used transcriptome sequencing from patients with intestinal I/R injury to validate the results. Then, we constructed transcription factors (TFs)-gene networks and gene-drug networks using mouse and human co-expressed FRGs (coFRG) and mouse and human co-expressed IRGs (coIRG). We also analyzed the composition of immune cells to reveal correlations between FRGs signatures and immune cells in the mouse and human gut. Finally, we validated these results through animal experiments. RESULTS We extracted 61 FRGs and 294 IRGs from mouse samples and performed PPI and functional analyses. We extracted 45 FRGs and 200 IRGs from human samples for validation, and identified 24 coFRGs,100 coIRGs and 6 hub genes (HSPA5, GDF15, TNFAIP3, HMOX1, CXCL2 and IL6) in both. We also predicted potential TF-gene networks for coFRGs and coIRGs, as well as predicted gene-drug pairs for hub genes. In addition, we found that the immune cells were altered in the early stages of intestinal I/R injury and that FRGs were closely associated with immune cells in mice and humans. Finally, we validated the hub genes in mouse samples. CONCLUSION In conclusion, we identified ferroptosis and immunity-related genes to predict their correlations in intestinal I/R injury. We also predicated potential TF-genes network and potential therapeutic targets (HSPA5, GDF15, TNFAIP3, HMOX1, CXCL2 and IL6) to provide clues for further investigation of intestinal I/R injury.
Collapse
Affiliation(s)
- Lin Zhu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Wanyi Lian
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Zhiwen Yao
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Xiao Yang
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Ziyi Wang
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Yupei Lai
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Shiting Xu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Bingcheng Zhao
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Kexuan Liu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| |
Collapse
|
40
|
Kadono K, Kageyama S, Nakamura K, Hirao H, Ito T, Kojima H, Dery KJ, Li X, Kupiec-Weglinski JW. Myeloid Ikaros-SIRT1 signaling axis regulates hepatic inflammation and pyroptosis in ischemia-stressed mouse and human liver. J Hepatol 2022; 76:896-909. [PMID: 34871625 PMCID: PMC9704689 DOI: 10.1016/j.jhep.2021.11.026] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 11/17/2021] [Accepted: 11/19/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Although Ikaros (IKZF1) is a well-established transcriptional regulator in leukocyte lymphopoiesis and differentiation, its role in myeloid innate immune responses remains unclear. Sirtuin 1 (SIRT1) is a histone/protein deacetylase involved in cellular senescence, inflammation, and stress resistance. Whether SIRT1 signaling is essential in myeloid cell activation remains uncertain, while the molecular communication between Ikaros and SIRT1, two major transcriptional regulators, has not been studied. METHODS We undertook molecular and functional studies to interrogate the significance of the myeloid Ikaros-SIRT1 axis in innate immune activation and whether it may serve as a homeostatic sentinel in human liver transplant recipients (hepatic biopsies) and murine models of sterile hepatic inflammation (liver warm ischemia-reperfusion injury in wild-type, myeloid-specific Sirt1-knockout, and CD11b-DTR mice) as well as primary bone marrow-derived macrophage (BMM) cultures (Ikaros silencing vs. overexpression). RESULTS In our clinical study, we identified increased post-reperfusion hepatic Ikaros levels, accompanied by augmented inflammasome signaling yet depressed SIRT1, as a mechanism of hepatocellular damage in liver transplant recipients. In our experimental studies, we identified infiltrating macrophages as the major source of Ikaros in IR-stressed mouse livers. Then, we demonstrated that Ikaros-regulated pyroptosis - induced by canonical inflammasome signaling in BMM cultures - was SIRT1 dependent. Consistent with the latter, myeloid-specific Ikaros signaling augmented hepatic pyroptosis to aggravate pro-inflammatory responses in vivo by negatively regulating SIRT1 in an AMPK-dependent manner. Finally, myeloid-specific SIRT1 was required to suppress pyroptosis, pro-inflammatory phenotype, and ultimately mitigate hepatocellular injury in ischemia-stressed murine livers. CONCLUSION These findings identify the Ikaros-SIRT1 axis as a novel mechanistic biomarker of pyroptosis and a putative checkpoint regulator of homeostasis in response to acute hepatic stress/injury in mouse and human livers. LAY SUMMARY This report describes how crosstalk between Ikaros and SIRT1, two major transcriptional regulators, influence acute hepatic inflammation in murine models of liver ischemia-reperfusion injury and liver transplant recipients. We show that the myeloid Ikaros-SIRT1 axis regulates inflammasome-pyroptotic cell death and hepatocellular damage in stressed livers. Thus, the Ikaros-SIRT1 axis may serve as a novel checkpoint regulator that is required for homeostasis in response to acute liver injury in mice and humans.
