1
|
Jin Z, Li X, Liu H, He T, Jiang W, Peng L, Wu X, Chen M, Fan Y, Lu Z, Fan D, Wang H. MEGF9 prevents lipopolysaccharide-induced cardiac dysfunction through activating AMPK pathway. Redox Rep 2025; 30:2435252. [PMID: 39737911 DOI: 10.1080/13510002.2024.2435252] [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] [Indexed: 01/01/2025] Open
Abstract
OBJECTIVE Inflammation and oxidative damage play critical roles in the pathogenesis of sepsis-induced cardiac dysfunction. Multiple EGF-like domains 9 (MEGF9) is essential for cell homeostasis; however, its role and mechanism in sepsis-induced cardiac injury and impairment remain unclear. METHODS Adenoviral and adeno-associated viral vectors were applied to overexpress or knock down the expression of MEGF9 in vivo and in vitro. To stimulate septic injury, cardiomyocytes and mice were treated lipopolysaccharide (LPS). To clarify the necessity of AMP-activated protein kinase (AMPK), global AMPK knockout mice were used. RESULTS We found that MEGF9 expressions were reduced in cardiomyocytes and mice by LPS stimulation. Compared with negative controls, plasma MEGF9 levels were also decreased in septic patients, and negatively correlated with LPS-induced cardiac dysfunction. In addition, MEGF9 overexpression attenuated, while MEGF9 knockdown aggravated LPS-induced inflammation and oxidative damage in vivo and in vitro, thereby regulating LPS-induced cardiac injury and impairment. Mechanistic studies revealed that MEGF9 overexpression alleviated LPS-induced cardiac dysfunction through activating AMPK pathway. CONCLUSION We for the first time demonstrate that MEGF9 prevents LPS-related inflammation, oxidative damage and cardiac injury through activating AMPK pathway, and provide a proof-of-concept for the treatment of LPS-induced cardiac dysfunction by targeting MEGF9.
Collapse
Affiliation(s)
- Zhili Jin
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
- Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, People's Republic of China
| | - Xianqing Li
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
- Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, People's Republic of China
| | - Huixia Liu
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
- Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, People's Republic of China
| | - Tao He
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
- Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, People's Republic of China
| | - Wanli Jiang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Li Peng
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
- Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, People's Republic of China
| | - Xiaoyan Wu
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
- Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, People's Republic of China
| | - Ming Chen
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
- Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, People's Republic of China
| | - Yongzhen Fan
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
- Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, People's Republic of China
| | - Zhibing Lu
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
- Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, People's Republic of China
| | - Di Fan
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
- Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, People's Republic of China
| | - Hairong Wang
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
- Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, People's Republic of China
| |
Collapse
|
2
|
Chen Y, Yang T, Wang S, Tong D, Liu X, Li Y, Zhao W, Zhao C. Hemocompatible nucleosome-inspired heparin-mimicking hydrogel microspheres for safe and efficient extracorporeal removal of circulating histones in critically ill patients. J Mater Chem B 2025; 13:2366-2381. [PMID: 39820716 DOI: 10.1039/d4tb01952k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Circulating histones have been identified as essential mediators that lead to hyperinflammation, platelet aggregation, coagulation cascade activation, endothelial cell injury, multiple organ dysfunction, and death in severe patients with sepsis, multiple trauma, COVID-19, acute liver failure, and pancreatitis. Clinical evidence suggests that plasma levels of circulating histones are positively associated with disease severity and survival in patients with such critical diseases. However, safe and efficient therapeutic strategies targeting circulating histones are lacking in current clinical practice. Extracorporeal blood purification, a widely used life support technique in intensive care units, is a promising therapeutic option for eliminating circulating histones. Inspired by electrostatic interactions between DNA chains and histones in natural nucleosomes, we propose a "one stone kills two birds" strategy to combat histone-related critical diseases by developing heparin-mimicking hydrogel microspheres (RCHMs). On one hand, the heparin-mimicking hydrogel structure inside RCHMs contains a large number of carboxyl and sulphonic acid groups by in situ cross-linking polymerization, which endows the RCHMs with excellent hemocompatibility. On the other hand, the RCHMs can adsorb circulating histones through electrostatic interactions. Our results demonstrate that the RCHMs do not cause significant hemolysis, blood cell activation and complement activation, with improved anti-protein contamination properties. The tailored RCHM microspheres (A3M1) can efficiently and selectively adsorb 91.16% of calf thymus histones with an adsorption capacity of 20.47 μg mg-1 within 4 h. Moreover, the RCHMs significantly attenuate histone-mediated thrombocytopenia, platelet aggregation, and endothelial cell death. Therefore, the RCHMs are promising hemoperfusion adsorbents for extracorporeal removal of circulating histones from the blood of critically ill patients, providing a new insight into the management of multiple histone-related disorders.
Collapse
Affiliation(s)
- Yu Chen
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China.
| | - Tinghang Yang
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu 610041, China.
| | - Shujing Wang
- Department of Nephrology, Kidney Research Institute, Frontiers Science Centre for Disease-related Molecular Network, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Dongmei Tong
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China.
| | - Xianda Liu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China.
| | - Yupei Li
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu 610041, China.
| | - Weifeng Zhao
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China.
| | - Changsheng Zhao
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China.
| |
Collapse
|
3
|
Marsman G, Zheng X, Čerina D, Lacey KA, Liu M, Humme D, Goosmann C, Brinkmann V, Harbort CJ, Torres VJ, Zychlinsky A. Histone H1 kills MRSA. Cell Rep 2024; 43:114969. [PMID: 39546397 DOI: 10.1016/j.celrep.2024.114969] [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: 03/11/2024] [Revised: 10/06/2024] [Accepted: 10/23/2024] [Indexed: 11/17/2024] Open
Abstract
The antimicrobial activity of histones was discovered in the 1940s, but their mechanism of action is not fully known. Here we show that methicillin-resistant Staphylococcus aureus (MRSA) is susceptible to histone H1 (H1), even in the presence of divalent cations and serum. Through selective evolution and a genome-wide screen of a transposon library, as well as physiological and pharmacological experiments, we elucidated how H1 kills MRSA. We show that H1 first binds to wall teichoic acids with high affinity. Once bound, H1 requires a potentiated membrane and a metabolically active bacterium to permeabilize the membrane and enter the cell. Upon entry, H1 accumulates intracellularly, in close association with the bacterial DNA. Of note, anti-H1 antibodies inhibit neutrophil extracellular trap killing of MRSA. Moreover, H1 colocalizes with bacterial DNA in abscess samples of MRSA-infected patients, suggesting a role for H1 in combating MRSA in vivo.
Collapse
Affiliation(s)
- Gerben Marsman
- Department of Cellular Microbiology, Max Planck Institute for Infection Biology, Charitéplatz 1, 10117 Berlin, Germany
| | - Xuhui Zheng
- Department of Microbiology, New York University Grossman School of Medicine, 430 East 29th Street, New York, NY 10016, USA
| | - Dora Čerina
- Department of Cellular Microbiology, Max Planck Institute for Infection Biology, Charitéplatz 1, 10117 Berlin, Germany
| | - Keenan A Lacey
- Department of Microbiology, New York University Grossman School of Medicine, 430 East 29th Street, New York, NY 10016, USA
| | - Menghan Liu
- Department of Microbiology, New York University Grossman School of Medicine, 430 East 29th Street, New York, NY 10016, USA
| | - Daniel Humme
- Department of Dermatology, Venerology and Allergology, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Christian Goosmann
- Department of Cellular Microbiology, Max Planck Institute for Infection Biology, Charitéplatz 1, 10117 Berlin, Germany
| | - Volker Brinkmann
- Department of Cellular Microbiology, Max Planck Institute for Infection Biology, Charitéplatz 1, 10117 Berlin, Germany
| | - C J Harbort
- Department of Cellular Microbiology, Max Planck Institute for Infection Biology, Charitéplatz 1, 10117 Berlin, Germany
| | - Victor J Torres
- Department of Microbiology, New York University Grossman School of Medicine, 430 East 29th Street, New York, NY 10016, USA; Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA.
| | - Arturo Zychlinsky
- Department of Cellular Microbiology, Max Planck Institute for Infection Biology, Charitéplatz 1, 10117 Berlin, Germany.
| |
Collapse
|
4
|
Wu J, Ai T, He P, Shi Q, Li Y, Zhang Z, Chen M, Huang Z, Wu S, Chen W, Han J. Cecal necroptosis triggers lethal cardiac dysfunction in TNF-induced severe SIRS. Cell Rep 2024; 43:114778. [PMID: 39325617 DOI: 10.1016/j.celrep.2024.114778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 07/25/2024] [Accepted: 09/04/2024] [Indexed: 09/28/2024] Open
Abstract
Tumor necrosis factor (TNF) induces systemic inflammatory response syndrome (SIRS), and severe SIRS can serve as a model for studying animal death caused by organ failure. Through strategic cecectomy, we demonstrate that necroptosis in the cecum initiates the death process in TNF-treated mice, but it is not the direct cause of death. Instead, we show that it is the cardiac dysfunction downstream of cecum damage that ultimately leads to the death of TNF-treated mice. By in vivo and ex vivo physiological analyses, we reveal that TNF and the damage-associated molecular patterns (DAMPs) released from necroptotic cecal cells jointly target cardiac endothelial cells, triggering caspase-8 activation and subsequent cardiac endothelial damage. Cardiac endothelial damage is a primary cause of the deterioration of diastolic function in the heart of TNF-treated mice. Our research provides insights into the pathophysiological process of TNF-induced lethality.
Collapse
Affiliation(s)
- Jianfeng Wu
- State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian 361102, China; Laboratory Animal Research Center, Xiamen University, Xiamen, Fujian 361102, China
| | - Tingting Ai
- State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian 361102, China
| | - Peng He
- State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian 361102, China; Research Unit of Cellular Stress of Chinese Academy of Medical Sciences, Xiang'an Hospital of Xiamen University, Xiamen, Fujian 361102, China
| | - Qilin Shi
- State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian 361102, China
| | - Yangxin Li
- State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian 361102, China
| | - Ziguan Zhang
- Xiamen Key Laboratory of Cardiac Electrophysiology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361102, China
| | - Minwei Chen
- Xiamen Key Laboratory of Cardiac Electrophysiology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361102, China
| | - Zhengrong Huang
- Xiamen Key Laboratory of Cardiac Electrophysiology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361102, China
| | - Suqin Wu
- Laboratory Animal Research Center, Xiamen University, Xiamen, Fujian 361102, China
| | - Wanze Chen
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Advanced Technology, Shenzhen, Guangdong 518000, China
| | - Jiahuai Han
- State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian 361102, China; Laboratory Animal Research Center, Xiamen University, Xiamen, Fujian 361102, China; Research Unit of Cellular Stress of Chinese Academy of Medical Sciences, Xiang'an Hospital of Xiamen University, Xiamen, Fujian 361102, China.
| |
Collapse
|
5
|
Li Y, Sun Y, Zhang X, Wang D, Yang X, Wei H, Wang C, Shi Z, Li X, Zhang F, Sun W, Yang Z, Song Y, Qing G. Selective Clearance of Circulating Histones Based on Dodecapeptide-Grafted Copolymer Material for Sepsis Blood Purification. ACS APPLIED MATERIALS & INTERFACES 2024; 16:47110-47123. [PMID: 39189050 DOI: 10.1021/acsami.4c07494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
Sepsis is defined as a life-threatening organ dysfunction caused by a dysregulated host response to infection. Research indicates that circulating histones, as pathogenic factors, may represent a therapeutic target for sepsis. However, effectively clearing circulating histones poses a challenge due to their structural similarity to normal blood proteins, their low abundance in the bloodstream, and serious interference from other blood biomacromolecules. Here we design a dodecapeptide-based functional polymer that can selectively adsorb circulating histones from the blood. The peptide, named P1 (HNHHQLALVESY), was discovered through phage display screening and demonstrated a strong affinity for circulating histones while exhibiting negligible affinities for common proteins in the blood, such as human serum albumin (HSA), immunoglobulin G (IgG), and transferrin (TRF). Furthermore, the P1 peptide was incorporated into a functional polymer design, poly(PEGMA-co-P1), which was immobilized onto a silica gel surface through reversible addition-fragmentation chain transfer polymerization. The resulting material was characterized using solid nuclear magnetic resonance, thermogravimetric analysis, and X-ray photoelectron spectroscopy. This material demonstrated the ability to selectively and efficiently capture circulating histones from both model solutions and whole blood samples while also exhibiting satisfactory blood compatibility, good antifouling properties, and resistance to interference. Satisfactory binding affinity and efficient capture capacity toward histone were also observed for the other screened peptide P2 (QMSMDLFGSNFV)-grafted polymer, validating phage display as a reliable ligand screening strategy. These findings present an approach for the specific clearance of circulating histones and hold promise for future clinical applications in blood purification toward sepsis.
Collapse
Affiliation(s)
- Yan Li
- College of Pharmaceutical and Bioengineering, Shenyang University of Chemical Technology, Shenyang 110142, P. R. China
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, P. R. China
| | - Yue Sun
- College of Pharmaceutical and Bioengineering, Shenyang University of Chemical Technology, Shenyang 110142, P. R. China
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, P. R. China
| | - Xiaoyu Zhang
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, P. R. China
| | - Dongdong Wang
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, P. R. China
| | - Xindi Yang
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, P. R. China
| | - Haijie Wei
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Cunli Wang
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, P. R. China
| | - Zhenqiang Shi
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, P. R. China
| | - Xiaonan Li
- Department of Spine Surgery, First Affiliated Hospital of Dalian Medical University, Dalian 116011, P. R. China
| | - Fenglin Zhang
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, P. R. China
- Department of Spine Surgery, First Affiliated Hospital of Dalian Medical University, Dalian 116011, P. R. China
| | - Wenjing Sun
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, P. R. China
| | - Zhiying Yang
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Yanling Song
- College of Pharmaceutical and Bioengineering, Shenyang University of Chemical Technology, Shenyang 110142, P. R. China
| | - Guangyan Qing
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| |
Collapse
|
6
|
Keulen GM, Huckriede J, Wichapong K, Nicolaes GAF. Histon activities in the extracellular environment: regulation and prothrombotic implications. Curr Opin Hematol 2024; 31:230-237. [PMID: 39087372 PMCID: PMC11296287 DOI: 10.1097/moh.0000000000000827] [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] [Indexed: 08/02/2024]
Abstract
PURPOSE OF REVIEW Thromboembolic complications are a major contributor to global mortality. The relationship between inflammation and coagulation pathways has become an emerging research topic where the role of the innate immune response, and specifically neutrophils in "immunothrombosis" are receiving much attention. This review aims to dissect the intricate interplay between histones (from neutrophils or cellular damage) and the haemostatic pathway, and to explore mechanisms that may counteract the potentially procoagulant effects of those histones that have escaped their nuclear localization. RECENT FINDINGS Extracellular histones exert procoagulant effects via endothelial damage, platelet activation, and direct interaction with coagulation proteins. Neutralization of histone activities can be achieved by complexation with physiological molecules, through pharmacological compounds, or via proteolytic degradation. Details of neutralization of extracellular histones are still being studied. SUMMARY Leveraging the understanding of extracellular histone neutralization will pave the way for development of novel pharmacological interventions to treat and prevent complications, including thromboembolism, in patients in whom extracellular histones contribute to their overall clinical status.
