1
|
Yasuda T, Deans K, Shankar A, Chilton R. The web of intrigue: unraveling the role of NETosis within the gut-microbiome-immune-heart axis in acute myocardial infarction and heart failure. Cardiovasc Endocrinol Metab 2024; 13:e0309. [PMID: 39130369 PMCID: PMC11315478 DOI: 10.1097/xce.0000000000000309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 06/06/2024] [Indexed: 08/13/2024]
Abstract
This review summarizes the role of NETosis, or the release of neutrophil extracellular traps (NETs), and its interplay with the gut microbiome in acute myocardial infarction (AMI) and heart failure. NETosis contributes to inflammation, thrombosis, and atherothrombosis, all central to the pathophysiology of AMI and heart failure. NETosis can be activated by inflammation and dietary factors, indicating association with metabolic conditions. In cases of heart failure, NETosis is regulated by inflammatory molecules such as C-reactive protein (CRP), and Krüppel-like factor 2 (KLF2) - a protein that plays a role in controlling inflammation, and angiotensin II. Changes in the gut microbiome are linked to the severity and recovery of cardiac injury post-AMI and heart failure progression. The microbiome's influence extends to immune modulation and inflammatory responses, potentially affecting NETosis.
Collapse
Affiliation(s)
- Tai Yasuda
- Department of Anesthesiology, University Hospital, UTHSC San Antonio
| | - Kate Deans
- Department of Cardiology, South Texas Department of Veteran Affairs
| | - Aditi Shankar
- Department of Cardiology, University Hospital, UTHSC San Antonio, San Antonio, Texas, USA
| | - Robert Chilton
- Department of Cardiology, South Texas Department of Veteran Affairs
- Department of Cardiology, University Hospital, UTHSC San Antonio, San Antonio, Texas, USA
| |
Collapse
|
2
|
Yang K, Gao R, Chen H, Hu J, Zhang P, Wei X, Shi J, Chen Y, Zhang L, Chen J, Lyu Y, Dong Z, Wei W, Hu K, Guo Y, Ge J, Sun A. Myocardial reperfusion injury exacerbation due to ALDH2 deficiency is mediated by neutrophil extracellular traps and prevented by leukotriene C4 inhibition. Eur Heart J 2024; 45:1662-1680. [PMID: 38666340 PMCID: PMC11089336 DOI: 10.1093/eurheartj/ehae205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 02/18/2024] [Accepted: 03/19/2024] [Indexed: 05/14/2024] Open
Abstract
BACKGROUND AND AIMS The Glu504Lys polymorphism in the aldehyde dehydrogenase 2 (ALDH2) gene is closely associated with myocardial ischaemia/reperfusion injury (I/RI). The effects of ALDH2 on neutrophil extracellular trap (NET) formation (i.e. NETosis) during I/RI remain unknown. This study aimed to investigate the role of ALDH2 in NETosis in the pathogenesis of myocardial I/RI. METHODS The mouse model of myocardial I/RI was constructed on wild-type, ALDH2 knockout, peptidylarginine deiminase 4 (Pad4) knockout, and ALDH2/PAD4 double knockout mice. Overall, 308 ST-elevation myocardial infarction patients after primary percutaneous coronary intervention were enrolled in the study. RESULTS Enhanced NETosis was observed in human neutrophils carrying the ALDH2 genetic mutation and ischaemic myocardium of ALDH2 knockout mice compared with controls. PAD4 knockout or treatment with NETosis-targeting drugs (GSK484, DNase1) substantially attenuated the extent of myocardial damage, particularly in ALDH2 knockout. Mechanistically, ALDH2 deficiency increased damage-associated molecular pattern release and susceptibility to NET-induced damage during myocardial I/RI. ALDH2 deficiency induced NOX2-dependent NETosis via upregulating the endoplasmic reticulum stress/microsomal glutathione S-transferase 2/leukotriene C4 (LTC4) pathway. The Food and Drug Administration-approved LTC4 receptor antagonist pranlukast ameliorated I/RI by inhibiting NETosis in both wild-type and ALDH2 knockout mice. Serum myeloperoxidase-DNA complex and LTC4 levels exhibited the predictive effect on adverse left ventricular remodelling at 6 months after primary percutaneous coronary intervention in ST-elevation myocardial infarction patients. CONCLUSIONS ALDH2 deficiency exacerbates myocardial I/RI by promoting NETosis via the endoplasmic reticulum stress/microsomal glutathione S-transferase 2/LTC4/NOX2 pathway. This study hints at the role of NETosis in the pathogenesis of myocardial I/RI, and pranlukast might be a potential therapeutic option for attenuating I/RI, particularly in individuals with the ALDH2 mutation.