Collapse
Affiliation(s)
- Kentaro Kadono
- Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Shoichi Kageyama
- Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA;,Division of Hepato-Biliary-Pancreatic Surgery and Transplantation, Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kojiro Nakamura
- Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA;,Division of Hepato-Biliary-Pancreatic Surgery and Transplantation, Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hirofumi Hirao
- Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Takahiro Ito
- Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Hidenobu Kojima
- Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Kenneth J. Dery
- Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Xiaoling Li
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences (NIEHS), Research Triangle Park, NC 27709, USA
| | - Jerzy W. Kupiec-Weglinski
- Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA;,Corresponding author. Address: Dumont-UCLA Transplant Center, 77-120 CHS, 10833 Le Conte Ave, Los Angeles, CA 90095, USA. Tel: (310) 825-4196; Fax: (310) 267-2358. (J.W. Kupiec-Weglinski)
| |
Collapse
|
41
|
Immune Regulation of Heme Oxygenase-1 in Allergic Airway Inflammation. Antioxidants (Basel) 2022; 11:antiox11030465. [PMID: 35326116 PMCID: PMC8944570 DOI: 10.3390/antiox11030465] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 02/09/2022] [Accepted: 02/23/2022] [Indexed: 11/17/2022] Open
Abstract
Heme oxygenase-1 (HO-1) is not only a rate-limiting enzyme in heme metabolism but is also regarded as a protective protein with an immunoregulation role in asthmatic airway inflammation. HO-1 exerts an anti-inflammation role in different stages of airway inflammation via regulating various immune cells, such as dendritic cells, mast cells, basophils, T cells, and macrophages. In addition, the immunoregulation role of HO-1 may differ according to subcellular locations.
Collapse
|
42
|
Schaefer REM, Callahan RC, Atif SM, Orlicky DJ, Cartwright IM, Fontenot AP, Colgan SP, Onyiah JC. Disruption of monocyte-macrophage differentiation and trafficking by a heme analog during active inflammation. Mucosal Immunol 2022; 15:244-256. [PMID: 34916594 PMCID: PMC8881314 DOI: 10.1038/s41385-021-00474-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 09/23/2021] [Accepted: 11/23/2021] [Indexed: 02/04/2023]
Abstract
Heme metabolism is a key regulator of inflammatory responses. Cobalt protoporphyrin IX (CoPP) is a heme analog and mimic that potently activates the NRF2/heme oxygenase-1 (HO-1) pathway, especially in monocytes and macrophages. We investigated the influence of CoPP on inflammatory responses using a murine model of colitis. Surprisingly, conditional deletion of myeloid HO-1 did not impact the colonic inflammatory response or the protective influence of CoPP in the setting of dextran sodium sulfate-induced colitis. Rather, we reveal that CoPP elicits a contradictory shift in blood myeloid populations relative to the colon during active intestinal inflammation. Major population changes include markedly diminished trafficking of CCR2+Ly6Chi monocytes to the inflamed colon, despite significant mobilization of this population into circulation. This resulted in significantly diminished colonic expansion of monocyte-derived macrophages and inflammatory cytokine expression. These findings were linked with significant induction of systemic CCL2 leading to a disrupted CCL2 chemoattractant gradient toward the colon and concentration-dependent suppression of circulating monocyte CCR2 expression. Administration of CoPP also induced macrophage differentiation toward a MarcohiHmox1hi anti-inflammatory erythrophagocytic phenotype, contributing to an overall decreased inflammatory profile. Such findings redefine protective influences of heme metabolism during inflammation, and highlight previously unreported immunosuppressive mechanisms of endogenous CCL2 induction.