Collapse
Affiliation(s)
- Gwen M Keulen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, The Netherlands
| | | | | | | |
Collapse
|
7
|
Toh JM, Yong J, Abrams ST, Wang L, Schofield J, Lane S, La Corte AC, Wang SS, Ariëns RAS, Philippou H, Xie J, Yu W, Wang G, Toh CH. Fibrinogen binding to histones in circulation protects against adverse cellular and clinical outcomes. J Thromb Haemost 2024; 22:2247-2260. [PMID: 38777257 DOI: 10.1016/j.jtha.2024.05.009] [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: 12/08/2023] [Revised: 04/17/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND Circulating histones are released by extensive tissue injury or cell death and play important pathogenic roles in critical illnesses. Their interaction with circulating plasma components and the potential roles in the clinical setting are not fully understood. OBJECTIVES We aimed to characterize the interaction of histones with fibrinogen and explore its roles in vitro, in vivo, and in patient samples. METHODS Histone-fibrinogen binding was assessed by electrophoresis and enzyme-linked immunosorbent assay-based affinity assay. Functional significance was explored using washed platelets and endothelial cells in vitro and histone-infusion mouse models in vivo. To determine clinical translatability, a retrospective single-center cohort study was conducted on patients requiring intensive care admission (n = 199) and validated in a cohort of hospitalized patients with COVID-19 (n = 69). RESULTS Fibrinogen binds histones through its D-domain with high affinity (calf thymus histones, KD = 18.0 ± 5.6 nM; histone 3, KD = 2.7 ± 0.8 nM; and histone 4, KD = 2.0 ± 0.7 nM) and significantly reduces histone-induced endothelial damage and platelet aggregation in vitro and in vivo in a histone-infusion mouse model. Physiologic concentrations of fibrinogen can neutralize low levels of circulating histones and increase the cytotoxicity threshold of histones to 50 μg/mL. In a cohort of patients requiring intensive care, a histone:fibrinogen ratio of ≥6 on admission was associated with moderate-severe thrombocytopenia and independently predicted mortality. This finding was validated in a cohort of hospitalized patients with COVID-19. CONCLUSION Fibrinogen buffers the cytotoxic properties of circulating histones. Detection and monitoring of circulating histones and histone:fibrinogen ratios will help identify critically ill patients at highest risk of adverse outcomes who might benefit from antihistone therapy.
Collapse
Affiliation(s)
- Julien M Toh
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, UK; Department of Anaesthetics and Critical Care, St George's University Hospitals NHS Foundation Trust, London, UK
| | - Jun Yong
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, UK; Roald Dahl Haemostasis & Thrombosis Centre, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
| | - Simon T Abrams
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, UK; Department of Haematology, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
| | - Lijun Wang
- The Medical School, Southeast University, Nanjing, China
| | - Jeremy Schofield
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, UK; Roald Dahl Haemostasis & Thrombosis Centre, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
| | - Steven Lane
- Department of Medical Statistics, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Amy Cilia La Corte
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Susan S Wang
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, UK; William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, Charterhouse Square, London, UK
| | - Robert A S Ariëns
- Leeds Institute of Cardiovascular & Metabolic Medicine, University of Leeds, Leeds, West Yorkshire, UK
| | - Helen Philippou
- Leeds Institute of Cardiovascular & Metabolic Medicine, University of Leeds, Leeds, West Yorkshire, UK
| | - Jianfeng Xie
- The Medical School, Southeast University, Nanjing, China
| | - Weiping Yu
- The Medical School, Southeast University, Nanjing, China
| | - Guozheng Wang
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, UK; Department of Haematology, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK.
| | - Cheng-Hock Toh
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, UK; Roald Dahl Haemostasis & Thrombosis Centre, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK.
| |
Collapse
|
8
|
杨 汀, 李 育, 苏 白. [Mechanism of Extracellular Histone-Induced Endothelial Dysfunction Leading to Sepsis-Induced Acute Respiratory Distress Syndrome]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2024; 55:902-910. [PMID: 39170019 PMCID: PMC11334276 DOI: 10.12182/20240760508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Indexed: 08/23/2024]
Abstract
Objective Sepsis-induced acute respiratory distress syndrome (ARDS) is an independent risk factor for mortality in critically ill septic patients. However, effective therapeutic targets are still unavailable due to the lack of understanding of its unclear pathogenesis. With increasing understanding in the roles of circulating histones and endothelial dysfunction in sepsis, we aimed to investigate the mechanism of histone-induced endothelial dysfunction leading to sepsis-induced ARDS and to provide experimental support for histone-targeted treatment of sepsis-induced ARDS. Methods First of all, in vitro experiments were conducted. Human umbilical vein endothelial cells (HUVEC) were stimulated with gradient concentrations of histones to explore for the optimal stimulation concentration in vitro. Then, HUVEC were exposed to histones at an optimal concentration with or without resatorvid (TAK-242), a selective inhibitor of Toll-like receptor 4 (TLR4), for 24 hours for modeling. The cells were divided into 4 groups: 1) the blank control group, 2) the blank control+TAK-242 intervention group, 3) the histone stimulation group, and 4) the histone+TAK-242 intervention group. HUVEC apoptosis was determined by flow cytometry, VE-Cadherin expression in endothelial cells was determined by Western blot, and the integrity of adhesion connections between endothelial cells was evaluated with confocal fluorescence microscopic images. Male C57BL/6 mice aged 6-8 weeks and weighing 22-25 g were used for the in vivo experiment. Then, the mice were given cecal ligation and puncture (CLP) as well as histone injection at 50 mg/kg via the tail vein for sepsis modeling. The experimental animals were divided into 6 groups: 1) the blank control group, 2) the blank control+TAK-242 intervention group, 3) the CLP model group, 4) the CLP+TAK-242 intervention group, 5) the histone model group, and 6) the histone+TAK-242 intervention group. After 24 h, the concentrations of serum interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) were determined using ELISA kits. Western blot was performed to determine the expression of vascular endothelial (VE)-cadherin in the lung tissue. Hematoxylin and eosin (HE) staining was performed to observe the pathological changes in the lung tissue of the mice. Evans Blue was injected via the tail vein 30 min before the mice were sacrificed. Lung tissue was collected after the mice were sacrificed. Then, the concentrations of Evans blue dye per unit mass in the lung tissue from mice of different groups were evaluated, the rates of pulmonary endothelial leakage were calculated, and the integrity of the pulmonary endothelial barrier was evaluated. Results The results of the in vitro experiment showed that, compared with those of the control group, HUVEC apoptosis was significantly increased under histone stimulation (P<0.05), the expression of VE-cadherin was decreased (P<0.05), and the integrity of adherens junctions between endothelial cells was damaged. TAK-242 can significantly inhibit histone-induced HUVEC apoptosis and VE-cadherin expression reduction and maintain the integrity of adherens junctions between endothelial cells. According to the findings from the in vivo experiments, in mice with CLP-induced and histone-induced sepsis, TAK-242 effectively alleviated the increase in serum concentrations of IL-6 and TNF-α, reduced the downregulation of VE-cadherin expression in the lung tissue (P<0.05), decreased endothelial permeability of the lung vessels, and improved pathological injury in the lung tissue. Conclusion By binding to TLR-4, histone decreases VE-cadherin expression on the surface of vascular endothelial cells, disrupts the integrity of intercellular adherens junctions, and triggers pathological damage to lung tissue. Using TLR-4 inhibitors can prevent sepsis-induced ARDS in histone-induced sepsis.
Collapse
Affiliation(s)
- 汀航 杨
- 四川大学华西医院 肾脏内科/肾脏病研究所 (成都 610041)Department of Nephrology/Kidney Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| | - 育霈 李
- 四川大学华西医院 肾脏内科/肾脏病研究所 (成都 610041)Department of Nephrology/Kidney Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| | - 白海 苏
- 四川大学华西医院 肾脏内科/肾脏病研究所 (成都 610041)Department of Nephrology/Kidney Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
9
|
McRae SA, Richards CM, Da Silva DE, Riar I, Yang SS, Zurfluh NE, Gibon J, Klegeris A. Pro-neuroinflammatory and neurotoxic potential of extracellular histones H1 and H3. Neurosci Res 2024; 204:34-45. [PMID: 38278218 DOI: 10.1016/j.neures.2024.01.004] [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: 05/17/2023] [Revised: 12/23/2023] [Accepted: 01/17/2024] [Indexed: 01/28/2024]
Abstract
Histones organize DNA within cellular nuclei, but they can be released from damaged cells. In peripheral tissues extracellular histones act as damage-associated molecular patterns (DAMPs) inducing pro-inflammatory activation of immune cells. Limited studies have considered DAMP-like activity of histones in the central nervous system (CNS); therefore, we studied the effects of extracellular histones on microglia, the CNS immunocytes, and on neuronal cells. Both the linker histone H1 and the core histone H3 induced pro-inflammatory activation of microglia-like cells by upregulating their secretion of NO and cytokines, including interferon-γ-inducible protein 10 (IP-10) and tumor necrosis factor-α (TNF). The selective inhibitors MMG-11 and TAK-242 were used to demonstrate involvement of toll-like receptors (TLR) 2 and 4, respectively, in H1-induced NO secretion by BV-2 microglia. H1, but not H3, downregulated the phagocytic activity of BV-2 microglia. H1 was also directly toxic to all neuronal cell types studied. We conclude that H1, and to a lesser extent H3, when released extracellularly, have the potential to act as a CNS DAMPs. Inhibition of the DAMP-like effects of extracellular histones on microglia and their neurotoxic activity represents a potential strategy for combating neurodegenerative diseases that are characterized by the adverse activation of microglia and neuronal death.
Collapse
Affiliation(s)
- Seamus A McRae
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, BC V1V 1V7, Canada
| | - Christy M Richards
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, BC V1V 1V7, Canada
| | - Dylan E Da Silva
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, BC V1V 1V7, Canada
| | - Ishvin Riar
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, BC V1V 1V7, Canada
| | - Sijie Shirley Yang
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, BC V1V 1V7, Canada
| | - Noah E Zurfluh
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, BC V1V 1V7, Canada
| | - Julien Gibon
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, BC V1V 1V7, Canada
| | - Andis Klegeris
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, BC V1V 1V7, Canada.
| |
Collapse
|
10
|
Affiliation(s)
- Guozheng Wang
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, United Kingdom
- Department of Haematology, Liverpool University Hospitals NHS Foundation Trust, Liverpool, United Kingdom
| | - Simon Timothy Abrams
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, United Kingdom
- Department of Haematology, Liverpool University Hospitals NHS Foundation Trust, Liverpool, United Kingdom
| | - Cheng-Hock Toh
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, United Kingdom
- Roald Dahl Haemostasis & Thrombosis Centre, Liverpool University Hospitals NHS Foundation Trust, Liverpool, United Kingdom
| |
Collapse
|
11
|
Hiraiwa H, Kasugai D, Okumura T, Murohara T. Clinical implications of septic cardiomyopathy: A narrative review. Medicine (Baltimore) 2024; 103:e37940. [PMID: 38669408 PMCID: PMC11049701 DOI: 10.1097/md.0000000000037940] [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: 10/31/2023] [Accepted: 03/29/2024] [Indexed: 04/28/2024] Open
Abstract
Sepsis is caused by the body's dysregulated response to infection, which can lead to multiorgan injury and death. Patients with sepsis may develop acute cardiac dysfunction, termed septic cardiomyopathy, which is a global but reversible dysfunction of both sides of the heart. This narrative review discusses the mechanistic changes in the heart during septic cardiomyopathy, its diagnosis, existing treatment options regarding severity and course, and emerging treatment approaches. Although no standardized definition for septic cardiomyopathy exists, it is described as a reversible myocardial dysfunction that typically resolves within 7 to 10 days. Septic cardiomyopathy is often diagnosed based on electrocardiography, cardiac magnetic resonance imaging, biomarkers, and direct invasive and noninvasive measures of cardiac output. Presently, the treatment of septic cardiomyopathy is similar to that of sepsis, primarily focusing on acute interventions. Treatments for cardiomyopathy often include angiotensin-converting enzyme inhibitors, angiotensin receptor blockers, and diuretics. However, because of profound hypotension in sepsis, many cardiomyopathy treatments are contraindicated in patients with septic cardiomyopathy. Substantial efforts have been made to study the pathophysiological mechanisms and diagnostic options; however, the lack of a uniform definition for septic cardiomyopathy is challenging for physicians when considering treatments. Another challenge for physicians is that the treatment for septic cardiomyopathy has only focused on acute intervention, whereas the treatment for other cardiomyopathies has been provided on a long-term basis. A better understanding of the underlying mechanisms of septic cardiomyopathy may contribute to the development of a unified definition of the condition and novel treatment options.
Collapse
Affiliation(s)
- Hiroaki Hiraiwa
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Daisuke Kasugai
- Department of Emergency and Critical Care Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takahiro Okumura
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
12
|
Ferreira GS, Frota ML, Gonzaga MJD, Vattimo MDFF, Lima C. The Role of Biomarkers in Diagnosis of Sepsis and Acute Kidney Injury. Biomedicines 2024; 12:931. [PMID: 38790893 PMCID: PMC11118225 DOI: 10.3390/biomedicines12050931] [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: 03/06/2024] [Revised: 04/05/2024] [Accepted: 04/13/2024] [Indexed: 05/26/2024] Open
Abstract
Sepsis and acute kidney injury (AKI) are two major public health concerns that contribute significantly to illness and death worldwide. Early diagnosis and prompt treatment are essential for achieving the best possible outcomes. To date, there are no specific clinical, imaging, or biochemical indicators available to diagnose sepsis, and diagnosis of AKI based on the KDIGO criterion has limitations. To improve the diagnostic process for sepsis and AKI, it is essential to continually evolve our understanding of these conditions. Delays in diagnosis and appropriate treatment can have serious consequences. Sepsis and AKI often occur together, and patients with kidney dysfunction are more prone to developing sepsis. Therefore, identifying potential biomarkers for both conditions is crucial. In this review, we talk about the main biomarkers that evolve the diagnostic of sepsis and AKI, namely neutrophil gelatinase-associated lipocalin (NGAL), proenkephalin (PENK), and cell-free DNA.