Collapse
Affiliation(s)
- Kun Yang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai 200032, China
| | - Rifeng Gao
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai 200032, China
- Department of Cardiology, The Fifth People’s Hospital of Shanghai, Fudan University, 128 Ruili Road, Shanghai 200240, China
- Department of Cardiac Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, China
| | - Hanchuan Chen
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai 200032, China
| | - Jingjing Hu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai 200032, China
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou 310006, China
| | - Peng Zhang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai 200032, China
- Department of Cardiology, Minhang Hospital affiliated to Fudan University, 170 Xinsong Road, Shanghai 201100, China
| | - Xiang Wei
- Department of Cardiology, The Fifth People’s Hospital of Shanghai, Fudan University, 128 Ruili Road, Shanghai 200240, China
| | - Jiaran Shi
- Department of Cardiology, Lihuili Hospital Facilitated to Ningbo University, 57 Xingning Road, Ningbo 315040, China
| | - Yinyin Chen
- Department of Radiology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Department of Medical Imaging, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Liwei Zhang
- Department of Cardiology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, 134 Dongjie Road, Fuzhou 350001, China
| | - Juntao Chen
- Department of Urology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Yang Lyu
- Department of Cardiology, The Fifth People’s Hospital of Shanghai, Fudan University, 128 Ruili Road, Shanghai 200240, China
| | - Zhen Dong
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai 200032, China
| | - Wei Wei
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, 241 West Huaihai Road, Shanghai 200030, China
| | - Kai Hu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai 200032, China
| | - Yansong Guo
- Department of Cardiology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, 134 Dongjie Road, Fuzhou 350001, China
- Fujian Provincial Key Laboratory of Cardiovascular Disease, Fujian Provincial Center for Geriatrics, Fujian Provincial Clinical Research Center for Severe Acute Cardiovascular Diseases, 134 Dongjie Road, Fuzhou 350001, China
- Fujian Heart Failure Center Alliance, 134 Dongjie Road, Fuzhou 350001, China
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai 200032, China
- Institutes of Biomedical Sciences, Fudan University, 131 Dongan Road, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, 180 Fenglin Road, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, 180 Fenglin Road, Shanghai 200032, China
| | - Aijun Sun
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai 200032, China
- Institutes of Biomedical Sciences, Fudan University, 131 Dongan Road, Shanghai 200032, China
| |
Collapse
|
3
|
Barbu E, Mihaila AC, Gan AM, Ciortan L, Macarie RD, Tucureanu MM, Filippi A, Stoenescu AI, Petrea SV, Simionescu M, Balanescu SM, Butoi E. The Elevated Inflammatory Status of Neutrophils Is Related to In-Hospital Complications in Patients with Acute Coronary Syndrome and Has Important Prognosis Value for Diabetic Patients. Int J Mol Sci 2024; 25:5107. [PMID: 38791147 PMCID: PMC11121518 DOI: 10.3390/ijms25105107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 05/03/2024] [Accepted: 05/05/2024] [Indexed: 05/26/2024] Open
Abstract
Despite neutrophil involvement in inflammation and tissue repair, little is understood about their inflammatory status in acute coronary syndrome (ACS) patients with poor outcomes. Hence, we investigated the potential correlation between neutrophil inflammatory markers and the prognosis of ACS patients with/without diabetes and explored whether neutrophils demonstrate a unique inflammatory phenotype in patients experiencing an adverse in-hospital outcome. The study enrolled 229 ACS patients with or without diabetes. Poor evolution was defined as either death, left ventricular ejection fraction (LVEF) <40%, Killip Class 3/4, ventricular arrhythmias, or mechanical complications. Univariate and multivariate analyses were employed to identify clinical and paraclinical factors associated with in-hospital outcomes. Neutrophils isolated from fresh blood were investigated using qPCR, Western blot, enzymatic assay, and immunofluorescence. Poor evolution post-myocardial infarction (MI) was associated with increased number, activity, and inflammatory status of neutrophils, as indicated by significant increase of Erythrocyte Sedimentation Rate (ESR), C-reactive protein (CRP), fibrinogen, interleukin-1β (IL-1β), and, interleukin-6 (IL-6). Among the patients with complicated evolution, neutrophil activity had an important prognosis value for diabetics. Neutrophils from patients with unfavorable evolution revealed a pro-inflammatory phenotype with increased expression of CCL3, IL-1β, interleukin-18 (IL-18), S100A9, intracellular cell adhesion molecule-1 (ICAM-1), matrix metalloprotease (MMP-9), of molecules essential in reactive oxygen species (ROS) production p22phox and Nox2, and increased capacity to form neutrophil extracellular traps. Inflammation is associated with adverse short-term prognosis in acute ACS, and inflammatory biomarkers exhibit greater specificity in predicting short-term outcomes in diabetics. Moreover, neutrophils from patients with unfavorable evolution exhibit distinct inflammatory patterns, suggesting that alterations in the innate immune response in this subgroup may exert detrimental effects on disease progression.