Collapse
Affiliation(s)
- Rachel E. M. Schaefer
- Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, CO,Department of Medicine, University of Colorado School of Medicine, Aurora, CO,Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO
| | - Rosemary C. Callahan
- Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, CO,Department of Medicine, University of Colorado School of Medicine, Aurora, CO,Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO
| | - Shaikh M. Atif
- Division of Allergy, Asthma and Clinical Immunology, University of Colorado School of Medicine, Aurora, CO
| | - David J. Orlicky
- Department of Pathology, University of Colorado School of Medicine, Aurora, CO
| | - Ian M. Cartwright
- Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, CO,Department of Medicine, University of Colorado School of Medicine, Aurora, CO,Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO
| | - Andrew P. Fontenot
- Division of Allergy, Asthma and Clinical Immunology, University of Colorado School of Medicine, Aurora, CO
| | - Sean P. Colgan
- Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, CO,Department of Medicine, University of Colorado School of Medicine, Aurora, CO,Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO
| | - Joseph C. Onyiah
- Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, CO,Department of Medicine, University of Colorado School of Medicine, Aurora, CO,Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO, Corresponding author: Joseph C. Onyiah, M.D., University of Colorado School of Medicine, Rocky Mountain Regional VA Medical Center, 12700 East 19th Ave. MS B-146, Aurora, CO 80045,
| |
Collapse
|
43
|
Noel JG, Ramser SW, Pitstick L, Bonamer JP, Mackenzie B, Seu KG, Kalfa TA, Cancelas JA, Gardner JC. M-CSF supports medullary erythropoiesis and erythroid iron demand following burn injury through its activity on homeostatic iron recycling. Sci Rep 2022; 12:1235. [PMID: 35075211 PMCID: PMC8786861 DOI: 10.1038/s41598-022-05360-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 01/11/2022] [Indexed: 11/09/2022] Open
Abstract
M-CSF receptor signaling supports the development and survival of mononuclear phagocytes and is thought to play a role in post burn anemia by promoting myeloid lineage bias. We found M-CSF secretion was increased in burn patients and a murine model of post burn ACI, so we neutralized M-CSF in ACI mice to determine if erythropoiesis was improved. Instead, M-CSF blockade further impaired erythropoiesis and erythroid cells access to iron. M-CSF blockade enhanced inflammatory cytokine secretion, further increased systemic neutrophil counts, and led to tissue iron sequestration that was dependent, in part, on augmented IL-6 secretion which induced hepcidin. Deleterious effects of post burn M-CSF blockade were associated with arrest of an iron recycling gene expression signature in the liver and spleen that included Spi-C transcription factor and heme oxygenase-1, which promote heme metabolism and confer a non-inflammatory tone in macrophages. Hepatic induction of these factors in ACI mice was consistent with a recovery of ferroportin gene expression and reflected an M-CSF dependent expansion and differentiation of Spi-C+ monocytes into Kupffer cells. Together, this data indicates M-CSF secretion supports a homeostatic iron recycling program that plays a key role in the maintenance of erythroid cells access to iron following burn injury.
Collapse
Affiliation(s)
- John G Noel
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Cincinnati College of Medicine, Cincinnati, 45267, USA
| | - Seth W Ramser
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Cincinnati College of Medicine, Cincinnati, 45267, USA
| | - Lori Pitstick
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Cincinnati College of Medicine, Cincinnati, 45267, USA
| | - John P Bonamer
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, 45267, USA
| | - Bryan Mackenzie
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, 45267, USA
| | - Katie G Seu
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, 45229, USA
| | - Theodosia A Kalfa
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, 45229, USA
| | - Jose A Cancelas
- Divisions of Pathology and Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, 45229, USA
| | - Jason C Gardner
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Cincinnati College of Medicine, Cincinnati, 45267, USA.
| |
Collapse
|
44
|
Reactive Oxygen Species (ROS) and Antioxidants as Immunomodulators in Exercise: Implications for Heme Oxygenase and Bilirubin. Antioxidants (Basel) 2022; 11:antiox11020179. [PMID: 35204062 PMCID: PMC8868548 DOI: 10.3390/antiox11020179] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/12/2022] [Accepted: 01/14/2022] [Indexed: 02/07/2023] Open
Abstract
Exercise is commonly prescribed as a lifestyle treatment for chronic metabolic diseases as it functions as an insulin sensitizer, cardio-protectant, and essential lifestyle tool for effective weight maintenance. Exercise boosts the production of reactive oxygen species (ROS) and subsequent transient oxidative damage, which also upregulates counterbalancing endogenous antioxidants to protect from ROS-induced damage and inflammation. Exercise elevates heme oxygenase-1 (HO-1) and biliverdin reductase A (BVRA) expression as built-in protective mechanisms, which produce the most potent antioxidant, bilirubin. Together, these mitigate inflammation and adiposity. Moderately raising plasma bilirubin protects in two ways: (1) via its antioxidant capacity to reduce ROS and inflammation, and (2) its newly defined function as a hormone that activates the nuclear receptor transcription factor PPARα. It is now understood that increasing plasma bilirubin can also drive metabolic adaptions, which improve deleterious outcomes of weight gain and obesity, such as inflammation, type II diabetes, and cardiovascular diseases. The main objective of this review is to describe the function of bilirubin as an antioxidant and metabolic hormone and how the HO-1-BVRA-bilirubin-PPARα axis influences inflammation, metabolic function and interacts with exercise to improve outcomes of weight management.