Collapse
Affiliation(s)
| | | | | | | | - Camila Lima
- Department of Medical-Surgical Nursing, School of Nursing, University of São Paulo, São Paulo 05403-000, Brazil; (G.S.F.); (M.L.F.); (M.J.D.G.); (M.d.F.F.V.)
| |
Collapse
|
13
|
Da Silva DE, Richards CM, McRae SA, Riar I, Yang S(S, Zurfluh NE, Gibon J, Klegeris A. Extracellular mixed histones are neurotoxic and modulate select neuroimmune responses of glial cells. PLoS One 2024; 19:e0298748. [PMID: 38630734 PMCID: PMC11023449 DOI: 10.1371/journal.pone.0298748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 01/29/2024] [Indexed: 04/19/2024] Open
Abstract
Although histone proteins are widely known for their intranuclear functions where they organize DNA, all five histone types can also be released into the extracellular space from damaged cells. Extracellular histones can interact with pattern recognition receptors of peripheral immune cells, including toll-like receptor 4 (TLR4), causing pro-inflammatory activation, which indicates they may act as damage-associated molecular patterns (DAMPs) in peripheral tissues. Very limited information is available about functions of extracellular histones in the central nervous system (CNS). To address this knowledge gap, we applied mixed histones (MH) to cultured cells modeling neurons, microglia, and astrocytes. Microglia are the professional CNS immunocytes, while astrocytes are the main support cells for neurons. Both these cell types are critical for neuroimmune responses and their dysregulated activity contributes to neurodegenerative diseases. We measured effects of extracellular MH on cell viability and select neuroimmune functions of microglia and astrocytes. MH were toxic to cultured primary murine neurons and also reduced viability of NSC-34 murine and SH-SY5Y human neuron-like cells in TLR4-dependent manner. MH did not affect the viability of resting or immune-stimulated BV-2 murine microglia or U118 MG human astrocytic cells. When applied to BV-2 cells, MH enhanced secretion of the potential neurotoxin glutamate, but did not modulate the release of nitric oxide (NO), tumor necrosis factor-α (TNF), C-X-C motif chemokine ligand 10 (CXCL10), or the overall cytotoxicity of lipopolysaccharide (LPS)- and/or interferon (IFN)-γ-stimulated BV-2 microglial cells towards NSC-34 neuron-like cells. We demonstrated, for the first time, that MH downregulated phagocytic activity of LPS-stimulated BV-2 microglia. However, MH also exhibited protective effect by ameliorating the cytotoxicity of LPS-stimulated U118 MG astrocytic cells towards SH-SY5Y neuron-like cells. Our data demonstrate extracellular MH could both damage neurons and alter neuroimmune functions of glial cells. These actions of MH could be targeted for treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Dylan E. Da Silva
- Department of Biology, University of British Columbia Okanagan Campus, University Way, Kelowna, British Columbia, Canada
| | - Christy M. Richards
- Department of Biology, University of British Columbia Okanagan Campus, University Way, Kelowna, British Columbia, Canada
| | - Seamus A. McRae
- Department of Biology, University of British Columbia Okanagan Campus, University Way, Kelowna, British Columbia, Canada
| | - Ishvin Riar
- Department of Biology, University of British Columbia Okanagan Campus, University Way, Kelowna, British Columbia, Canada
| | - Sijie (Shirley) Yang
- Department of Biology, University of British Columbia Okanagan Campus, University Way, Kelowna, British Columbia, Canada
| | - Noah E. Zurfluh
- Department of Biology, University of British Columbia Okanagan Campus, University Way, Kelowna, British Columbia, Canada
| | - Julien Gibon
- Department of Biology, University of British Columbia Okanagan Campus, University Way, Kelowna, British Columbia, Canada
| | - Andis Klegeris
- Department of Biology, University of British Columbia Okanagan Campus, University Way, Kelowna, British Columbia, Canada
| |
Collapse
|
14
|
Hegemann N, Barth L, Döring Y, Voigt N, Grune J. Implications for neutrophils in cardiac arrhythmias. Am J Physiol Heart Circ Physiol 2024; 326:H441-H458. [PMID: 38099844 PMCID: PMC11219058 DOI: 10.1152/ajpheart.00590.2023] [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: 09/22/2023] [Revised: 12/08/2023] [Accepted: 12/13/2023] [Indexed: 02/03/2024]
Abstract
Cardiac arrhythmias commonly occur as a result of aberrant electrical impulse formation or conduction in the myocardium. Frequently discussed triggers include underlying heart diseases such as myocardial ischemia, electrolyte imbalances, or genetic anomalies of ion channels involved in the tightly regulated cardiac action potential. Recently, the role of innate immune cells in the onset of arrhythmic events has been highlighted in numerous studies, correlating leukocyte expansion in the myocardium to increased arrhythmic burden. Here, we aim to call attention to the role of neutrophils in the pathogenesis of cardiac arrhythmias and their expansion during myocardial ischemia and infectious disease manifestation. In addition, we will elucidate molecular mechanisms associated with neutrophil activation and discuss their involvement as direct mediators of arrhythmogenicity.
Collapse
Affiliation(s)
- Niklas Hegemann
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), Berlin, Germany
| | - Lukas Barth
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), Berlin, Germany
| | - Yannic Döring
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg August University Göttingen, Göttingen, Germany
- German Centre for Cardiovascular Research (DZHK), Göttingen, Germany
| | - Niels Voigt
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg August University Göttingen, Göttingen, Germany
- German Centre for Cardiovascular Research (DZHK), Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Jana Grune
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), Berlin, Germany
| |
Collapse
|
15
|
Shahriari S, Damodara S, Selvaganapathy PR. Isoelectric trapping and discrimination of histones from plasma in a microfluidic device using dehydrated isoelectric gate. Mikrochim Acta 2024; 191:131. [PMID: 38351209 DOI: 10.1007/s00604-024-06223-5] [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: 09/20/2023] [Accepted: 01/19/2024] [Indexed: 02/16/2024]
Abstract
Histones are basic proteins with an isoelectric point around 11. It has been shown that the level of plasma circulating histones increases significantly during sepsis, and circulating free histones are associated with sepsis severity and mortality. It was found that the median plasma total free histone concentration of sepsis ICU non-survivors is higher compared to survivors. Therefore, histone concentration can serve as a prognostic indicator and there is a need for a simple, low-cost, and rapid method for measuring histone levels. In this work, we have developed a microfluidic device containing an isoelectric membrane made of dehydrated agarose gel of a specific pH embedded in a porous membrane for isoelectric trapping of histones rapidly. Although isoelectric gates have been used for trapping proteins before, they have to be introduced at the time of the experiment. Here, we show that isoelectric gates formed by gels loaded in a scaffold can be integrated directly into the fabrication process flow, dehydrated for storage, and rehydrated during the experiment and still function effectively to achieve isoelectric trapping. A low-cost and rapid microfabrication technique, xurography, was used for agarose integration and device fabrication. The integrated device was tested with samples containing buffered histone, histone in the presence of high-concentration bovine serum albumin (BSA), and histone spiked in blood plasma. The results show that the device can be used to distinguish between survivors and non-survivors of sepsis in less than 10 min, making it suitable as a point-of-care device for sepsis prognosis.
Collapse
Affiliation(s)
- Shadi Shahriari
- Department of Mechanical Engineering, McMaster University, Hamilton, ON, Canada
| | - Sreekant Damodara
- Department of Mechanical Engineering, McMaster University, Hamilton, ON, Canada
| | - P Ravi Selvaganapathy
- Department of Mechanical Engineering, McMaster University, Hamilton, ON, Canada.
- School of Biomedical Engineering, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
16
|
Reutelingsperger CPM, Gijbels MJ, Spronk H, Van Oerle R, Schrijver R, Ekhart P, de Kimpe S, Nicolaes GAF. M6229 Protects against Extracellular-Histone-Induced Liver Injury, Kidney Dysfunction, and Mortality in a Rat Model of Acute Hyperinflammation. Int J Mol Sci 2024; 25:1376. [PMID: 38338654 PMCID: PMC10855969 DOI: 10.3390/ijms25031376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/16/2024] [Accepted: 01/18/2024] [Indexed: 02/12/2024] Open
Abstract
Extracellular histones have been shown to act as DAMPs in a variety of inflammatory diseases. Moreover, they have the ability to induce cell death. In this study, we show that M6229, a low-anticoagulant fraction of unfractionated heparin (UFH), rescues rats that were challenged by continuous infusion of calf thymus histones at a rate of 25 mg histones/kg/h. Histone infusion by itself induced hepatic and homeostatic dysfunction characterized by elevated activity of hepatic enzymes (ASAT and ALAT) and serum lactate levels as well as by a renal dysfunction, which contributed to the significantly increased mortality rate. M6229 was able to restore normal levels of both hepatic and renal parameters at 3 and 9 mg M6229/kg/h and prevented mortality of the animals. We conclude that M6229 is a promising therapeutic agent to treat histone-mediated disease.
Collapse
Affiliation(s)
- Chris P. M. Reutelingsperger
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6200 MD Maastricht, The Netherlands; (C.P.M.R.); (H.S.); (R.V.O.)
| | - Marion J. Gijbels
- Department of Pathology, Maastricht University Medical Center, MUMC+, 6202 AZ Maastricht, The Netherlands;
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences—Atherosclerosis & Ischemic Syndrome, Amsterdam Infection and Immunity—Inflammatory Diseases, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Henri Spronk
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6200 MD Maastricht, The Netherlands; (C.P.M.R.); (H.S.); (R.V.O.)
- Coagulation Profile B.V., 6229 EV Maastricht, The Netherlands
| | - Rene Van Oerle
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6200 MD Maastricht, The Netherlands; (C.P.M.R.); (H.S.); (R.V.O.)
| | - Roy Schrijver
- Matisse Pharmaceuticals B.V., 6163 JT Geleen, The Netherlands; (R.S.); (P.E.); (S.d.K.)
| | - Peter Ekhart
- Matisse Pharmaceuticals B.V., 6163 JT Geleen, The Netherlands; (R.S.); (P.E.); (S.d.K.)
| | - Sjef de Kimpe
- Matisse Pharmaceuticals B.V., 6163 JT Geleen, The Netherlands; (R.S.); (P.E.); (S.d.K.)
| | - Gerry A. F. Nicolaes
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6200 MD Maastricht, The Netherlands; (C.P.M.R.); (H.S.); (R.V.O.)
| |
Collapse
|
17
|
Garcia B, Su F, Dewachter L, Wang Y, Li N, Remmelink M, Eycken MV, Khaldi A, Favory R, Herpain A, Moreau A, Moiroux-Sahraoui A, Manicone F, Annoni F, Shi L, Vincent JL, Creteur J, Taccone FS. Neutralization of extracellular histones by sodium-Β-O-methyl cellobioside sulfate in septic shock. Crit Care 2023; 27:458. [PMID: 38001494 PMCID: PMC10675855 DOI: 10.1186/s13054-023-04741-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: 10/09/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
BACKGROUND Extracellular histones have been associated with severity and outcome in sepsis. The aim of the present study was to assess the effects of sodium-β-O-Methyl cellobioside sulfate (mCBS), a histone-neutralizing polyanion, on the severity and outcome of sepsis in an experimental model. METHODS This randomized placebo-controlled experimental study was performed in 24 mechanically ventilated female sheep. Sepsis was induced by fecal peritonitis. Animals were randomized to three groups: control, early treatment, and late treatment (n = 8 each). mCBS was given as a bolus (1 mg/kg) followed by a continuous infusion (1 mg/kg/h) just after sepsis induction in the early treatment group, and 4 h later in the late treatment group. Fluid administration and antimicrobial therapy were initiated 4 h T4 after feces injection, peritoneal lavage performed, and a norepinephrine infusion titrated to maintain mean arterial pressure (MAP) between 65-75 mmHg. The experiment was blinded and lasted maximum 24 h. RESULTS During the first 4 h, MAP remained > 65 mmHg in the early treatment group but decreased significantly in the others (p < 0.01 for interaction, median value at T4: (79 [70-90] mmHg for early treatment, 57 [70-90] mmHg for late treatment, and 55 [49-60] mmHg for the control group). mCBS-treated animals required significantly less norepinephrine to maintain MAP than controls (p < 0.01 for interaction) and had lower creatinine (p < 0.01), lactate (p < 0.01), and interleukin-6 (p < 0.01) levels, associated with reduced changes in H3.1 nucleosome levels (p = 0.02). Early treatment was associated with lower norepinephrine requirements than later treatment. Two control animals died; all the mCBS-treated animals survived. CONCLUSIONS Neutralization of extracellular histones with mCBS was associated with reduced norepinephrine requirements, improved tissue perfusion, less renal dysfunction, and lower circulating IL-6 in experimental septic shock and may represent a new therapeutic approach to be tested in clinical trials.
Collapse
Affiliation(s)
- Bruno Garcia
- Experimental Laboratory of the Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles, 1070, Brussels, Belgium
- Department of Intensive Care, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Fuhong Su
- Experimental Laboratory of the Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles, 1070, Brussels, Belgium
| | - Laurence Dewachter
- Laboratory of Physiology and Pharmacology, Université Libre de Bruxelles, Brussels, Belgium
| | - Yong Wang
- Grand Pharma (China) Co., Ltd, Wuhan, China
| | - Ning Li
- Grand Pharma (China) Co., Ltd, Wuhan, China
| | - Myriam Remmelink
- Pathology Laboratory, Erasme Hospital, Hôpitaux Universitaires de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
| | - Marie Van Eycken
- Pathology Laboratory, Erasme Hospital, Hôpitaux Universitaires de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
| | - Amina Khaldi
- Experimental Laboratory of the Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles, 1070, Brussels, Belgium
| | - Raphaël Favory
- Department of Intensive Care, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Antoine Herpain
- Experimental Laboratory of the Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles, 1070, Brussels, Belgium
| | - Anthony Moreau
- Experimental Laboratory of the Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles, 1070, Brussels, Belgium
- Department of Intensive Care, Erasme Hospital, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
| | - Alexander Moiroux-Sahraoui
- Experimental Laboratory of the Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles, 1070, Brussels, Belgium
| | - Francesca Manicone
- Experimental Laboratory of the Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles, 1070, Brussels, Belgium
| | - Filippo Annoni
- Experimental Laboratory of the Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles, 1070, Brussels, Belgium
- Department of Intensive Care, Erasme Hospital, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
| | - Lin Shi
- Grand Pharma (China) Co., Ltd, Wuhan, China
| | - Jean-Louis Vincent
- Experimental Laboratory of the Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles, 1070, Brussels, Belgium.
| | - Jacques Creteur
- Experimental Laboratory of the Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles, 1070, Brussels, Belgium
| | - Fabio S Taccone
- Experimental Laboratory of the Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles, 1070, Brussels, Belgium
- Department of Intensive Care, Erasme Hospital, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
18
|
Pei XB, Liu B. Research Progress on the Mechanism and Management of Septic Cardiomyopathy: A Comprehensive Review. Emerg Med Int 2023; 2023:8107336. [PMID: 38029224 PMCID: PMC10681771 DOI: 10.1155/2023/8107336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 11/08/2023] [Accepted: 11/13/2023] [Indexed: 12/01/2023] Open
Abstract
Sepsis is defined as a kind of life-threatening organ dysfunction due to a dysregulated host immune response to infection and is a leading cause of mortality in the intensive care unit. Sepsis-induced myocardial dysfunction, also called septic cardiomyopathy, is a common and serious complication in patients with sepsis, which may indicate a bad prognosis. Although efforts have been made to uncover the pathophysiology of septic cardiomyopathy, a number of uncertainties remain. This article sought to review available literature to summarize the existing knowledge on current diagnostic tools and biomarkers, pathogenesis, and treatments for septic cardiomyopathy.