Collapse
Affiliation(s)
- Elena Barbu
- Department of Cardiology, Elias Emergency Hospital, 011461, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (A.I.S.); (S.M.B.)
| | - Andreea Cristina Mihaila
- Biopathology and Therapy of Inflammation, Institute of Cellular Biology and Pathology “Nicolae Simionescu”, 050568 Bucharest, Romania; (A.C.M.); (A.-M.G.); (L.C.); (R.D.M.); (M.M.T.); (M.S.)
| | - Ana-Maria Gan
- Biopathology and Therapy of Inflammation, Institute of Cellular Biology and Pathology “Nicolae Simionescu”, 050568 Bucharest, Romania; (A.C.M.); (A.-M.G.); (L.C.); (R.D.M.); (M.M.T.); (M.S.)
| | - Letitia Ciortan
- Biopathology and Therapy of Inflammation, Institute of Cellular Biology and Pathology “Nicolae Simionescu”, 050568 Bucharest, Romania; (A.C.M.); (A.-M.G.); (L.C.); (R.D.M.); (M.M.T.); (M.S.)
| | - Razvan Daniel Macarie
- Biopathology and Therapy of Inflammation, Institute of Cellular Biology and Pathology “Nicolae Simionescu”, 050568 Bucharest, Romania; (A.C.M.); (A.-M.G.); (L.C.); (R.D.M.); (M.M.T.); (M.S.)
| | - Monica Madalina Tucureanu
- Biopathology and Therapy of Inflammation, Institute of Cellular Biology and Pathology “Nicolae Simionescu”, 050568 Bucharest, Romania; (A.C.M.); (A.-M.G.); (L.C.); (R.D.M.); (M.M.T.); (M.S.)
| | - Alexandru Filippi
- Department of Biochemistry and Biophysics, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania;
| | - Andra Ioana Stoenescu
- Department of Cardiology, Elias Emergency Hospital, 011461, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (A.I.S.); (S.M.B.)
| | | | - Maya Simionescu
- Biopathology and Therapy of Inflammation, Institute of Cellular Biology and Pathology “Nicolae Simionescu”, 050568 Bucharest, Romania; (A.C.M.); (A.-M.G.); (L.C.); (R.D.M.); (M.M.T.); (M.S.)
| | - Serban Mihai Balanescu
- Department of Cardiology, Elias Emergency Hospital, 011461, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (A.I.S.); (S.M.B.)
| | - Elena Butoi
- Biopathology and Therapy of Inflammation, Institute of Cellular Biology and Pathology “Nicolae Simionescu”, 050568 Bucharest, Romania; (A.C.M.); (A.-M.G.); (L.C.); (R.D.M.); (M.M.T.); (M.S.)
| |
Collapse
|
4
|
Cao C, Yu P, Chu C, Wang Z, Xu W, Cheng F, Zhao H, Qiu Z. Magnesium hydride attenuates intestinal barrier injury during hemorrhage shock by regulating neutrophil extracellular trap formation via the ROS/MAPK/PAD4 pathway. Int Immunopharmacol 2024; 130:111688. [PMID: 38394886 DOI: 10.1016/j.intimp.2024.111688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 01/31/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024]
Abstract
Magnesium hydride (MgH2) is a hydrogen storage material that is known for its high capacity and safety and is capable of releasing hydrogen in a controlled manner when administered orally. This release of hydrogen has been associated with a range of biological effects, including anti-inflammatory properties, antioxidant activity, and protection of the intestinal barrier. Previous research has shown that neutrophil extracellular traps (NETs) play a role in the dysfunction of the intestinal barrier in conditions such as sepsis and critical illnesses. However, it remains unclear as to whether MgH2 can protect the intestinal barrier by inhibiting NET formation, and the underlying mechanisms have yet to be elucidated. A rat model of hemorrhagic shock was created, and pretreatment or posttreatment procedures with MgH2 were performed. After 24 h, samples from the small intestine and blood were collected for analysis. In vitro, human neutrophils were incubated with either phorbol-12-myristate-13-acetate (PMA) or MgH2. Reactive oxygen species generation and the expression of key proteins were assessed. The results demonstrated that MgH2 administration led to a decrease in inflammatory cytokines in the serum and mitigated distant organ dysfunction in rats with HS. Furthermore, MgH2 treatment reversed histopathological damage in the intestines, improved intestinal permeability, and enhanced the expression of tight junction proteins (TJPs) during HS. Additionally, MgH2 treatment was found to suppress NET formation in the intestines. In vitro pretreatment with MgH2 alleviated intestinal monolayer barrier disruption that was induced by NETs. Mechanistically, MgH2 pretreatment reduced ROS production and NET formation, inhibited the activation of ERK and p38, and suppressed the expression of the PAD4 protein. These findings indicated that MgH2 may inhibit NET formation in a ROS/MAPK/PAD4-dependent manner, which reduces NET-related intestinal barrier damage, thus offering a novel protective role in preventing intestinal barrier dysfunction during HS.