Collapse
|
45
|
Yu S, Lu J. Macrophages in transplant rejection. Transpl Immunol 2022; 71:101536. [PMID: 35017096 DOI: 10.1016/j.trim.2022.101536] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 01/04/2022] [Accepted: 01/06/2022] [Indexed: 11/17/2022]
Abstract
Transplant rejection is one of the primary factors leading to loss of allograft function, and macrophages are involved in allograft rejection. Macrophages polarize into different phenotypes according to stimulation by different external factors. Different types of macrophages play distinct roles in inflammation, tumors, and autoimmune diseases and are involved in transplant rejection. In this review, we introduce the origin and migration of macrophages, outline the classification of macrophages and their polarization mechanisms, and review the currently understood mechanisms of their involvement in transplant rejection. Finally, we discuss the regulation of macrophage polarization and miRNA expression with respect to transplant rejection, which is important for the development of new anti-rejection therapies.
Collapse
Affiliation(s)
- Shaochen Yu
- Department of Emergency and Critical Care Medicine, Guangdong Second Provincial General Hospital, No. 466, Xingang Middle Road, Haizhu District, Guangzhou, Guangdong 510317, China.
| | - Jian Lu
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Shushan District, Hefei, Anhui 230022, China.
| |
Collapse
|
46
|
Trichosanthis Semen Suppresses Lipopolysaccharide-Induced Neuroinflammation by Regulating the NF-κB Signaling Pathway and HO-1 Expression in Microglia. Toxins (Basel) 2021; 13:toxins13120898. [PMID: 34941735 PMCID: PMC8704237 DOI: 10.3390/toxins13120898] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 12/10/2021] [Indexed: 01/08/2023] Open
Abstract
Neuroinflammation, which is mediated by microglia that release various inflammatory cytokines, is a typical feature of neurodegenerative diseases (NDDs), such as Alzheimer’s disease and Parkinson’s disease. Hence, alleviating neuroinflammation by downregulating pro-inflammatory action, and upregulating anti-inflammatory action of microglia is an efficient therapeutic target for NDDs. In this study, we evaluated whether trichosanthis semen (TS), a dried ripe seed of Trichosanthes kirilowii Maximowicz, reduces lipopolysaccharide (LPS)-induced neuroinflammation by regulating microglial responses in vitro and in vivo. Our results presented that TS reduced the release of pro-inflammatory mediators, such as nitric oxide (NO), inducible NO synthase, tumor necrosis factor-α, interleukin-1β, and interleukin-6 via inhibition of the nuclear factor kappa B (NF-κB) signaling pathway in LPS-treated BV2 microglial cells. Moreover, TS induced anti-inflammatory mediators, such as interleukin-10, found in inflammatory zone 1, and chitinase 3-like 3 by the upregulation of heme oxygenase 1 (HO-1). We further confirmed that TS administration suppressed microglial activation, but enhanced HO-1 expression in LPS-injected mice. These results suggest that TS has anti-neuroinflammatory effects via inhibition of NF-κB signaling through the activation of HO-1, and that TS may be a therapeutical candidate for NDDs treatment.