Collapse
Affiliation(s)
- Xue-Bin Pei
- Emergency Medicine Clinical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Bo Liu
- Department of Emergency Medicine, Beijing YouAn Hospital, Capital Medical University, Beijing 100069, China
| |
Collapse
|
19
|
Taha AM, Mahmoud AM, Ghonaim MM, Kamran A, AlSamhori JF, AlBarakat MM, Shrestha AB, Jaiswal V, Reiter RJ. Melatonin as a potential treatment for septic cardiomyopathy. Biomed Pharmacother 2023; 166:115305. [PMID: 37619482 DOI: 10.1016/j.biopha.2023.115305] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 08/02/2023] [Accepted: 08/07/2023] [Indexed: 08/26/2023] Open
Abstract
Septic cardiomyopathy (SCM) is a common complication of sepsis contributing to high mortality rates. Its pathophysiology involves complex factors, including inflammatory cytokines, mitochondrial dysfunction, oxidative stress, and immune dysregulation. Despite extensive research, no effective pharmacological agent has been established for sepsis-induced cardiomyopathy. Melatonin, a hormone with diverse functions in the body, has emerged as a potential agent for SCM through its anti-oxidant, anti-inflammatory, anti-apoptotic, and cardioprotective roles. Through various molecular levels of its mechanism of action, it counterattacks the adverse event of sepsis. Experimental studies have mentioned that melatonin protects against many cardiovascular diseases and exerts preventive effects on SCM. Moreover, melatonin has been investigated in combination with other drugs such as antibiotics, resveratrol, and anti-oxidants showing synergistic effects in reducing inflammation, anti-oxidant, and improving cardiac function. While preclinical studies have demonstrated positive results, clinical trials are required to establish the optimal dosage, route of administration, and treatment duration for melatonin in SCM. Its safety profile, low toxicity, and natural occurrence in the human body provide a favorable basis for its clinical use. This review aims to provide an overview of the current evidence of the use of melatonin in sepsis-induced cardiomyopathy (SICM). Melatonin appears to be promising as a possible treatment for sepsis-induced cardiomyopathy and demands further investigation.
Collapse
Affiliation(s)
- Amira Mohamed Taha
- Faculty of Medicine, Fayoum University, Fayoum, Egypt; Medical Research Group of Egypt (MRGE), Negida Academy, Arlington, MA, USA
| | | | | | - Ateeba Kamran
- Bachelor of Medicine, Bachelor of Surgery, Karachi Medical and Dental College, Karachi, Pakistan
| | | | - Majd M AlBarakat
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Abhigan Babu Shrestha
- Department of Internal Medicine, M Abdur Rahim Medical College, Dinajpur, Bangladesh.
| | | | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA
| |
Collapse
|
20
|
Ortega-Martorell S, Olier I, Johnston BW, Welters ID. Sepsis-induced coagulopathy is associated with new episodes of atrial fibrillation in patients admitted to critical care in sinus rhythm. Front Med (Lausanne) 2023; 10:1230854. [PMID: 37780563 PMCID: PMC10540306 DOI: 10.3389/fmed.2023.1230854] [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: 05/29/2023] [Accepted: 08/29/2023] [Indexed: 10/03/2023] Open
Abstract
Background Sepsis is a life-threatening disease commonly complicated by activation of coagulation and immune pathways. Sepsis-induced coagulopathy (SIC) is associated with micro- and macrothrombosis, but its relation to other cardiovascular complications remains less clear. In this study we explored associations between SIC and the occurrence of atrial fibrillation (AF) in patients admitted to the Intensive Care Unit (ICU) in sinus rhythm. We also aimed to identify predictive factors for the development of AF in patients with and without SIC. Methods Data were extracted from the publicly available AmsterdamUMCdb database. Patients with sepsis and documented sinus rhythm on admission to ICU were included. Patients were stratified into those who fulfilled the criteria for SIC and those who did not. Following univariate analysis, logistic regression models were developed to describe the association between routinely documented demographics and blood results and the development of at least one episode of AF. Machine learning methods (gradient boosting machines and random forest) were applied to define the predictive importance of factors contributing to the development of AF. Results Age was the strongest predictor for the development of AF in patients with and without SIC. Routine coagulation tests activated Partial Thromboplastin Time (aPTT) and International Normalized Ratio (INR) and C-reactive protein (CRP) as a marker of inflammation were also associated with AF occurrence in SIC-positive and SIC-negative patients. Cardiorespiratory parameters (oxygen requirements and heart rate) showed predictive potential. Conclusion Higher INR, elevated CRP, increased heart rate and more severe respiratory failure are risk factors for occurrence of AF in critical illness, suggesting an association between cardiac, respiratory and immune and coagulation pathways. However, age was the most dominant factor to predict the first episodes of AF in patients admitted in sinus rhythm with and without SIC.
Collapse
Affiliation(s)
- Sandra Ortega-Martorell
- School of Computer Science and Mathematics, Liverpool John Moores University, Liverpool, United Kingdom
- Liverpool Centre for Cardiovascular Science, Liverpool, United Kingdom
| | - Ivan Olier
- School of Computer Science and Mathematics, Liverpool John Moores University, Liverpool, United Kingdom
- Liverpool Centre for Cardiovascular Science, Liverpool, United Kingdom
| | - Brian W. Johnston
- Liverpool Centre for Cardiovascular Science, Liverpool, United Kingdom
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
- Liverpool University Hospitals NHS Foundation Trust, Liverpool, United Kingdom
| | - Ingeborg D. Welters
- Liverpool Centre for Cardiovascular Science, Liverpool, United Kingdom
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
- Liverpool University Hospitals NHS Foundation Trust, Liverpool, United Kingdom
| |
Collapse
|
21
|
Richards CM, McRae SA, Ranger AL, Klegeris A. Extracellular histones as damage-associated molecular patterns in neuroinflammatory responses. Rev Neurosci 2023; 34:533-558. [PMID: 36368030 DOI: 10.1515/revneuro-2022-0091] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 10/18/2022] [Indexed: 07/20/2023]
Abstract
The four core histones H2A, H2B, H3, H4, and the linker histone H1 primarily bind DNA and regulate gene expression within the nucleus. Evidence collected mainly from the peripheral tissues illustrates that histones can be released into the extracellular space by activated or damaged cells. In this article, we first summarize the innate immune-modulatory properties of extracellular histones and histone-containing complexes, such as nucleosomes, and neutrophil extracellular traps (NETs), described in peripheral tissues. There, histones act as damage-associated molecular patterns (DAMPs), which are a class of endogenous molecules that trigger immune responses by interacting directly with the cellular membranes and activating pattern recognition receptors (PRRs), such as toll-like receptors (TLR) 2, 4, 9 and the receptor for advanced glycation end-products (RAGE). We then focus on the available evidence implicating extracellular histones as DAMPs of the central nervous system (CNS). It is becoming evident that histones are present in the brain parenchyma after crossing the blood-brain barrier (BBB) or being released by several types of brain cells, including neurons, microglia, and astrocytes. However, studies on the DAMP-like effects of histones on CNS cells are limited. For example, TLR4 is the only known molecular target of CNS extracellular histones and their interactions with other PRRs expressed by brain cells have not been observed. Nevertheless, extracellular histones are implicated in the pathogenesis of a variety of neurological disorders characterized by sterile neuroinflammation; therefore, detailed studies on the role these proteins and their complexes play in these pathologies could identify novel therapeutic targets.
Collapse
Affiliation(s)
- Christy M Richards
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna V1V 1V7, BC, Canada
| | - Seamus A McRae
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna V1V 1V7, BC, Canada
| | - Athena L Ranger
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna V1V 1V7, BC, Canada
| | - Andis Klegeris
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna V1V 1V7, BC, Canada
| |
Collapse
|
22
|
Yong J, Abrams ST, Wang G, Toh CH. Cell-free histones and the cell-based model of coagulation. J Thromb Haemost 2023; 21:1724-1736. [PMID: 37116754 DOI: 10.1016/j.jtha.2023.04.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 04/06/2023] [Accepted: 04/20/2023] [Indexed: 04/30/2023]
Abstract
The cell-based model of coagulation remains the basis of our current understanding of clinical hemostasis and thrombosis. Its advancement on the coagulation cascade model has enabled new prohemostatic and anticoagulant treatments to be developed. In the past decade, there has been increasing evidence of the procoagulant properties of extracellular, cell-free histones (CFHs). Although high levels of circulating CFHs released following extensive cell death in acute critical illnesses, such as sepsis and trauma, have been associated with adverse coagulation outcomes, including disseminated intravascular coagulation, new information has also emerged on how its local effects contribute to physiological clot formation. CFHs initiate coagulation by tissue factor exposure, either by destruction of the endovascular barrier or induction of endoluminal tissue factor expression on endothelia and monocytes. CFHs can also bind prothrombin directly, generating thrombin via the alternative prothrombinase pathway. In amplifying and augmenting the procoagulant signal, CFHs activate and aggregate platelets, increase procoagulant material bioavailability through platelet degranulation and Weibel-Palade body exocytosis, activate intrinsic coagulation via platelet polyphosphate release, and induce phosphatidylserine exposure. CFHs also inhibit protein C activation and downregulate thrombomodulin expression to reduce anti-inflammatory and anticoagulant effects. In consolidating clot formation, CFHs augment the fibrin polymer to confer fibrinolytic resistance and integrate neutrophil extracellular traps into the clot structure. Such new information holds the promise of new therapeutic developments, including improved targeting of immunothrombotic pathologies in acute critical illnesses.
Collapse
Affiliation(s)
- Jun Yong
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, UK
| | - Simon T Abrams
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, UK; Liverpool Clinical Laboratories, Liverpool, UK
| | - Guozheng Wang
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, UK; Liverpool Clinical Laboratories, Liverpool, UK
| | - Cheng-Hock Toh
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, UK; The Roald Dahl Haemostasis and Thrombosis Centre, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK.
| |
Collapse
|
23
|
Li Y, Chen Y, Yang T, Chang K, Deng N, Zhao W, Su B. Targeting circulating high mobility group box-1 and histones by extracorporeal blood purification as an immunomodulation strategy against critical illnesses. Crit Care 2023; 27:77. [PMID: 36855150 PMCID: PMC9972334 DOI: 10.1186/s13054-023-04382-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 02/23/2023] [Indexed: 03/02/2023] Open
Abstract
Both high mobility group box-1 (HMGB1) and histones are major damage-associated molecular patterns (DAPMs) that mediate lethal systemic inflammation, activation of the complement and coagulation system, endothelial injury and multiple organ dysfunction syndrome in critical illnesses. Although accumulating evidence collectively shows that targeting HMGB1 or histones by their specific antibodies or inhibitors could significantly mitigate aberrant immune responses in multiple critically ill animal models, routine clinical use of such agents is still not recommended by any guideline. In contrast, extracorporeal blood purification, which has been widely used to replace dysfunctional organs and remove exogenous or endogenous toxins in intensive care units, may also exert an immunomodulatory effect by eliminating inflammatory mediators such as cytokines, endotoxin, HMGB1 and histones in patients with critical illnesses. In this review, we summarize the multiple immunopathological roles of HMGB1 and histones in mediating inflammation, immune thrombosis and organ dysfunction and discuss the rationale for the removal of these DAMPs using various hemofilters. The latest preclinical and clinical evidence for the use of extracorporeal blood purification to improve the clinical outcome of critically ill patients by targeting circulating HMGB1 and histones is also gathered.
Collapse
Affiliation(s)
- Yupei Li
- grid.13291.380000 0001 0807 1581Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Chen
- grid.13291.380000 0001 0807 1581State Key Laboratory of Polymer Materials Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu, China
| | - Tinghang Yang
- grid.13291.380000 0001 0807 1581Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China
| | - Kaixi Chang
- grid.13291.380000 0001 0807 1581Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China
| | - Ningyue Deng
- grid.13291.380000 0001 0807 1581Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China
| | - Weifeng Zhao
- State Key Laboratory of Polymer Materials Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu, China. .,Med-X Center for Materials, Sichuan University, Chengdu, China.
| | - Baihai Su
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China. .,Med-X Center for Materials, Sichuan University, Chengdu, China. .,Med+ Biomaterial Institute of West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
24
|
Hogwood J, Gray E, Mulloy B. Heparin, Heparan Sulphate and Sepsis: Potential New Options for Treatment. Pharmaceuticals (Basel) 2023; 16:271. [PMID: 37259415 PMCID: PMC9959362 DOI: 10.3390/ph16020271] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/22/2023] [Accepted: 02/07/2023] [Indexed: 08/31/2023] Open
Abstract
Sepsis is a life-threatening hyperreaction to infection in which excessive inflammatory and immune responses cause damage to host tissues and organs. The glycosaminoglycan heparan sulphate (HS) is a major component of the cell surface glycocalyx. Cell surface HS modulates several of the mechanisms involved in sepsis such as pathogen interactions with the host cell and neutrophil recruitment and is a target for the pro-inflammatory enzyme heparanase. Heparin, a close structural relative of HS, is used in medicine as a powerful anticoagulant and antithrombotic. Many studies have shown that heparin can influence the course of sepsis-related processes as a result of its structural similarity to HS, including its strong negative charge. The anticoagulant activity of heparin, however, limits its potential in treatment of inflammatory conditions by introducing the risk of bleeding and other adverse side-effects. As the anticoagulant potency of heparin is largely determined by a single well-defined structural feature, it has been possible to develop heparin derivatives and mimetic compounds with reduced anticoagulant activity. Such heparin mimetics may have potential for use as therapeutic agents in the context of sepsis.