Collapse
Affiliation(s)
- Changkui Cao
- Department of Emergency Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu City, Anhui Province, China; Department of Emergency, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Pan Yu
- Department of Burn and Plastic Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Chengnan Chu
- Department of Emergency, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Zhenjie Wang
- Department of Emergency Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu City, Anhui Province, China
| | - Weiqi Xu
- Department of Emergency, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Feng Cheng
- Department of Emergency Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu City, Anhui Province, China
| | - Heng Zhao
- Department of Emergency Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu City, Anhui Province, China
| | - Zhaolei Qiu
- Department of Emergency Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu City, Anhui Province, China.
| |
Collapse
|
5
|
Chen Q, Gu Q, Yin A, Cai D, Xiao T, Wang Y, Ji Y, Wang Q, Wei J, Sun L. Neutrophil Percentage as a Potential Biomarker of Acute Kidney Injury Risk and Short-Term Prognosis in Patients with Acute Myocardial Infarction in the Elderly. Clin Interv Aging 2024; 19:503-515. [PMID: 38525316 PMCID: PMC10959300 DOI: 10.2147/cia.s455588] [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: 12/18/2023] [Accepted: 03/12/2024] [Indexed: 03/26/2024] Open
Abstract
Objective This study aimed to explore the association of preoperative neutrophil percentage (NEUT%) with the risk of acute kidney injury (AKI) in patients with acute myocardial infarction (AMI) having undergone coronary interventional therapy. Methods A single-center, retrospective and observational study was conducted. From December 2012 to June 2021, patients with AMI were enrolled and divided into AKI group and non-AKI group. The NEUT% in the two groups was compared. The association between NEUT% with the risk of post-AMI AKI was analyzed by univariate and multivariable logistic regression. Kaplan-Meier survival curve was drawn to evaluate the prognostic ability of NEUT% for short-term all-cause death following AMI. Results A total of 3001 consecutive patients were enrolled with an average age of 64.38 years. AKI occurred in 327 (10.9%) patients. The NEUT% was higher in the AKI group than in the non-AKI group ([76.65±11.43]% versus [73.22±11.83]%, P<0.001). NEUT% was also identified as an independent risk factor for AKI in AMI patients after adjustment (OR=1.021, 95% CI: 1.010-1.033, P < 0.001). Compared with those at the lowest quartile of NEUT%, the patients at quartiles 2-4 had a higher risk of AKI (P for trend = 0.003). The odds of AKI increased by 29.0% as NEUT% increased by 1 standard deviation (OR=1.290, 95% CI: 1.087-1.531, P = 0.004). After a median of 35 days follow-up, 93 patients died. Patients with a higher NEUT% presented a higher risk of all-cause death after AMI (Log rank: χ2 =24.753, P<0.001). Conclusion In AMI patients, the peripheral blood NEUT% was positively associated with the odds of AKI and short-term all-cause mortality. NEUT% may provide physicians with more information about disease development and prognosis.
Collapse
Affiliation(s)
- Qianwen Chen
- Department of Cardiology, the Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, 213000, People’s Republic of China
| | - Qingqing Gu
- Department of Cardiology, the Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, 213000, People’s Republic of China
| | - Anwen Yin
- Department of Cardiology, the Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, Jiangsu, 214023, People’s Republic of China
| | - Dabei Cai
- Department of Cardiology, the Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, 213000, People’s Republic of China
| | - Tingting Xiao
- Department of Cardiology, the Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, 213000, People’s Republic of China
| | - Yu Wang
- Department of Cardiology, the Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, 213000, People’s Republic of China
| | - Yuan Ji
- Department of Cardiology, the Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, 213000, People’s Republic of China
| | - Qingjie Wang
- Department of Cardiology, the Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, 213000, People’s Republic of China
| | - Jun Wei
- Department of Cardiovascular Surgery, the First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui, 241000, People’s Republic of China
| | - Ling Sun
- Department of Cardiology, the Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, 213000, People’s Republic of China
| |
Collapse
|
6
|
Li N, Liu H, Xue Y, Zhu Q, Fan Z. The chromatin remodeling protein BRG1 contributes to liver ischemia-reperfusion injury by regulating NOXA expression. Life Sci 2023; 334:122235. [PMID: 37926300 DOI: 10.1016/j.lfs.2023.122235] [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: 09/24/2023] [Revised: 10/27/2023] [Accepted: 10/31/2023] [Indexed: 11/07/2023]
Abstract
AIMS Hepatic ischemia-reperfusion injury (IRI) is a common complication secondary to liver transplantation. Extensive death of hepatocytes, typically in the form of apoptosis, is observed in and contributes to IRI. In the present study we investigated the role of BRG1 (encoded by Smarca4), a chromatin remodeling protein, in the pathogenesis of liver IRI focusing on the transcriptional mechanism and translational potential. METHODS Smarca4f/f mice were crossed to Alb-Cre mice to generate hepatocytes-specific BRG1 knockout mice (CKO). Alterations in cellular transcriptome were evaluated by RNA-seq. RESULTS BRG1 expression was up-regulated in liver tissues of mice subjected to I/R and in hepatocytes exposed to hypoxia-reoxygenation (H/R). Compared to wild type (WT) littermates, the BRG1 CKO mice displayed significant amelioration of liver injury following ischemia-reperfusion as evidenced by decreased ALT/AST levels and cell apoptosis. Primary hepatocytes isolated from the CKO mice were protected from H/R-induced apoptosis compared to those from the WT mice. RNA-seq analysis revealed phorbol-12-myristate-13-acetate-induced protein 1 (PMAIP1, also known as NOXA) as a novel target for BRG1. Consistently, NOXA knockdown attenuated liver IRI in mice. More importantly, administration of a small-molecule BRG1 inhibitor (PFI-3) protected the mice from liver IRI. CONCLUSIONS Our data uncover a pivotal role for BRG1 in liver IRI and suggest that targeting BRG1 with small-molecule inhibitors can be considered as a reasonable therapeutic strategy.