Collapse
|
47
|
Kang IS, Kim RI, Kim C. Carbon Monoxide Regulates Macrophage Differentiation and Polarization toward the M2 Phenotype through Upregulation of Heme Oxygenase 1. Cells 2021; 10:3444. [PMID: 34943953 PMCID: PMC8700076 DOI: 10.3390/cells10123444] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/30/2021] [Accepted: 12/02/2021] [Indexed: 12/24/2022] Open
Abstract
Carbon monoxide (CO) is generated by heme oxygenase (HO), and HO-1 is highly induced in monocytes and macrophages upon stimulation. Monocytes differentiate into macrophages, including pro-inflammatory (M1) and anti-inflammatory (M2) cells, in response to environmental signals. The present study investigated whether CO modulates macrophage differentiation and polarization, by applying the CO-releasing molecule-3 (CORM-3). Results showed that murine bone marrow cells are differentiated into macrophages by CORM-3 in the presence of macrophage colony-stimulating factor. CORM-3 increases expressions of macrophage markers, including F4/80 and CD11b, and alters the cell morphology into elongated spindle-shaped cells, which is a typical morphology of M2 cells. CORM-3 upregulates the expressions of genes and molecules involved in M2 polarization and M2 phenotype markers, such as STAT6, PPARγ, Ym1, Fizz1, arginase-1, and IL-10. However, exposure to CORM-3 inhibits the iNOS expression, suggesting that CO enhances macrophage differentiation and polarization toward M2. Increased HO-1 expression is observed in differentiated macrophages, and CORM-3 further increases this expression. Hemin, an HO-1 inducer, results in increased macrophage differentiation, whereas the HO-1 inhibitor zinc protoporphyrin IX inhibits differentiation. In addition, CORM-3 increases the proportion of macrophages in peritoneal exudate cells and enhances the expression of HO-1 and arginase-1 but inhibits iNOS. Taken together, these results suggest that the abundantly produced CO in activated macrophages enhances proliferation, differentiation, and polarization toward M2. It will probably help clear apoptotic cells, resolve inflammation, and promote wound healing and tissue remodeling.
Collapse
Affiliation(s)
- In-Soon Kang
- Laboratory of Leukocyte Signaling Research, Department of Pharmacology, College of Medicine, Inha University, Incheon 22212, Korea; (I.-S.K.); (R.-I.K.)
| | - Rang-Ie Kim
- Laboratory of Leukocyte Signaling Research, Department of Pharmacology, College of Medicine, Inha University, Incheon 22212, Korea; (I.-S.K.); (R.-I.K.)
| | - Chaekyun Kim
- Laboratory of Leukocyte Signaling Research, Department of Pharmacology, College of Medicine, Inha University, Incheon 22212, Korea; (I.-S.K.); (R.-I.K.)
- BK21 Program in Biomedical Science & Engineering, Inha University, Incheon 22212, Korea
- Convergent Research Center for Metabolism and Immunoregulation, Inha University, Incheon 22212, Korea
| |
Collapse
|
48
|
Guo C, Zhang C, Xia Z, Song B, Hu W, Cui Y, Xue Y, Xia M, Xu D, Zhang S, Fang J. Nano-designed CO donor ameliorates bleomycin-induced pulmonary fibrosis via macrophage manipulation. J Control Release 2021; 341:566-577. [PMID: 34864115 DOI: 10.1016/j.jconrel.2021.11.047] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 11/29/2021] [Indexed: 02/08/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and irreversible interstitial pulmonary disease due to chronic inflammatory responses. The prognosis of IPF is very poor, however, the therapeutic options are very limited. Previously we developed a polymeric micellar drug delivery system of carbon monoxide (CO) that is a pivotal anti-inflammatory gaseous molecule, i.e., SMA/CORM2, which exhibited therapeutic potentials against dextran sulfate sodium (DSS)-induced mouse colitis and acetaminophen (APAP) induced liver injury. Along this line, here we investigate the applicability of SMA/CORM2 on IPF using a bleomycin (BLM)-induced pulmonary fibrosis model. Severe inflammation and the consequent pulmonary fibrosis were triggered by BLM, whereas SMA/CORM2 treatment remarkably suppressed the inflammation progression and ameliorated the formation of fibrosis. CO is the effector molecule of SMA/CORM2, which exerted the therapeutic/protective effect mostly through suppressing the reprogramming of anti-inflammatory macrophages as revealed by the decreased expressions of CD206 and arginase-1 that were remarkably upregulated by BLM exposure. The suppression of macrophage polarization accompanied the downregulated hypoxia-inducible factor-1α (HIF-1α) and its target molecule heme oxygenase-1 (HO-1), suggesting a HIF-1α/HO-1 pathway for modulating macrophage reprogramming. As the downstream event of anti-inflammatory macrophage polarization, the alveolar epithelial to mesenchymal transition that is the major source of myofibroblast, the hallmark of IPF, was significantly suppressed by SMA/CORM2 via a TGF-β/Smad2/3 pathway. Compared to native CORM2 of equivalent dose, SMA/CROM2 exhibited a much better protective effect indicating its superior bioavailability as an enhanced permeability and retention (EPR) effect-based nanomedicine. We thus anticipate the application of SMA/CORM2 as a therapeutic candidate for IPF as well as other inflammatory diseases and disorders.