Collapse
Affiliation(s)
- John Hogwood
- National Institute for Biological Standards and Control, Blanche Lane, South Mimms EN6 3QG, UK
| | - Elaine Gray
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King’s College London, Stamford St., London SE1 9NH, UK
| | - Barbara Mulloy
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King’s College London, Stamford St., London SE1 9NH, UK
| |
Collapse
|
25
|
Abrams ST, Alhamdi Y, Zi M, Guo F, Du M, Wang G, Cartwright EJ, Toh CH. Extracellular Histone-Induced Protein Kinase C Alpha Activation and Troponin Phosphorylation Is a Potential Mechanism of Cardiac Contractility Depression in Sepsis. Int J Mol Sci 2023; 24:ijms24043225. [PMID: 36834636 PMCID: PMC9967552 DOI: 10.3390/ijms24043225] [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: 01/06/2023] [Revised: 01/29/2023] [Accepted: 02/01/2023] [Indexed: 02/10/2023] Open
Abstract
Reduction in cardiac contractility is common in severe sepsis. However, the pathological mechanism is still not fully understood. Recently it has been found that circulating histones released after extensive immune cell death play important roles in multiple organ injury and disfunction, particularly in cardiomyocyte injury and contractility reduction. How extracellular histones cause cardiac contractility depression is still not fully clear. In this work, using cultured cardiomyocytes and a histone infusion mouse model, we demonstrate that clinically relevant histone concentrations cause significant increases in intracellular calcium concentrations with subsequent activation and enriched localization of calcium-dependent protein kinase C (PKC) α and βII into the myofilament fraction of cardiomyocytes in vitro and in vivo. Furthermore, histones induced dose-dependent phosphorylation of cardiac troponin I (cTnI) at the PKC-regulated phosphorylation residues (S43 and T144) in cultured cardiomyocytes, which was also confirmed in murine cardiomyocytes following intravenous histone injection. Specific inhibitors against PKCα and PKCβII revealed that histone-induced cTnI phosphorylation was mainly mediated by PKCα activation, but not PKCβII. Blocking PKCα also significantly abrogated histone-induced deterioration in peak shortening, duration and the velocity of shortening, and re-lengthening of cardiomyocyte contractility. These in vitro and in vivo findings collectively indicate a potential mechanism of histone-induced cardiomyocyte dysfunction driven by PKCα activation with subsequent enhanced phosphorylation of cTnI. These findings also indicate a potential mechanism of clinical cardiac dysfunction in sepsis and other critical illnesses with high levels of circulating histones, which holds the potential translational benefit to these patients by targeting circulating histones and downstream pathways.
Collapse
Affiliation(s)
- Simon T. Abrams
- Department of Clinical Infection Microbiology and Immunology, University of Liverpool, Liverpool L69 7BE, UK
- Coagulation Department, Liverpool University Hospitals NHS Foundation Trust, Liverpool L7 8XP, UK
| | - Yasir Alhamdi
- Department of Clinical Infection Microbiology and Immunology, University of Liverpool, Liverpool L69 7BE, UK
- Sheffield Teaching Hospital NHS Foundation Trust, Sheffield S5 7AU, UK
| | - Min Zi
- Institute of Cardiovascular Sciences, Centre for Cardiac Research, University of Manchester, Manchester M13 9PT, UK
| | - Fengmei Guo
- Department of Clinical Infection Microbiology and Immunology, University of Liverpool, Liverpool L69 7BE, UK
- The Medical School, Southeast University, Nanjing 210009, China
| | - Min Du
- Department of Clinical Infection Microbiology and Immunology, University of Liverpool, Liverpool L69 7BE, UK
| | - Guozheng Wang
- Department of Clinical Infection Microbiology and Immunology, University of Liverpool, Liverpool L69 7BE, UK
- Coagulation Department, Liverpool University Hospitals NHS Foundation Trust, Liverpool L7 8XP, UK
- Correspondence: (G.W.); (C.-H.T.)
| | - Elizabeth J. Cartwright
- Institute of Cardiovascular Sciences, Centre for Cardiac Research, University of Manchester, Manchester M13 9PT, UK
| | - Cheng-Hock Toh
- Department of Clinical Infection Microbiology and Immunology, University of Liverpool, Liverpool L69 7BE, UK
- Roald Dahl Haemostasis & Thrombosis Centre, Royal Liverpool University Hospital, Liverpool L7 8XP, UK
- Correspondence: (G.W.); (C.-H.T.)
| |
Collapse
|
26
|
The Importance of Pore-Forming Toxins in Multiple Organ Injury and Dysfunction. Biomedicines 2022; 10:biomedicines10123256. [PMID: 36552012 PMCID: PMC9776026 DOI: 10.3390/biomedicines10123256] [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: 11/03/2022] [Revised: 12/03/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Multiple organ injury and dysfunction often occurs in acute critical illness and adversely affects survival. However, in patients who survive, organ function usually recovers without permanent damage. It is, therefore, likely that there are reversible mechanisms, but this is poorly understood in the pathogenesis of multiple organ dysfunction syndrome (MODS). AIMS Based on our knowledge of extracellular histones and pneumolysin, as endogenous and exogenous pore-forming toxins, respectively, here we clarify if the extent of cell membrane disruption and recovery is important in MODS. METHODS This is a combination of retrospective clinical studies of a cohort of 98 patients from an intensive care unit (ICU) in a tertiary hospital, with interventional animal models and laboratory investigation. RESULTS In patients without septic shock and/or disseminate intravascular coagulation (DIC), circulating histones also strongly correlated with sequential organ failure assessment (SOFA) scores, suggesting their pore-forming property might play an important role. In vivo, histones or pneumolysin infusion similarly caused significant elevation of cell damage markers and multiple organ injury. In trauma and sepsis models, circulating histones strongly correlated with these markers, and anti-histone reagents significantly reduced their release. Comparison of pneumolysin deletion and its parental strain-induced sepsis mouse model showed that pneumolysin was not essential for sepsis development, but enhanced multiple organ damage and reduced survival time. In vitro, histones and pneumolysin treatment disrupt cell membrane integrity, resulting in changes in whole-cell currents and elevated intracellular Ca2+ to lead to Ca2+ overload. Cell-specific damage markers, lactate dehydrogenase (LDH), alanine aminotransferase (ALT), and cardiac troponin I (cTnI), were released from damaged cells. Once toxins were removed, cell membrane damage could be rapidly repaired and cellular function recovered. CONCLUSION This work has confirmed the importance of pore-forming toxins in the development of MODS and proposed a potential mechanism to explain the reversibility of MODS. This may form the foundation for the development of effective therapies.
Collapse
|
27
|
Chia PY, Teo A, Yeo TW. Association of Neutrophil Mediators With Dengue Disease Severity and Cardiac Impairment in Adults. J Infect Dis 2022; 226:1974-1984. [PMID: 36208158 DOI: 10.1093/infdis/jiac383] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 09/19/2022] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Cardiac impairment contributes to hypotension in severe dengue (SD). However, studies examining pathogenic factors affecting dengue-associated cardiac impairment are lacking. We examined the role of neutrophil mediators on cardiac impairment in clinical dengue. METHODS We prospectively enrolled adult patients with dengue and controls. Cardiac parameters were measured using a bioimpedance device. Neutrophils mediators were measured, including myeloperoxidase (MPO) and citrullinated histone H3. RESULTS We recruited 107 dengue patients and 30 controls. Patients with dengue were classified according to World Health Organization 2009 guidelines (44 with dengue fever [DF], 51 with DF with warning signs, and 12 with SD). During critical phase, stroke index (P < .001), cardiac index (P = .03), and Granov-Goor index (P < .001) were significantly lower in patients with dengue than in controls. During critical phase, MPO was significantly higher in patients with dengue than in controls (P < .001) and also significantly higher in patients with SD than in those with DF. In addition, MPO was inversely associated with the stroke, cardiac, and Granov-Goor indexes, during the critical phase, and longitudinally as well. CONCLUSIONS Cardiac function was decreased, and MPO increased, during with critical phase in patients SD compared with those with DF and controls. MPO may mediate dengue-associated cardiac impairment.
Collapse
Affiliation(s)
- Po Ying Chia
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore.,National Centre for Infectious Diseases, Singapore.,Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore
| | - Andrew Teo
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore.,Department of Medicine, The Doherty Institute, University of Melbourne, Victoria, Australia
| | - Tsin Wen Yeo
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore.,National Centre for Infectious Diseases, Singapore.,Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore
| |
Collapse
|
28
|
Circulating Histones and Severity of Illness in Children with Sepsis: A Prospective Observational Study. Indian J Pediatr 2022; 89:989-995. [PMID: 35181838 DOI: 10.1007/s12098-022-04084-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 10/25/2021] [Indexed: 11/05/2022]
Abstract
OBJECTIVES To measure circulating histone H3 levels in children with severe sepsis and explore its relationship with severity of illness and organ failures. METHODS Children aged 3 mo to 12 y with severe sepsis admitted to pediatric intensive care unit (PICU) were prospectively studied. Healthy controls were enrolled from the outpatient department for comparison. Levels of H3 histones were measured on day 1 and day 3. RESULTS Thirty-seven patients and 14 controls with median (IQR) age 5 (0.67, 8) and 5 (3, 7) y, respectively were enrolled. Common diagnoses included severe pneumonia (n = 9), staphylococcal sepsis (n = 6), and seasonal tropical infections (n = 4). Two-third (n = 26, 70%) had septic shock. One third (35%) had an unfavorable outcome; 11 died and 2 discontinued care. Median (IQR) H3 levels were not statistically different among patients with sepsis and controls [0.84 (0.62, 1.13) vs. 0.72 (0.52, 0.87) ng/mL; p = 0.10]. There was no significant change in H3 between day 1 and day 3 [0.84 (0.62, 1.13) vs 0.74 (0.5, 0.98) ng/mL; p = 0.22]. Children with thrombocytopenia (n = 27) showed a trend towards higher H3 compared to those without thrombocytopenia (n = 10) [0.9 vs. 0.67 ng/mL; p = 0.06]. However, H3 levels were not elevated in patients with cardiovascular dysfunction, those needing renal-replacement therapy, or unfavorable outcomes. CONCLUSION The present data provides early evidence that in children hospitalized with severe sepsis, histone H3 is not elevated as compared to healthy controls. H3 levels during initial days of sepsis requiring PICU admission were not different with regards to severity of illness, organ dysfunction, and clinical outcome. There was a trend towards elevated H3 in children with thrombocytopenia, which needs further evaluation.
Collapse
|
29
|
Carbone F, Liberale L, Preda A, Schindler TH, Montecucco F. Septic Cardiomyopathy: From Pathophysiology to the Clinical Setting. Cells 2022; 11:2833. [PMID: 36139408 PMCID: PMC9496713 DOI: 10.3390/cells11182833] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/05/2022] [Accepted: 09/07/2022] [Indexed: 11/21/2022] Open
Abstract
The onset of cardiomyopathy is a common feature in sepsis, with relevant effects on its pathophysiology and clinical care. Septic cardiomyopathy is characterized by reduced left ventricular (LV) contractility eventually associated with LV dilatation with or without right ventricle failure. Unfortunately, such a wide range of ultrasonographic findings does not reflect a deep comprehension of sepsis-induced cardiomyopathy, but rather a lack of consensus about its definition. Several echocardiographic parameters intrinsically depend on loading conditions (both preload and afterload) so that it may be challenging to discriminate which is primitive and which is induced by hemodynamic perturbances. Here, we explore the state of the art in sepsis-related cardiomyopathy. We focus on the shortcomings in its definition and point out how cardiac performance dynamically changes in response to different hemodynamic clusters. A special attention is also given to update the knowledge about molecular mechanisms leading to myocardial dysfunction and that recall those of myocardial hibernation. Ultimately, the aim of this review is to highlight the unsolved issue in the field of sepsis-induced cardiomyopathy as their implementation would lead to improve risk stratification and clinical care.
Collapse
Affiliation(s)
- Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa-Italian Cardiovascular Network, 16132 Genoa, Italy
| | - Luca Liberale
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa-Italian Cardiovascular Network, 16132 Genoa, Italy
| | - Alberto Preda
- Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Thomas Hellmut Schindler
- Mallinckrodt Institute of Radiology, Division of Nuclear Medicine, School of Medicine, Washington University, Saint Louis, MO 63110, USA
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa-Italian Cardiovascular Network, 16132 Genoa, Italy
| |
Collapse
|
30
|
Laudanski K, Liu D, Hajj J, Ghani D, Szeto WY. Serum level of total histone 3, H3K4me3, and H3K27ac after non-emergent cardiac surgery suggests the persistence of smoldering inflammation at 3 months in an adult population. Clin Epigenetics 2022; 14:112. [PMID: 36068552 PMCID: PMC9446722 DOI: 10.1186/s13148-022-01331-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 08/24/2022] [Indexed: 11/29/2022] Open
Abstract
Background Despite clinical relevance of immunological activation due to histone leakage into the serum following cardiac surgery, long-term data describing their longitudinal dynamic are lacking. Therefore, this study examines the serum levels of histone 3 (tH3) and its modifications (H3K4me3 and H3K27ac) alongside immune system activation during the acute and convalescence phases of cardiac surgery. Methods Blood samples from fifty-nine individuals were collected before non-emergent cardiac surgery (tpre-op) and 24 h (t24hr), seven days (t7d), and three months (t3m) post-procedure to examine serum levels of tH3, H3K4me3, and H3K27ac. Serum heat shock protein-60 (HSP-60) was a surrogate of the cellular damage marker. Serum C-reactive protein (CRP) and interleukin 6 (IL-6) assessed smoldering inflammation. TNFα and IL-6 production by whole blood in response to lipopolysaccharide (LPS) evaluated immunological activation. Electronic medical records provided demographic, peri-operative, and clinical information. Paired longitudinal analyses were employed with data expressed as mean and standard deviation (X ± SD) or median and interquartile range (Me[IQ25; 75%]. Results Compared to pre-operative levels (tH3Pre-op = 1.6[0.33;2.4]), post-operative serum tH3 significantly (p > 0.0001) increased after heart surgery (tH324hr = 2.2[0.3;28]), remained elevated at 7 days (tH37d = 2.4[0.37;5.3]), and at 3 months (tH33m = 2.0[0.31;2.9]). Serum H3K27ac was elevated at 24 h (H3K27ac24hr = 0.66 ± 0.51; p = 0.025) and seven days (H3K27ac7d = 0.94 ± 0.95; p = 0.032) as compared to baseline hours (H3K27acPre-op = 0.55 ± 0.54). Serum H3K4me3 was significantly diminished at three months (H3K4me3Pre-op = 0.94 ± 0.54 vs. H3K27ac3m = 0.59 ± 0.89; p = 0.008). tH3 correlated significantly with the duration of anesthesia (r2 = 0.38). In contrast, HSP-60 normalized seven days after surgery. Peri-operative intake of acetaminophen, but no acetylsalicylic acid (ASA), acid, ketorolac or steroids, resulted in the significant depression of serum H3K4me3 at 24 h (H3K4me3acetom- = 1.26[0.71; 3.21] vs H3K4me3acetom+ = 0.54[0.07;1.01]; W[50] = 2.26; p = 0.021). CRP, but not IL-6, remained elevated at 3 months compared to pre-surgical levels and correlated with tH324hrs (r2 = 0.43), tH37d (r2 = 0.71; p < 0.05), H3K4me37d (r2 = 0.53), and H3K27ac7d (r2 = 0.49). Production of TNFα by whole blood in response to LPS was associated with serum tH324hrs (r2 = 0.67). Diminished H3K4me324hrs, H3K27ac24hrs, and H3K27ac3m, accompanied the emergence of liver failure. Conclusions We demonstrated a prolonged elevation in serum histone 3 three months after cardiac surgery. Furthermore, histone 3 modifications had a discrete time evolution indicating differential immune activation.