Collapse
Affiliation(s)
- Nan Li
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Departments of Human Anatomy and Pathophysiology, Nanjing Medial University, Nanjing, China
| | - Hong Liu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Departments of Human Anatomy and Pathophysiology, Nanjing Medial University, Nanjing, China
| | - Yujia Xue
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Departments of Human Anatomy and Pathophysiology, Nanjing Medial University, Nanjing, China
| | - Qiang Zhu
- Department of General Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China.
| | - Zhiwen Fan
- Department of Pathology, Nanjing Drum Tower Hospital, Nanjing, China.
| |
Collapse
|
7
|
Wang L, Liu Y, Tian R, Zuo W, Qian H, Wang L, Yang X, Liu Z, Zhang S. What do we know about platelets in myocardial ischemia-reperfusion injury and why is it important? Thromb Res 2023; 229:114-126. [PMID: 37437517 DOI: 10.1016/j.thromres.2023.06.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 05/22/2023] [Accepted: 06/23/2023] [Indexed: 07/14/2023]
Abstract
Myocardial ischemia-reperfusion injury (MIRI), the joint result of ischemic injury and reperfusion injury, is associated with poor outcomes in patients with acute myocardial infarction undergoing primary percutaneous coronary intervention. Accumulating evidence demonstrates that activated platelets directly contribute to the pathogenesis of MIRI through participating in the formation of microthrombi, interaction with leukocytes, secretion of active substances, constriction of microvasculature, and activation of spinal afferent nerves. The molecular mechanisms underlying the above detrimental effects of activated platelets include the homotypic and heterotypic interactions through surface receptors, transduction of intracellular signals, and secretion of active substances. Revealing the roles of platelet activation in MIRI and the associated mechanisms would provide potential targets/strategies for the clinical evaluation and treatment of MIRI. Further studies are needed to characterize the temporal (ischemia phase vs. reperfusion phase) and spatial (systemic vs. local) distributions of platelet activation in MIRI by multi-omics strategies. To improve the likelihood of translating novel cardioprotective interventions into clinical practice, basic researches maximally replicating the complexity of clinical scenarios would be necessary.
Collapse
Affiliation(s)
- Lun Wang
- Department of Internal Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| | - Yifan Liu
- Department of Internal Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| | - Ran Tian
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| | - Wei Zuo
- Department of Pharmacy, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| | - Hao Qian
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| | - Liang Wang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| | - Xinglin Yang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| | - Zhenyu Liu
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China.
| | - Shuyang Zhang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China.
| |
Collapse
|
8
|
Sikora JP, Karawani J, Sobczak J. Neutrophils and the Systemic Inflammatory Response Syndrome (SIRS). Int J Mol Sci 2023; 24:13469. [PMID: 37686271 PMCID: PMC10488036 DOI: 10.3390/ijms241713469] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 08/24/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023] Open
Abstract
We are not entirely able to understand, assess, and modulate the functioning of the immune system in clinical situations that lead to a systemic inflammatory response. In the search for diagnostic and treatment strategies (which are still far from perfect), it became very important to study the pathogenesis and participation of endogenous inflammation mediators. This study attempts to more precisely establish the role of neutrophils in individual phenomena occurring during an inflammatory and anti-inflammatory reaction, taking into account their cidal, immunoregulatory, and reparative abilities. Pro- and anticoagulatory properties of endothelium in systemic inflammatory response syndrome (SIRS) are emphasised, along with the resulting clinical implications (the application of immunotherapy using mesenchymal stem/stromal cells (MSCs) or IL-6 antagonists in sepsis and COVID-19 treatment, among others). Special attention is paid to reactive oxygen species (ROS), produced by neutrophils activated during "respiratory burst" in the course of SIRS; the protective and pathogenic role of these endogenous mediators is highlighted. Moreover, clinically useful biomarkers of SIRS (neutrophil extracellular traps, cell-free DNA, DAMP, TREMs, NGAL, miRNA, selected cytokines, ROS, and recognised markers of endothelial damage from the group of adhesins by means of immunohistochemical techniques) related to the neutrophils are presented, and their role in the diagnosing and forecasting of sepsis, burn disease, and COVID-19 is emphasised. Finally, examples of immunomodulation of sepsis and antioxidative thermal injury therapy are presented.