Collapse
Affiliation(s)
- Chunyu Guo
- Department of Toxicology, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei 230032, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, School of Public Health, Anhui Medical University, Hefei 230022, China
| | - Cheng Zhang
- Department of Toxicology, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei 230032, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, School of Public Health, Anhui Medical University, Hefei 230022, China
| | - Zhengmei Xia
- Department of Toxicology, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei 230032, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, School of Public Health, Anhui Medical University, Hefei 230022, China
| | - Bingdong Song
- Department of Toxicology, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei 230032, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, School of Public Health, Anhui Medical University, Hefei 230022, China
| | - Weirong Hu
- Department of Toxicology, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei 230032, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, School of Public Health, Anhui Medical University, Hefei 230022, China
| | - Yingying Cui
- Department of Toxicology, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei 230032, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, School of Public Health, Anhui Medical University, Hefei 230022, China
| | - Yanni Xue
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei 230022, China; MOE Key Laboratory of Population Health Across Life Cycle, Anhui Provincial Key Laboratory of Population Health and Aristogenics, No. 81 Meishan Road, Hefei 230032, China
| | - Mizhen Xia
- School of Life Science, Anhui Medical University, No. 81 Meishan Road, Hefei 230032, China
| | - Dexiang Xu
- Department of Toxicology, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei 230032, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, School of Public Health, Anhui Medical University, Hefei 230022, China
| | - Shichen Zhang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei 230022, China; MOE Key Laboratory of Population Health Across Life Cycle, Anhui Provincial Key Laboratory of Population Health and Aristogenics, No. 81 Meishan Road, Hefei 230032, China; School of Public Health and Health Management, Anhui Medical College, No. 632 Furong Road, Hefei 230601, China.
| | - Jun Fang
- Department of Toxicology, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei 230032, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, School of Public Health, Anhui Medical University, Hefei 230022, China; Faculty of Pharmaceutical Science, Sojo University, Ikeda 4-22-1, Kumamoto 860-0082, Japan.
| |
Collapse
|
49
|
Seong JB, Kim B, Kim S, Kim MH, Park YH, Lee Y, Lee HJ, Hong CW, Lee DS. Macrophage peroxiredoxin 5 deficiency promotes lung cancer progression via ROS-dependent M2-like polarization. Free Radic Biol Med 2021; 176:322-334. [PMID: 34637923 DOI: 10.1016/j.freeradbiomed.2021.10.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 10/08/2021] [Accepted: 10/09/2021] [Indexed: 12/24/2022]
Abstract
Strategies for cancer treatment have traditionally focused on suppressing cancer cell behavior, but many recent studies have demonstrated that regulating the tumor microenvironment (TME) can also inhibit disease progression. Macrophages are major TME components, and the direction of phenotype polarization is known to regulate tumor behavior, with M2-like polarization promoting progression. It is also known that reactive oxygen species (ROS) in macrophages drive M2 polarization, and M2 polarization promote lung cancer progression. Lung cancer patients with lower expression of the antioxidant enzyme peroxiredoxin 5 (Prx5) demonstrate poorer survival. This study revealed that Prx5 deficiency in macrophages induced M2 macrophage polarization by lung cancer. We report that injection of lung cancer cells produced larger tumors in Prx5-deficit mice than wild-type mice independent of cancer cell Prx5 expression. Through co-culture with lung cancer cell lines, Prx5-deficient macrophages exhibited M2 polarization, and reduced expression levels of the M1-associated inflammatory factors iNOS, TNFα, and Il-1β. Moreover, these Prx5-deficient macrophages promoted the proliferation and migration of co-cultured lung cancer cells. Conversely, suppression of ROS generation by N-acetyl cysteine (NAC) inhibited the M2-like polarization of Prx5-deficient macrophages, increased expression levels of inflammatory factors, inhibited the proliferation and migration of co-cultured lung cancer cells, and suppressed tumor growth in mice. These findings suggest that blocking the M2 polarization of macrophages may promote lung cancer regression.