Collapse
Affiliation(s)
- Krzysztof Laudanski
- Department of Anesthesiology and Critical Care, University of Pennsylvania, JMB 127, 3620 Hamilton Walk, Philadelphia, PA, 19146, USA. .,Department of Neurology, University of Pennsylvania, JMB 127, 3620 Hamilton Walk, Philadelphia, PA, 19146, USA. .,Leonard Davis Institute for Health Economics, University of Pennsylvania, JMB 127, 3620 Hamilton Walk, Philadelphia, PA, 19146, USA.
| | - Da Liu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Jihane Hajj
- School of Nursing, Widener University, Philadelphia, PA, USA
| | - Danyal Ghani
- Department of Cardiac Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Wilson Y Szeto
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
31
|
Zhu CL, Wang Y, Liu Q, Li HR, Yu CM, Li P, Deng XM, Wang JF. Dysregulation of neutrophil death in sepsis. Front Immunol 2022; 13:963955. [PMID: 36059483 PMCID: PMC9434116 DOI: 10.3389/fimmu.2022.963955] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 08/02/2022] [Indexed: 11/13/2022] Open
Abstract
Sepsis is a prevalent disease that has alarmingly high mortality rates and, for several survivors, long-term morbidity. The modern definition of sepsis is an aberrant host response to infection followed by a life-threatening organ dysfunction. Sepsis has a complicated pathophysiology and involves multiple immune and non-immune mediators. It is now believed that in the initial stages of sepsis, excessive immune system activation and cascading inflammation are usually accompanied by immunosuppression. During the pathophysiology of severe sepsis, neutrophils are crucial. Recent researches have demonstrated a clear link between the process of neutrophil cell death and the emergence of organ dysfunction in sepsis. During sepsis, spontaneous apoptosis of neutrophils is inhibited and neutrophils may undergo some other types of cell death. In this review, we describe various types of neutrophil cell death, including necrosis, apoptosis, necroptosis, pyroptosis, NETosis, and autophagy, to reveal their known effects in the development and progression of sepsis. However, the exact role and mechanisms of neutrophil cell death in sepsis have not been fully elucidated, and this remains a major challenge for future neutrophil research. We hope that this review will provide hints for researches regarding neutrophil cell death in sepsis and provide insights for clinical practitioners.
Collapse
|
32
|
Beltrán-García J, Osca-Verdegal R, Pérez-Cremades D, Novella S, Hermenegildo C, Pallardó FV, García-Giménez JL. Extracellular Histones Activate Endothelial NLRP3 Inflammasome and are Associated with a Severe Sepsis Phenotype. J Inflamm Res 2022; 15:4217-4238. [PMID: 35915852 PMCID: PMC9338392 DOI: 10.2147/jir.s363693] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 06/29/2022] [Indexed: 12/27/2022] Open
Affiliation(s)
- Jesús Beltrán-García
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Investigación Sanitaria INCLIVA, Valencia, Spain
- Departamento de Fisiología, Facultad de Medicina y Odontología, Universitat de València, València, Spain
| | - Rebeca Osca-Verdegal
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Departamento de Fisiología, Facultad de Medicina y Odontología, Universitat de València, València, Spain
| | - Daniel Pérez-Cremades
- Instituto de Investigación Sanitaria INCLIVA, Valencia, Spain
- Departamento de Fisiología, Facultad de Medicina y Odontología, Universitat de València, València, Spain
| | - Susana Novella
- Instituto de Investigación Sanitaria INCLIVA, Valencia, Spain
- Departamento de Fisiología, Facultad de Medicina y Odontología, Universitat de València, València, Spain
| | - Carlos Hermenegildo
- Instituto de Investigación Sanitaria INCLIVA, Valencia, Spain
- Departamento de Fisiología, Facultad de Medicina y Odontología, Universitat de València, València, Spain
| | - Federico V Pallardó
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Investigación Sanitaria INCLIVA, Valencia, Spain
- Departamento de Fisiología, Facultad de Medicina y Odontología, Universitat de València, València, Spain
| | - José Luis García-Giménez
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Investigación Sanitaria INCLIVA, Valencia, Spain
- Departamento de Fisiología, Facultad de Medicina y Odontología, Universitat de València, València, Spain
- Correspondence: José Luis García-Giménez, Departamento de Fisiología, Facultad de Medicina y Odontología, Universitat de València, València, 46010, Spain, Tel +34 963 864 646, Email
| |
Collapse
|
33
|
Block H, Rossaint J, Zarbock A. The Fatal Circle of NETs and NET-Associated DAMPs Contributing to Organ Dysfunction. Cells 2022; 11:1919. [PMID: 35741047 PMCID: PMC9222025 DOI: 10.3390/cells11121919] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/02/2022] [Accepted: 06/10/2022] [Indexed: 02/07/2023] Open
Abstract
The innate immune system is the first line of defense against invading pathogens or sterile injuries. Pattern recognition receptors (PRR) sense molecules released from inflamed or damaged cells, or foreign molecules resulting from invading pathogens. PRRs can in turn induce inflammatory responses, comprising the generation of cytokines or chemokines, which further induce immune cell recruitment. Neutrophils represent an essential factor in the early immune response and fulfill numerous tasks to fight infection or heal injuries. The release of neutrophil extracellular traps (NETs) is part of it and was originally attributed to the capture and elimination of pathogens. In the last decade studies revealed a detrimental role of NETs during several diseases, often correlated with an exaggerated immune response. Overwhelming inflammation in single organs can induce remote organ damage, thereby further perpetuating release of inflammatory molecules. Here, we review recent findings regarding damage-associated molecular patterns (DAMPs) which are able to induce NET formation, as well as NET components known to act as DAMPs, generating a putative fatal circle of inflammation contributing to organ damage and sequentially occurring remote organ injury.
Collapse
Affiliation(s)
| | | | - Alexander Zarbock
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, 48149 Muenster, Germany; (H.B.); (J.R.)
| |
Collapse
|
34
|
Endothelial Dysfunction Induced by Extracellular Neutrophil Traps Plays Important Role in the Occurrence and Treatment of Extracellular Neutrophil Traps-Related Disease. Int J Mol Sci 2022; 23:ijms23105626. [PMID: 35628437 PMCID: PMC9147606 DOI: 10.3390/ijms23105626] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/16/2022] [Accepted: 05/17/2022] [Indexed: 01/27/2023] Open
Abstract
Many articles have demonstrated that extracellular neutrophil traps (NETs) are often described as part of the antibacterial function. However, since the components of NETs are non-specific, excessive NETs usually cause inflammation and tissue damage. Endothelial dysfunction (ED) caused by NETs is the major focus of tissue damage, which is highly related to many inflammatory diseases. Therefore, this review summarizes the latest advances in the primary and secondary mechanisms between NETs and ED regarding inflammation as a mediator. Moreover, the detailed molecular mechanisms with emphasis on the disadvantages from NETs are elaborated: NETs can use its own enzymes, release particles as damage-associated molecular patterns (DAMPs) and activate the complement system to interact with endothelial cells (ECs), drive ECs damage and eventually aggravate inflammation. In view of the role of NETs-induced ED in different diseases, we also discussed possible molecular mechanisms and the treatments of NETs-related diseases.
Collapse
|
35
|
Huang Z, Zhang H, Fu X, Han L, Zhang H, Zhang L, Zhao J, Xiao D, Li H, Li P. Autophagy-driven neutrophil extracellular traps: The dawn of sepsis. Pathol Res Pract 2022; 234:153896. [PMID: 35462228 DOI: 10.1016/j.prp.2022.153896] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 03/28/2022] [Accepted: 04/11/2022] [Indexed: 12/12/2022]
Abstract
Sepsis is a systemic inflammatory syndrome caused by infection disorders. The core mechanism of sepsis is immune dysfunction. Neutrophils are the most abundant circulating white blood cells, which play a crucial role in mediating the innate immune response. Previous studies have shown that an effective way to treat sepsis is through the regulation of neutrophil functions. Autophagy, a highly conserved degradation process, is responsible for removing denatured proteins or damaged organelles within cells and protecting cells from external stimuli. It is a key homeostasis process that promotes neutrophil function and differentiation. Autophagy has been shown to be closely associated with inflammation and immunity. Neutrophils, the first line of innate immunity, migrate to inflammatory sites upon their activation. Neutrophil-mediated autophagy may participate in the clinical course of sepsis. In this review, we summarized and analyzed the latest research findings on the changes in neutrophil external traps during sepsis, the regulatory role of autophagy in neutrophil, and the potential application of autophagy-driven NETs in sepsis, so as to guide clinical treatment of sepsis.
Collapse
Affiliation(s)
- Zhenzhen Huang
- Department of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, China
| | - Haodong Zhang
- Department of Hypertension Center, Lanzhou University Second Hospital, Lanzhou, China
| | - Xu Fu
- Key Laboratory of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, China
| | - Li Han
- Key Laboratory of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, China
| | - Haidan Zhang
- Department of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, China
| | - Ling Zhang
- Department of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, China
| | - Jing Zhao
- Department of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, China
| | - Danyang Xiao
- Department of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, China
| | - Hongyao Li
- Department of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, China
| | - Peiwu Li
- Department of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, China.
| |
Collapse
|
36
|
Yehya N, Fazelinia H, Taylor DM, Lawrence GG, Spruce LA, Thompson JM, Margulies SS, Seeholzer SH, Worthen GS. Differentiating children with sepsis with and without acute respiratory distress syndrome using proteomics. Am J Physiol Lung Cell Mol Physiol 2022; 322:L365-L372. [PMID: 34984927 PMCID: PMC8873032 DOI: 10.1152/ajplung.00164.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Both sepsis and acute respiratory distress syndrome (ARDS) rely on imprecise clinical definitions leading to heterogeneity, which has contributed to negative trials. Because circulating protein/DNA complexes have been implicated in sepsis and ARDS, we aimed to develop a proteomic signature of DNA-bound proteins to discriminate between children with sepsis with and without ARDS. We performed a prospective case-control study in 12 children with sepsis with ARDS matched to 12 children with sepsis without ARDS on age, severity of illness score, and source of infection. We performed co-immunoprecipitation and downstream proteomics in plasma collected ≤ 24 h of intensive care unit admission. Expression profiles were generated, and a random forest classifier was used on differentially expressed proteins to develop a signature which discriminated ARDS. The classifier was tested in six independent blinded samples. Neutrophil and nucleosome proteins were over-represented in ARDS, including two S100A proteins, superoxide dismutase (SOD), and three histones. Random forest produced a 10-protein signature that accurately discriminated between children with sepsis with and without ARDS. This classifier perfectly assigned six independent blinded samples as having ARDS or not. We validated higher expression of the most informative discriminating protein, galectin-3-binding protein, in children with ARDS. Our methodology has applicability to isolation of DNA-bound proteins from plasma. Our results support the premise of a molecular definition of ARDS, and give preliminary insight into why some children with sepsis, but not others, develop ARDS.
Collapse
Affiliation(s)
- Nadir Yehya
- 1Department of Anesthesiology and Critical Care Medicine, The Children’s Hospital of Philadelphia and University of Pennsylvania, Philadelphia, Pennsylvania
| | - Hossein Fazelinia
- 2Proteomics Core, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Deanne M. Taylor
- 3Department of Biomedical and Health Informatics, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania,6Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Gladys G. Lawrence
- 4Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Lynn A. Spruce
- 2Proteomics Core, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Jill M. Thompson
- 1Department of Anesthesiology and Critical Care Medicine, The Children’s Hospital of Philadelphia and University of Pennsylvania, Philadelphia, Pennsylvania
| | - Susan S. Margulies
- 5Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia
| | - Steven H. Seeholzer
- 2Proteomics Core, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - G. Scott Worthen
- 6Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| |
Collapse
|
37
|
Donis N, Jiang Z, D'Emal C, Hulin A, Debuisson M, Dulgheru R, Nguyen ML, Postolache A, Lallemand F, Coucke P, Martinive P, Herzog M, Pamart D, Terrell J, Pincemail J, Drion P, Delvenne P, Nchimi A, Lancellotti P, Oury C. Differential Biological Effects of Dietary Lipids and Irradiation on the Aorta, Aortic Valve, and the Mitral Valve. Front Cardiovasc Med 2022; 9:839720. [PMID: 35295264 PMCID: PMC8918952 DOI: 10.3389/fcvm.2022.839720] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 01/24/2022] [Indexed: 01/12/2023] Open
Abstract
Aims Dietary cholesterol and palmitic acid are risk factors for cardiovascular diseases (CVDs) affecting the arteries and the heart valves. The ionizing radiation that is frequently used as an anticancer treatment promotes CVD. The specific pathophysiology of these distinct disease manifestations is poorly understood. We, therefore, studied the biological effects of these dietary lipids and their cardiac irradiation on the arteries and the heart valves in the rabbit models of CVD. Methods and Results Cholesterol-enriched diet led to the thickening of the aortic wall and the aortic valve leaflets, immune cell infiltration in the aorta, mitral and aortic valves, as well as aortic valve calcification. Numerous cells expressing α-smooth muscle actin were detected in both the mitral and aortic valves. Lard-enriched diet induced massive aorta and aortic valve calcification, with no detectable immune cell infiltration. The addition of cardiac irradiation to the cholesterol diet yielded more calcification and more immune cell infiltrates in the atheroma and the aortic valve than cholesterol alone. RNA sequencing (RNAseq) analyses of aorta and heart valves revealed that a cholesterol-enriched diet mainly triggered inflammation-related biological processes in the aorta, aortic and mitral valves, which was further enhanced by cardiac irradiation. Lard-enriched diet rather affected calcification- and muscle-related processes in the aorta and aortic valve, respectively. Neutrophil count and systemic levels of platelet factor 4 and ent-8-iso-15(S)-PGF2α were identified as early biomarkers of cholesterol-induced tissue alterations, while cardiac irradiation resulted in elevated levels of circulating nucleosomes. Conclusion Dietary cholesterol, palmitic acid, and cardiac irradiation combined with a cholesterol-rich diet led to the development of distinct vascular and valvular lesions and changes in the circulating biomarkers. Hence, our study highlights unprecedented specificities related to common risk factors that underlie CVD.