Collapse
Affiliation(s)
- Janusz P. Sikora
- Department of Paediatric Emergency Medicine, 2nd Chair of Paediatrics, Central Clinical Hospital, Medical University of Łódź, ul. Sporna 36/50, 91-738 Łódź, Poland;
| | - Jakub Karawani
- Faculty of Medicine, Lazarski University, ul. Świeradowska 43, 02-662 Warsaw, Poland;
| | - Jarosław Sobczak
- Department of Paediatric Emergency Medicine, 2nd Chair of Paediatrics, Central Clinical Hospital, Medical University of Łódź, ul. Sporna 36/50, 91-738 Łódź, Poland;
- Department of Management and Logistics in Healthcare, Medical University of Łódź, ul. Lindleya 6, 90-131 Łódź, Poland
| |
Collapse
|
9
|
Alaygut D, Ozturk I, Ulu S, Gungor O. NETosis and kidney disease: what do we know? Int Urol Nephrol 2023:10.1007/s11255-023-03527-y. [PMID: 36840801 DOI: 10.1007/s11255-023-03527-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 02/18/2023] [Indexed: 02/26/2023]
Abstract
Neutrophils are the most abundant leukocytes in the blood. They are rapidly mobilized from the circulation to sites of inflammation and/or infection. In affected tissues, neutrophils exhibit some dramatic antimicrobial functions, including degranulation, reactive oxygen species (ROS) production, phagocytosis, and formation of neutrophil extracellular traps (NETs). Like other cells of the immune system, after fulfilling their biological duties, they enter the path of death. Depending on the conditions, they may undergo different types of cell death (apoptosis, necrosis, necroptosis, autophagy, NETosis, and pyroptosis) that require the participation of multiple signaling pathways. NETosis is a unique neutrophil cell death mechanism that gives rise to different inflammatory and autoimmune pathological conditions. Recent studies have shown that NETosis also plays a role in the formation and/or progression of kidney diseases. This review discusses the underlying mechanism of NETosis and its relationship with some major kidney diseases in light of the current knowledge.
Collapse
Affiliation(s)
- Demet Alaygut
- Department of Pediatric Nephrology, University of Health Sciences, Izmir Faculty of Medicine, Tepecik Training and Research Hospital, Izmir, Turkey
| | - Ilyas Ozturk
- Department of Internal Medicine, Division of Nephrology, Kahramanmaras Sutcu Imam University Faculty of Medicine, Kahramanmaras, Turkey.
| | - Sena Ulu
- Department of Internal Medicine, Division of Nephrology, Bahcesehir University Faculty of Medicine, Istanbul, Turkey
| | - Ozkan Gungor
- Department of Internal Medicine, Division of Nephrology, Kahramanmaras Sutcu Imam University Faculty of Medicine, Kahramanmaras, Turkey
| |
Collapse
|
10
|
Elrod J, Kiwit A, Lenz M, Rohde H, Börnigen D, Alawi M, Mohr C, Pagerols Raluy L, Trochimiuk M, Knopf J, Reinshagen K, Herrmann M, Boettcher M. Midgut Volvulus Adds a Murine, Neutrophil-Driven Model of Septic Condition to the Experimental Toolbox. Cells 2023; 12:cells12030366. [PMID: 36766707 PMCID: PMC9913099 DOI: 10.3390/cells12030366] [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: 11/07/2022] [Revised: 01/13/2023] [Accepted: 01/14/2023] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Severe infections that culminate in sepsis are associated with high morbidity and mortality. Despite continuous efforts in basis science and clinical research, evidence based-therapy is mostly limited to basic causal and supportive measures. Adjuvant therapies often remain without clear evidence. The objective of this study was to evaluate the septic volvulus ischemia-reperfusion model in comparison to two already established models and the role of neutrophil extacellular traps (NETs) in this model. METHODS The technique of the murine model of midgut volvulus was optimized and was compared to two established models of murine sepsis, namely cecal ligation and puncture (CLP) and intra-peritoneal (i.p.) injection of lipopolysaccharide (LPS). RESULTS Midgut volvulus for 15 min caused a comparable mortality (38%) as CLP (55%) and peritoneal LPS injection (25%) at 48 h. While oxidative stress was comparable, levels of circulating free DNA (cfDNA), and splenic/hepatic and pulmonary translocation of bacteria were decreased and increased, respectively at 48 h. DNases were increased compared to the established models. Proteomic analysis revealed an upregulation of systemic Epo, IL-1b, Prdx5, Parp1, Ccl2 and IL-6 at 48 h in comparison to the healthy controls. DISCUSSION AND CONCLUSION Midgut volvulus is a stable and physiological model for sepsis. Depending on the duration and subsequent tissue damage, it represents a combination of ischemia-reperfusion injury and hyperinflammation.