Collapse
Affiliation(s)
- Jung Bae Seong
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, South Korea; College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea; National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, South Korea
| | - Bokyung Kim
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, South Korea; College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Soyoon Kim
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, South Korea; College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Mi Hye Kim
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, South Korea; College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Young-Ho Park
- Futuristic Animal Resource & Research Center (FARRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, South Korea
| | - Youngjeon Lee
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, South Korea
| | - Hong Jun Lee
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea; Research Institute eBiogen Inc., Seoul, Republic of Korea
| | - Chang-Won Hong
- Department of Physiology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Dong-Seok Lee
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, South Korea; College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea.
| |
Collapse
|
50
|
Gwak SY, Kim SJ, Park J, Kim SH, Joe Y, Lee HN, Kim W, Muna IA, Na HK, Chung HT, Surh YJ. Potential Role of Heme Oxygenase-1 in the Resolution of Experimentally Induced Colitis through Regulation of Macrophage Polarization. Gut Liver 2021; 16:246-258. [PMID: 34737242 PMCID: PMC8924814 DOI: 10.5009/gnl210058] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 05/20/2021] [Accepted: 05/24/2021] [Indexed: 12/01/2022] Open
Abstract
Background/Aims Heme oxygenase-1 (HO-1) plays a central role in cellular defense against inflammatory insults, and its induction in macrophages potentiates their efferocytic activity. In this study, we explored the potential role of macrophage HO-1 in the resolution of experimentally induced colitis. Methods To induce colitis, male C57BL/6 mice were treated with 2% dextran sulfate sodium (DSS) in the drinking water for 7 days. To investigate efferocytosis, apoptotic colon epithelial CCD 841 CoN cells were coincubated with bone marrow-derived macrophages (BMDMs). Results Administration of the HO-1 inhibitor zinc protoporphyrin IX (ZnPP) blunted the resolution of DSS-induced intestinal inflammation and expression of the proresolving M2 macrophage marker CD206. BMDMs treated with apoptotic colonic epithelial cells showed significantly elevated expression of HO-1 and its regulator Nrf2. Under the same experimental conditions, the proportion of CD206-expressing macrophages was also enhanced. ZnPP treatment abrogated the upregulation of CD206 expression in BMDMs engulfing apoptotic colonic epithelial cells. This result was verified with BMDMs isolated from HO-1-knockout mice. BMDMs, when stimulated with lipopolysaccharide, exhibited increased expression of CD86, a marker of M1 macrophages. Coculture of lipopolysaccharide-stimulated BMDMs with apoptotic colonic epithelial cell debris dampened the expression of CD86 as well as the pro-inflammatory cytokines in an HO-1-dependent manner. Genetic ablation as well as pharmacologic inhibition of HO-1 significantly reduced the proportion of efferocytic BMDMs expressing the scavenger receptor CD36. Conclusions HO-1 plays a key role in the resolution of experimentally induced colitis by modulating the polarization of macrophages.
Collapse
Affiliation(s)
- Shin-Young Gwak
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul, Korea
| | - Su-Jung Kim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Korea
| | - Jeongmin Park
- Department of Biological Sciences, University of Ulsan, Ulsan, Korea
| | - Seung Hyeon Kim
- Cancer Research Institute, Seoul National University, Seoul, Korea
| | - Yeonsoo Joe
- Department of Biological Sciences, University of Ulsan, Ulsan, Korea
| | - Ha-Na Lee
- Laboratory of Immunology, Division of Biotechnology Review and Research-III, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD
| | - Wonki Kim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Korea
| | - Ishrat Aklima Muna
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Korea
| | - Hye-Kyung Na
- Department of Food Science and Biotechnology, College of Knowledge-Based Services Engineering, Sungshin Women's University, Seoul, Korea
| | - Hun Taeg Chung
- Department of Biological Sciences, University of Ulsan, Ulsan, Korea
| | - Young-Joon Surh
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul, Korea.,Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Korea.,Cancer Research Institute, Seoul National University, Seoul, Korea
| |
Collapse
|