Collapse
Affiliation(s)
- Nathalie Donis
- Laboratory of Cardiology, Department of Cardiology, GIGA Institute, University of Liège Hospital, CHU Sart Tilman, Liège, Belgium
| | - Zheshen Jiang
- Laboratory of Cardiology, Department of Cardiology, GIGA Institute, University of Liège Hospital, CHU Sart Tilman, Liège, Belgium
| | - Céline D'Emal
- Laboratory of Cardiology, Department of Cardiology, GIGA Institute, University of Liège Hospital, CHU Sart Tilman, Liège, Belgium
| | - Alexia Hulin
- Laboratory of Cardiology, Department of Cardiology, GIGA Institute, University of Liège Hospital, CHU Sart Tilman, Liège, Belgium
| | - Margaux Debuisson
- Laboratory of Cardiology, Department of Cardiology, GIGA Institute, University of Liège Hospital, CHU Sart Tilman, Liège, Belgium
| | - Raluca Dulgheru
- Laboratory of Cardiology, Department of Cardiology, GIGA Institute, University of Liège Hospital, CHU Sart Tilman, Liège, Belgium
| | - Mai-Linh Nguyen
- Laboratory of Cardiology, Department of Cardiology, GIGA Institute, University of Liège Hospital, CHU Sart Tilman, Liège, Belgium
| | - Adriana Postolache
- Laboratory of Cardiology, Department of Cardiology, GIGA Institute, University of Liège Hospital, CHU Sart Tilman, Liège, Belgium
| | | | | | - Philippe Martinive
- Department Radiation Oncology, Institut Jules Bordet, Université Libre Bruxelles, Brussels, Belgium
| | - Marielle Herzog
- Belgian Volition Société à Responsabilité Limitée, Gembloux, Belgium
| | - Dorian Pamart
- Belgian Volition Société à Responsabilité Limitée, Gembloux, Belgium
| | - Jason Terrell
- Department of Oncology and Livestrong Cancer Institutes, Dell Medical School, University of Texas at Austin, Austin, TX, United States
- Volition America, Austin, TX, United States
| | | | - Pierre Drion
- Experimental Surgery Unit, Centre de Recherche du Département de Chrirurgie, Groupe Interdisciplinaire de Géno-Protéomique Appliquée Institute, University of Liège, Liège, Belgium
| | - Philippe Delvenne
- Department of Pathology, Centre Hospitalier Universitaire of Liège, Liège, Belgium
- Laboratory of Experimental Pathology, GIGA Institute, University of Liège, Liège, Belgium
| | - Alain Nchimi
- Laboratory of Cardiology, Department of Cardiology, GIGA Institute, University of Liège Hospital, CHU Sart Tilman, Liège, Belgium
| | - Patrizio Lancellotti
- Laboratory of Cardiology, Department of Cardiology, GIGA Institute, University of Liège Hospital, CHU Sart Tilman, Liège, Belgium
- Gruppo Villa Maria Care and Research, Maria Cecilia Hospital, Cotignola, Italy
- Anthea Hospital, Bari, Italy
| | - Cécile Oury
- Laboratory of Cardiology, Department of Cardiology, GIGA Institute, University of Liège Hospital, CHU Sart Tilman, Liège, Belgium
| |
Collapse
|
38
|
Chen C, Lin Z, Zhang X, Zhang X, Cheng Z, Jin T, Liu T, Deng L, Guo J, Wang G, Xia Q. Extracellular histones cause intestinal epithelium injury and disrupt its barrier function in vitro and in vivo. Toxicology 2022; 469:153117. [PMID: 35122914 DOI: 10.1016/j.tox.2022.153117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 01/29/2022] [Accepted: 02/01/2022] [Indexed: 02/05/2023]
Abstract
Extracellular histones are cytotoxic to various cells and have been extensively proven a vital mediator of multiple organ injuries. However, the effect of extracellular histones on the intestine remains largely unknown. This study aimed to clarify the effect of extracellular histones on the intestine. IEC-6, a cell line of rat small intestinal epithelial crypt, and C57BL/6 or ICR mice were treated with histones. The IEC-6 cells treated with histones from 20 μg/mL to 200 μg/mL for 0-24 h displayed a decline of cell viability and an increase of cell death in a concentration- and time-dependent manner. Moreover, histones (100 μg/mL) induced IEC-6 apoptosis through activating caspase 3 and necroptosis through up-regulation of receptor-interacting serine/threonine protein kinase 1 and 3 (RIPK1 and RIPK3), phosphorylated mixed-lineage kinase domain-like protein (p-MLKL) along with the decrease of caspase-8. Histones treatment disturbed zonular occludens 1 (ZO-1) expression and increased permeability of IEC-6 cell monolayer. In vivo, histones 50 mg/kg injection caused mice intestinal edema, loss apex of villus, epithelial lifting down the sides of the villi, and increased neutrophil infiltration. Elevation of serum intestinal fatty acid binding protein (I-FABP), D-lactate, or Diamine oxidase (DAO) and loss of tight junction protein, ZO-1, at 3 h and 6 h after histones injection strongly indicated severe intestinal epithelium injury, which led to increased permeability of the intestine. In conclusion, extracellular histones cause intestinal epithelial damage via direct cytotoxicity. Consequently, intestinal epithelial tight junction and barrier integrity are disrupted, which may play pivotal roles in diverse diseases.
Collapse
Affiliation(s)
- Chanjuan Chen
- Department of Integrated Traditional Chinese and Western Medicine, Sichuan Provincial Pancreatitis Centre and West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ziqi Lin
- Department of Integrated Traditional Chinese and Western Medicine, Sichuan Provincial Pancreatitis Centre and West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Xiaoxin Zhang
- Department of Integrated Traditional Chinese and Western Medicine, Sichuan Provincial Pancreatitis Centre and West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiaoying Zhang
- Department of Integrated Traditional Chinese and Western Medicine, Sichuan Provincial Pancreatitis Centre and West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhenxing Cheng
- The Medical School, Southeast University, Nanjing, 210009, China; Department of Gastroenterology, the First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Tao Jin
- Department of Integrated Traditional Chinese and Western Medicine, Sichuan Provincial Pancreatitis Centre and West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Tingting Liu
- Department of Integrated Traditional Chinese and Western Medicine, Sichuan Provincial Pancreatitis Centre and West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lihui Deng
- Department of Integrated Traditional Chinese and Western Medicine, Sichuan Provincial Pancreatitis Centre and West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jia Guo
- Department of Integrated Traditional Chinese and Western Medicine, Sichuan Provincial Pancreatitis Centre and West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Guozheng Wang
- The Medical School, Southeast University, Nanjing, 210009, China
| | - Qing Xia
- Department of Gastroenterology, the First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
39
|
Yehya N. Potential therapeutics in pediatric acute respiratory distress syndrome: what does the immune system have to offer? A narrative review. Transl Pediatr 2021; 10:2689-2699. [PMID: 34765494 PMCID: PMC8578784 DOI: 10.21037/tp-20-341] [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: 10/14/2020] [Accepted: 01/22/2021] [Indexed: 11/09/2022] Open
Abstract
Since first described, acute respiratory distress syndrome (ARDS) has been understood to be an inflammatory disease with a dysregulated hyperinflammatory response. While fewer investigations have studied these phenomena in pediatric ARDS (PARDS), similar pathways are believed to be involved. Significant attention has been paid to the innate immune system, particularly neutrophils and neutrophil-related signaling, more recent studies have provided additional nuance regarding the role of upstream damage-associated molecular patterns (DAMPs) and subsequent neutrophil-mediated inflammation, lung permeability, and alveolar epithelial damage. For example, neutrophil extracellular traps (NETs) and inflammasome signaling have been identified as critical mediators existing at the junction of DAMPs and downstream inflammation. We demonstrate how the conclusions obtained from pre-clinical studies of lung injury are highly dependent upon the model chosen, and how this can lead us astray when developing therapies. More recently the adaptive immune system, specifically select T cell subpopulations, have also been implicated in ARDS. This raises the possibility of antigen-specific immunomodulation as a potential therapeutic avenue in ARDS. Finally, we briefly review randomized controlled trials attempting to manipulate the immune dysregulation in ARDS, including pleiotropic immunomodulators like corticosteroids and interferon-β, and what these studies can teach us about the design of novel therapeutics and the design of future trials.
Collapse
Affiliation(s)
- Nadir Yehya
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
40
|
Neutrophil extracellular traps and organ dysfunction in sepsis. Clin Chim Acta 2021; 523:152-162. [PMID: 34537216 DOI: 10.1016/j.cca.2021.09.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/09/2021] [Accepted: 09/14/2021] [Indexed: 12/12/2022]
Abstract
Sepsis is a clinical syndrome resulting from infection followed by inflammation and is one of the significant causes of mortality worldwide. The underlying reason is the host's uncontrolled inflammatory response due to an infection led to multiple organ dysfunction/failure. Neutrophils, an innate immune cell, are forerunners to reach the site of infection/inflammation for clearing the infection and resolute the inflammation during sepsis. A relatively new neutrophil effector function, neutrophil extracellular traps (NETs), have been demonstrated to kill the pathogens by releasing DNA decorated with histone and granular proteins. A growing number of pieces of shreds of evidence suggest that unregulated incidence of NETs have a significant influence on the pathogenesis of sepsis-induced multiple organ damage, including arterial hypotension, hypoxemia, coagulopathy, renal, neurological, and hepatic dysfunction. Thus, excessive production and improper resolution of NETs are of significant therapeutic value in combating sepsis-induced multiple organ failure. The purpose of this review is intended to highlight the role of NETs in sepsis-induced organ failure. Furthermore, the current status of therapeutic strategies to intersect the harmful effects of NETs to restore organ functions is discussed.
Collapse
|
41
|
Comparative Analysis of Chromatin-Delivered Biomarkers in the Monitoring of Sepsis and Septic Shock: A Pilot Study. Int J Mol Sci 2021; 22:ijms22189935. [PMID: 34576097 PMCID: PMC8465401 DOI: 10.3390/ijms22189935] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/10/2021] [Accepted: 09/11/2021] [Indexed: 12/12/2022] Open
Abstract
Sepsis management remains one of the most important challenges in modern clinical practice. Rapid progression from sepsis to septic shock is practically unpredictable, hence the critical need for sepsis biomarkers that can help clinicians in the management of patients to reduce the probability of a fatal outcome. Circulating nucleoproteins released during the inflammatory response to infection, including neutrophil extracellular traps, nucleosomes, and histones, and nuclear proteins like HMGB1, have been proposed as markers of disease progression since they are related to inflammation, oxidative stress, endothelial damage, and impairment of the coagulation response, among other pathological features. The aim of this work was to evaluate the actual potential for decision making/outcome prediction of the most commonly proposed chromatin-related biomarkers (i.e., nucleosomes, citrullinated H3, and HMGB1). To do this, we compared different ELISA measuring methods for quantifying plasma nucleoproteins in a cohort of critically ill patients diagnosed with sepsis or septic shock compared to nonseptic patients admitted to the intensive care unit (ICU), as well as to healthy subjects. Our results show that all studied biomarkers can be used to monitor sepsis progression, although they vary in their effectiveness to separate sepsis and septic shock patients. Our data suggest that HMGB1/citrullinated H3 determination in plasma is potentially the most promising clinical tool for the monitoring and stratification of septic patients.
Collapse
|
42
|
Makatsariya AD, Slukhanchuk EV, Bitsadze VO, Khizroeva JK, Tretyakova MV, Makatsariya NA, Akinshina SV, Shkoda AS, Pankratyeva LL, Di Renzo GC, Rizzo G, Grigorieva KN, Tsibizova VI, Gris JC, Elalamy I. Neutrophil extracellular traps: a role in inflammation and dysregulated hemostasis as well as in patients with COVID-19 and severe obstetric pathology. OBSTETRICS, GYNECOLOGY AND REPRODUCTION 2021; 15:335-350. [DOI: 10.17749/2313-7347/ob.gyn.rep.2021.238] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Numerous studies have proven a close relationship between inflammatory diseases and the state of hypercoagulability. In fact, thromboembolic complications represent one of the main causes of disability and mortality in acute and chronic inflammatory diseases, cancer and obstetric complications. Despite this, the processes of hemostasis and immune responses have long been considered separately; currently, work is underway to identify the molecular basis for a relationship between such systems. It has been identified that various pro-inflammatory stimuli are capable of triggering a coagulation cascade, which in turn modulates inflammatory responses. Neutrophil extracellular traps (NETs) are the networks of histones of extracellular DNA generated by neutrophils in response to inflammatory stimuli. The hemostasis is activated against infection in order to minimize the spread of infection and, if possible, inactivate the infectious agent. Another molecular network is based on fibrin. Over the last 10 years, there has been accumulated a whole body of evidence that NETs and fibrin are able to form a united network within a thrombus, stabilizing each other. Similarities and molecular cross-reactions are also present in the processes of fibrinolysis and lysis of NETs. Both NETs and von Willebrand factor (vWF) are involved in thrombosis as well as inflammation. During the development of these conditions, a series of events occurs in the microvascular network, including endothelial activation, NETs formation, vWF secretion, adhesion, aggregation, and activation of blood cells. The activity of vWF multimers is regulated by the specific metalloproteinase ADAMTS-13 (a disintegrin and metalloproteinase with a thrombospondin type 1 motif, member 13). Studies have shown that interactions between NETs and vWF can lead to arterial and venous thrombosis and inflammation. In addition, the contents released from activated neutrophils or NETs result in decreased ADAMTS-13 activity, which can occur in both thrombotic microangiopathies and acute ischemic stroke. Recently, NETs have been envisioned as a cause of endothelial damage and immunothrombosis in COVID-19. In addition, vWF and ADAMTS-13 levels predict COVID-19 mortality. In this review, we summarize the biological characteristics and interactions of NETs, vWF, and ADAMTS-13, the effect of NETs on hemostasis regulation and discuss their role in thrombotic conditions, sepsis, COVID-19, and obstetric complications.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - A. S. Shkoda
- Vorokhobov City Clinical Hospital № 67, Moscow Healthcare Department
| | - L. L. Pankratyeva
- Vorokhobov City Clinical Hospital № 67, Moscow Healthcare Department; Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Health Ministry of Russian Federation
| | - G. C. Di Renzo
- Sechenov University; Center for Prenatal and Reproductive Medicine, University of Perugia
| | - G. Rizzo
- Sechenov University; University of Rome Tor Vergata
| | | | - V. I. Tsibizova
- Almazov National Medical Research Centre, Health Ministry of Russian Federation
| | - J.-C. Gris
- Sechenov University; University of Montpellier
| | - I. Elalamy
- Sechenov University; Medicine Sorbonne University; Hospital Tenon
| |
Collapse
|
43
|
Shaw RJ, Abrams ST, Austin J, Taylor JM, Lane S, Dutt T, Downey C, Du M, Turtle L, Baillie JK, Openshaw PJM, Wang G, Semple MG, Toh CH. Circulating histones play a central role in COVID-19-associated coagulopathy and mortality. Haematologica 2021; 106:2493-2498. [PMID: 33832219 PMCID: PMC8409036 DOI: 10.3324/haematol.2021.278492] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Indexed: 12/14/2022] Open
Affiliation(s)
- Rebecca J Shaw
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, UK; Roald Dahl Haemostasis and Thrombosis Centre, Liverpool University Hospitals NHS Foundation Trust, Liverpool
| | - Simon T Abrams
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool
| | - James Austin
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool
| | - Joseph M Taylor
- Liverpool Clinical Laboratories, Liverpool University Hospitals NHS Foundation Trust, Liverpool
| | - Steven Lane
- Department of Biostatistics, University of Liverpool, Liverpool
| | - Tina Dutt
- Roald Dahl Haemostasis and Thrombosis Centre, Liverpool University Hospitals NHS Foundation Trust, Liverpool
| | - Colin Downey
- Liverpool Clinical Laboratories, Liverpool University Hospitals NHS Foundation Trust, Liverpool
| | - Min Du
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool
| | - Lance Turtle
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, UK; Infectious Diseases Unit, Royal Liverpool University Hospital, Liverpool
| | - J Kenneth Baillie
- Roslin Institute, University of Edinburgh, Edinburgh, UK; Intensive Care Unit, Royal Infirmary Edinburgh, Edinburgh
| | | | - Guozheng Wang
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool
| | - Malcolm G Semple
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, UK; Respiratory Medicine, Alder Hey Children's Hospital NHS Foundation Trust, Liverpool
| | - Cheng-Hock Toh
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, UK; Roald Dahl Haemostasis and Thrombosis Centre, Liverpool University Hospitals NHS Foundation Trust, Liverpool.