Collapse
Affiliation(s)
- Julia Elrod
- Department of Pediatric Surgery, University Medical Center Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Antonia Kiwit
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Moritz Lenz
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Holger Rohde
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Daniela Börnigen
- Bioinformatics Core, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Malik Alawi
- Bioinformatics Core, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Christoph Mohr
- Department of Pediatric Surgery, University Medical Center Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Laia Pagerols Raluy
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Magdalena Trochimiuk
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Jasmin Knopf
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Ulmenweg 18, 91054 Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Ulmenweg 18, 91054 Erlangen, Germany
| | - Konrad Reinshagen
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Martin Herrmann
- Department of Pediatric Surgery, University Medical Center Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Ulmenweg 18, 91054 Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Ulmenweg 18, 91054 Erlangen, Germany
| | - Michael Boettcher
- Department of Pediatric Surgery, University Medical Center Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
- Correspondence:
| |
Collapse
|
11
|
Geerdink RJ, Pascoal Ramos MI, van den Hoogen LL, Radstake TRDJ, Shibayama S, Shibuya A, Bont L, Meyaard L. Differential isoform expression of Allergin-1 during acute and chronic inflammation. Immun Inflamm Dis 2022; 10:e739. [PMID: 36444625 PMCID: PMC9695092 DOI: 10.1002/iid3.739] [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: 05/11/2022] [Revised: 10/08/2022] [Accepted: 10/28/2022] [Indexed: 11/27/2022] Open
Abstract
INTRODUCTION Neutrophils are crucial to antimicrobial defense, but excessive neutrophilic inflammation elicits immune pathology. Currently, no effective treatment exists to curb neutrophil activation. However, neutrophils express a variety of inhibitory receptors which may represent potential therapeutic targets to limit neutrophilic inflammation. Indeed, we previously showed that the inhibitory collagen receptor leukocyte-associated immunoglobulin-like receptor 1 (LAIR-1) regulates neutrophilic airway inflammation and inhibits neutrophil extracellular trap formation. The inhibitory receptor Allergin-1 is expressed by myeloid cells and B cells. Allergin-1 suppresses mast cell and basophil activation, but a potential regulatory role on neutrophils remains unexplored. We aimed to demonstrate the regulation of neutrophils by Allergin-1. METHODS We examine Allergin-1 isoform expression on human neutrophils during homeostatic (healthy donors) and chronic inflammatory (systemic lupus erythematosus patients) conditions in comparison to other circulating leukocytes by flow cytometry. To reveal a potential role for Allergin-1 in regulating neutrophilic inflammation, we experimentally infect wild-type (WT) and Allergin-1-deficient mice with a respiratory syncytial virus (RSV) and monitor disease severity and examine cellular airway infiltrate. Flow cytometry was used to confirm Allergin-1 expression by airway-infiltrated neutrophils in RSV infection-induced bronchiolitis patients. RESULTS Only the short 1 (S1) isoform, but not the long (L) or S2 isoform could be detected on blood leukocytes, with the exception of nonclassical monocytes, which exclusively express the S2 isoform. Allergin-1 expression levels did not vary significantly between healthy individuals and patients with the systemic inflammatory disease on any interrogated cell type. Airway-infiltrated neutrophils of pediatric RSV bronchiolitis patients were found to express Allergin-1S1. However, Allergin-1-deficient mice experimentally infected with RSV did not show exacerbated disease or increased neutrophil airway infiltration compared to WT littermates. CONCLUSION Allergin-1 isoform expression is unaffected by chronic inflammatory conditions. In stark contrast to fellow inhibitory receptor LAIR-1, Allergin-1 does not regulate neutrophilic inflammation in a mouse model of RSV bronchiolitis.
Collapse
Affiliation(s)
- Ruben J. Geerdink
- Center for Translational Immunology, University Medical Centre UtrechtUtrecht UniversityUtrechtThe Netherlands
| | - Maria Inês Pascoal Ramos
- Center for Translational Immunology, University Medical Centre UtrechtUtrecht UniversityUtrechtThe Netherlands
| | - Luuk L. van den Hoogen
- Department of Rheumatology and Clinical ImmunologyUniversity Medical Centre UtrechtUtrechtThe Netherlands
| | - Timothy R. D. J. Radstake
- Center for Translational Immunology, University Medical Centre UtrechtUtrecht UniversityUtrechtThe Netherlands
| | - Shiro Shibayama
- Research Centre of Immunology, Tsukuba InstituteONO Pharmaceutical Co., Ltd.TsukubaIbarakiJapan
| | - Akira Shibuya
- Department of Immunology, Faculty of MedicineUniversity of TsukubaTsukubaIbarakiJapan
| | - Louis Bont
- Center for Translational Immunology, University Medical Centre UtrechtUtrecht UniversityUtrechtThe Netherlands
- Department of Paediatrics, Wilhelmina Children's HospitalUniversity Medical Centre UtrechtUtrechtThe Netherlands
| | - Linde Meyaard
- Center for Translational Immunology, University Medical Centre UtrechtUtrecht UniversityUtrechtThe Netherlands
| |
Collapse
|
12
|
Kuzmin DO, Manukovsky VA, Bagnenko SF, Reznik ON, Ananiev AN, Vorobyeva OA, Vorobyev SL, Gogolev DV, Daineko VS, Kutenkov AA, Chichagova NA, Uliankina IV. Use of polyclonal antibodies in brain-dead donors in kidney transplantation. RUSSIAN JOURNAL OF TRANSPLANTOLOGY AND ARTIFICIAL ORGANS 2022. [DOI: 10.15825/1995-1191-2022-4-124-134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Objective. The objective of this study is to develop a therapeutic strategy for protecting grafts in order to improve the efficiency of kidney transplantation (KT) using polyclonal antibodies (pAbs) through elimination of activated forms of neutrophils, chemo- and cytokines from the donor’s bloodstream, and a decrease in the level of expression of adhesion molecules on the renal vascular endothelium at the pre-transplant stage.Materials and methods. In 2017, we developed and for the first time applied a therapeutic strategy for ischemia-reperfusion injury (IRI) in a brain-dead donor (BDD). Given the limited time interval after brain death has been diagnosed, Timoglobulin (Sanofi Genzyme, France) was administered to the donor at a dose of 8 mg/kg intravenously for 6 hours. Before drug administration and immediately before the start of cold perfusion, a complete blood count and renal transplant biopsy were performed. The study group included 10 BDDs (mean age 39.3 ± 4.4 years) who received anti-thymocyte globulin (ATG). The comparison group included 10 BDDs (mean age 38.5 ± 4.3 years) who did not undergo the new strategy. Donor kidneys were transplanted to 40 recipients (average age 47.5 ± 4.3 years), who were also divided into 2 groups, depending on the graft received (with and without ATG). At the organ donation center, a biobank of specimens from donors of various categories, including those using the IRI therapeutic strategy and recipients for retrospective assessment of the effectiveness of pAbs, was formed.Results. Clinical blood test results show that in the ATG group, there was stable leukopenia (neutropenia and lymphopenia) of 1.46 ± 0.18x109/l. Fifteen (75%) recipients of kidneys obtained from donors with ATG had immediate graft function; in the control group – 10 (50%) recipients.Conclusion. Data obtained testify to the prospects of implementing the proposed strategy in clinical practice, which will improve the quality of the resulting grafts and their suitability for subsequent transplantation, prolong graft functioning due to elimination of leukocytes as a factor of IRI, prevention of early allograft nephropathy, increase in the donor pool by using expanded criteria donors (ECDs).
Collapse
Affiliation(s)
- D. O. Kuzmin
- St. Petersburg Research Institute of Emergency Medicine; Pavlov First St. Petersburg State Medical University
| | | | | | - O. N. Reznik
- St. Petersburg Research Institute of Emergency Medicine; Pavlov First St. Petersburg State Medical University; North-Western State Medical University
| | - A. N. Ananiev
- St. Petersburg Research Institute of Emergency Medicine; Pavlov First St. Petersburg State Medical University
| | | | | | - D. V. Gogolev
- St. Petersburg Research Institute of Emergency Medicine; Pavlov First St. Petersburg State Medical University
| | - V. S. Daineko
- St. Petersburg Research Institute of Emergency Medicine; North-Western State Medical University
| | - A. A. Kutenkov
- St. Petersburg Research Institute of Emergency Medicine; Pavlov First St. Petersburg State Medical University
| | - N. A. Chichagova
- St. Petersburg Research Institute of Emergency Medicine; Pavlov First St. Petersburg State Medical University
| | - I. V. Uliankina
- St. Petersburg Research Institute of Emergency Medicine; Pavlov First St. Petersburg State Medical University
| |
Collapse
|