| |
Collapse
|
44
|
Shen Y, Zhang Y, Li W, Chen K, Xiang M, Ma H. Glutamine metabolism: from proliferating cells to cardiomyocytes. Metabolism 2021; 121:154778. [PMID: 33901502 DOI: 10.1016/j.metabol.2021.154778] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 04/16/2021] [Accepted: 04/19/2021] [Indexed: 02/06/2023]
Abstract
Glutamine is a major energy source for rapidly dividing cells, such as hematopoietic stem cells and cancer cells. Reliance on glutamine is therefore regarded as a metabolic hallmark of proliferating cells. Moreover, reprogramming glutamine metabolism by various factors, including tissue type, microenvironment, pro-oncogenes, and tumor suppressor genes, can facilitate stem cell fate decisions, tumor recurrence, and drug resistance. However, the significance of glutamine metabolism in cardiomyocytes, an end-differentiated cell type, is not fully understood. Existing evidence suggests important roles of glutamine metabolism in the development of cardiovascular diseases. In this review, we have focused on glutaminolysis and its regulatory network in proliferating cells. We have summarized current findings about the role of glutamine utilization in cardiomyocytes and have discussed possibilities of targeting glutamine metabolism for the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Yimin Shen
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Yuhao Zhang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Wudi Li
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Kaijie Chen
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Meixiang Xiang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China.
| | - Hong Ma
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China.
| |
Collapse
|
45
|
Suhaimi NF, Jalaludin J, Abu Bakar S. The Influence of Traffic-Related Air Pollution (TRAP) in Primary Schools and Residential Proximity to Traffic Sources on Histone H3 Level in Selected Malaysian Children. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18157995. [PMID: 34360284 PMCID: PMC8345469 DOI: 10.3390/ijerph18157995] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/12/2021] [Accepted: 07/21/2021] [Indexed: 02/07/2023]
Abstract
This study aimed to investigate the association between traffic-related air pollution (TRAP) exposure and histone H3 modification among school children in high-traffic (HT) and low-traffic (LT) areas in Malaysia. Respondents' background information and personal exposure to traffic sources were obtained from questionnaires distributed to randomly selected school children. Real-time monitoring instruments were used for 6-h measurements of PM10, PM2.5, PM1, NO2, SO2, O3, CO, and total volatile organic compounds (TVOC). Meanwhile, 24-h measurements of PM2.5-bound black carbon (BC) were performed using air sampling pumps. The salivary histone H3 level was captured using an enzyme-linked immunosorbent assay (ELISA). HT schools had significantly higher PM10, PM2.5, PM1, BC, NO2, SO2, O3, CO, and TVOC than LT schools, all at p < 0.001. Children in the HT area were more likely to get higher histone H3 levels (z = -5.13). There were positive weak correlations between histone H3 level and concentrations of NO2 (r = 0.37), CO (r = 0.36), PM1 (r = 0.35), PM2.5 (r = 0.34), SO2 (r = 0.34), PM10 (r = 0.33), O3 (r = 0.33), TVOC (r = 0.25), and BC (r = 0.19). Overall, this study proposes the possible role of histone H3 modification in interpreting the effects of TRAP exposure via non-genotoxic mechanisms.
Collapse
Affiliation(s)
- Nur Faseeha Suhaimi
- Department of Environmental and Occupational Health, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia;
| | - Juliana Jalaludin
- Department of Environmental and Occupational Health, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia;
- Department of Occupational Health and Safety, Faculty of Public Health, Universitas Airlangga, Surabaya 60115, Indonesia
- Correspondence: ; Tel.: +603-97692401
| | - Suhaili Abu Bakar
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia;
| |
Collapse
|
46
|
Early Vitamin C, Hydrocortisone, and Thiamine Treatment for Septic Cardiomyopathy: A Propensity Score Analysis. J Pers Med 2021; 11:jpm11070610. [PMID: 34203183 PMCID: PMC8304272 DOI: 10.3390/jpm11070610] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/22/2021] [Accepted: 06/25/2021] [Indexed: 12/29/2022] Open
Abstract
This study investigated the effectiveness of early vitamin C, hydrocortisone, and thiamine among patients with septic cardiomyopathy. In total, 91 patients with septic cardiomyopathy received a vitamin C protocol in September 2018–February 2020. These patients were compared to 75 patients with septic cardiomyopathy who did not receive a vitamin C protocol in September 2016–February 2018. Relative to the control patients, the treated patients were older and more likely to require mechanical ventilation. The vitamin C protocol was associated with a lower risk of intensive care unit mortality in the propensity score (PS)-matched cohort (aHR: 0.55, 95% CI: 0.30–0.99) and inverse probability of treatment weighting-matched cohort (aHR: 0.67, 95% CI: 0.45–1.00). In the PS-matched cohort (59 patients per group), the vitamin C protocol was associated with decreased values for vasopressor dosage, C-reactive protein concentration, and the Sequential Organ Failure Assessment score during the 4-day treatment period. Patients who started the vitamin C protocol within 2 h after diagnosis (vs. 2–8 h or ≥8 h) had the highest vasopressor weaning rate and the lowest mortality rate. These results suggest that early treatment using a vitamin C protocol might improve organ dysfunction and reduce mortality among patients with septic cardiomyopathy.
Collapse
|
47
|
Moxon C, Alhamdi Y, Storm J, Toh J, Ko JY, Murphy G, Taylor T, Seydel K, Wang G, García-Car G, Molyneux M, Craig A, Abrams S, Toh CH. Parasite histones mediate blood-brain barrier disruption in cerebral malaria. Clin Med (Lond) 2021; 20:s96-s97. [PMID: 32409404 DOI: 10.7861/clinmed.20-2-s96] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
| | | | - Janet Storm
- Liverpool School of Tropical Medicine, Liverpool, UK
| | - Julien Toh
- University of Sheffield Medical School, Sheffield, UK
| | - Joo Yeon Ko
- Hanyang University College of Medicine, Seoul, South Korea
| | | | | | - Karl Seydel
- Michigan State University, East Lansing, USA
| | | | | | | | - Alister Craig
- Liverpool School of Tropical Medicine, Liverpool, UK
| | | | | |
Collapse
|
48
|
Parasite histones are toxic to brain endothelium and link blood barrier breakdown and thrombosis in cerebral malaria. Blood Adv 2021; 4:2851-2864. [PMID: 32579667 DOI: 10.1182/bloodadvances.2019001258] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 05/17/2020] [Indexed: 12/16/2022] Open
Abstract
Microvascular thrombosis and blood-brain barrier (BBB) breakdown are key components of cerebral malaria (CM) pathogenesis in African children and are implicated in fatal brain swelling. How Plasmodium falciparum infection causes this endothelial disruption and why this occurs, particularly in the brain, is not fully understood. In this study, we have demonstrated that circulating extracellular histones, equally of host and parasite origin, are significantly elevated in CM patients. Higher histone levels are associated with brain swelling on magnetic resonance imaging. On postmortem brain sections of CM patients, we found that histones are colocalized with P falciparum-infected erythrocytes sequestered inside small blood vessels, suggesting that histones might be expelled locally during parasite schizont rupture. Histone staining on the luminal vascular surface colocalized with thrombosis and leakage, indicating a possible link between endothelial surface accumulation of histones and coagulation activation and BBB breakdown. Supporting this, patient sera or purified P falciparum histones caused disruption of barrier function and were toxic to cultured human brain endothelial cells, which were abrogated with antihistone antibody and nonanticoagulant heparin. Overall, our data support a role for histones of parasite and host origin in thrombosis, BBB breakdown, and brain swelling in CM, processes implicated in the causal pathway to death. Neutralizing histones with agents such as nonanticoagulant heparin warrant exploration to prevent brain swelling in the development or progression of CM and thereby to improve outcomes.
Collapse
|
49
|
Moiana M, Aranda F, de Larrañaga G. A focus on the roles of histones in health and diseases. Clin Biochem 2021; 94:12-19. [PMID: 33895124 DOI: 10.1016/j.clinbiochem.2021.04.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/13/2021] [Accepted: 04/18/2021] [Indexed: 11/28/2022]
Abstract
Over time, the knowledge on the role of histones has significantly changed. Initially, histones were only known as DNA packaging proteins but later, it was discovered that they act extracellularly as powerful antimicrobial agents and also as potentially self-detrimental agents. Indeed, histones were found to be the most abundant proteins within neutrophil extracellular traps what ultimately highlighted their microbicidal function. In addition, extracellular histones proved to be involved in triggering exacerbated inflammatory and coagulation responses, depending on the cell type affected. Consequently, several investigations were conducted towards studying the potential of histones and their derivatives as either biomarkers or therapeutic target candidates in different diseases in which inflammation and thrombosis have a key pathophysiological role, such as sepsis, thrombosis and different types of cancer. The main objective of this review is to summarize and discuss the current state of the art with regard to both beneficial and harmful roles of histones and also their possible use as biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Mauro Moiana
- Hemostasis and Thrombosis Laboratory, Hospital of Infectious Diseases "F. J. Muñiz", Buenos Aires, Argentina
| | - Federico Aranda
- Hemostasis and Thrombosis Laboratory, Hospital of Infectious Diseases "F. J. Muñiz", Buenos Aires, Argentina
| | - Gabriela de Larrañaga
- Hemostasis and Thrombosis Laboratory, Hospital of Infectious Diseases "F. J. Muñiz", Buenos Aires, Argentina.
| |
Collapse
|
50
|
Li Y, Wan D, Luo X, Song T, Wang Y, Yu Q, Jiang L, Liao R, Zhao W, Su B. Circulating Histones in Sepsis: Potential Outcome Predictors and Therapeutic Targets. Front Immunol 2021; 12:650184. [PMID: 33868288 PMCID: PMC8044749 DOI: 10.3389/fimmu.2021.650184] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 03/11/2021] [Indexed: 02/05/2023] Open
Abstract
Sepsis is defined as a life-threatening organ dysfunction caused by a dysregulated host response to infection and is associated with high morbidity and mortality. Circulating histones (CHs), a group of damage-associated molecular pattern molecules mainly derived from neutrophil extracellular traps, play a crucial role in sepsis by mediating inflammation response, organ injury and death through Toll-like receptors or inflammasome pathways. Herein, we first elucidate the molecular mechanisms of histone-induced inflammation amplification, endothelium injury and cascade coagulation activation, and discuss the close correlation between elevated level of CHs and disease severity as well as mortality in patients with sepsis. Furthermore, current state-of-the-art on anti-histone therapy with antibodies, histone-binding proteins (namely recombinant thrombomodulin and activated protein C), and heparin is summarized to propose promising approaches for sepsis treatment.
Collapse
Affiliation(s)
- Yupei Li
- Department of Nephrology of West China Hospital, Institute for Disaster Management and Reconstruction, Sichuan University, Chengdu, China.,Department of Emergency Medicine of West China Hospital, Disaster Medical Center, Sichuan University, Chengdu, China.,Med-X Center for Materials, Sichuan University, Chengdu, China
| | - Dingyuan Wan
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Xinyao Luo
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Tao Song
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, China
| | - Yiran Wang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, China
| | - Qiao Yu
- Department of Nephrology of West China Hospital, Institute for Disaster Management and Reconstruction, Sichuan University, Chengdu, China.,Department of Emergency Medicine of West China Hospital, Disaster Medical Center, Sichuan University, Chengdu, China.,Med-X Center for Materials, Sichuan University, Chengdu, China
| | - Luojia Jiang
- Department of Nephrology of West China Hospital, Institute for Disaster Management and Reconstruction, Sichuan University, Chengdu, China
| | - Ruoxi Liao
- Department of Nephrology of West China Hospital, Institute for Disaster Management and Reconstruction, Sichuan University, Chengdu, China
| | - Weifeng Zhao
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, China
| | - Baihai Su
- Department of Nephrology of West China Hospital, Institute for Disaster Management and Reconstruction, Sichuan University, Chengdu, China.,Department of Emergency Medicine of West China Hospital, Disaster Medical Center, Sichuan University, Chengdu, China.,Med-X Center for Materials, Sichuan University, Chengdu, China.,West China School of Medicine, Sichuan University, Chengdu, China
| |
Collapse
|