1
|
Sun Y, Pu Z, Zhao H, Deng Y, Zhang J, Li S, Jiang Y, Sun M, Zhu J, Alam A, Ma D, Han R. Vitamin D can mitigate sepsis-associated neurodegeneration by inhibiting exogenous histone-induced pyroptosis and ferroptosis: Implications for brain protection and cognitive preservation. Brain Behav Immun 2025; 124:40-54. [PMID: 39566666 DOI: 10.1016/j.bbi.2024.11.019] [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: 07/06/2024] [Revised: 11/14/2024] [Accepted: 11/17/2024] [Indexed: 11/22/2024] Open
Abstract
BACKGROUND Sepsis-induced neurodegeneration and cognitive dysfunction remain critical challenges worldwide. Vitamin D was reported to reduce neuronal injury and neurotoxicity and its deficiency was associated with neurocognitive disorders. This study investigates the mechanisms by which vitamin D exerts neuroprotective potential against damage-associated molecular patterns (DAMPs), specifically extracellular histones, in sepsis-related brain dysfunction. METHODS The cultured mouse hippocampal neuronal HT22 cells were exposed to 20 µg/ml exogenous histone for 24 h to induce pyroptosis and ferroptosis in the presence or absence of the active form of vitamin D, calcitriol (1 nM). A cecal ligation and puncture mouse sepsis model was used to evaluate histone release and pyroptosis/ferroptosis biomarkers in the brain together with neurobehavioral performance with or without calcitriol treatment (1 µg/kg, i.p. injection) at 24 h or 1 week after sepsis onset. RESULTS In vitro, histone exposure triggered both pyroptosis and ferroptosis in neuronal cells, which was significantly suppressed by calcitriol treatment with the reduced expression of caspase-1 by 38 %, GSDMD by 30 %, ACSL4 by 33 %, and the increased expression of GPX4 by 35 % (n = 6, P < 0.05). Similarly, in vivo, calcitriol treatment inhibited both neuronal pyroptosis and ferroptosis by reducing expression of pyroptosis marker, GSDMD/NeuN (11.6 ± 1.2 % vs. 19.4 ± 1.1 %) and increasing expression of ferroptosis marker, GPX4/NeuN (21.4 ± 1.7 % vs. 13.5 ± 1.1 %), in the brain of septic mice (n = 6, P < 0.01). In addition, calcitriol increased survival rate (72 % vs. 41 %) and ameliorated cognitive dysfunction of septic mice (n = 8-13, P < 0.05). CONCLUSIONS This study demonstrates that vitamin D exerts a neuroprotective effect against sepsis by attenuating histone-induced pyroptosis and ferroptosis. These findings highlight the potential therapeutic role of vitamin D supplementation in mitigating brain dysfunction associated with sepsis which needs for further investigation.
Collapse
Affiliation(s)
- Yibing Sun
- Department of Anesthesiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, PR China
| | - Zhuonan Pu
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, PR China
| | - Hailin Zhao
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London, UK
| | - Yuxuan Deng
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, PR China
| | - Jing Zhang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, PR China
| | - Shiwei Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, PR China
| | - Yingying Jiang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, PR China
| | - Ming Sun
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, PR China
| | - Jinpiao Zhu
- Perioperative and Systems Medicine Laboratory, Department of Anesthesiology and Rehabilitation, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Zhejiang, PR China
| | - Azeem Alam
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London, UK
| | - Daqing Ma
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London, UK; Perioperative and Systems Medicine Laboratory, Department of Anesthesiology and Rehabilitation, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Zhejiang, PR China.
| | - Ruquan Han
- Department of Anesthesiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, PR China.
| |
Collapse
|
2
|
Loaiza R, Fattahi F, Kalbitz M, Grailer JJ, Russell MW, Jalife J, Valdivia HH, Zetoune FS, Ward PA. The Impact of Extracellular Histones and Absence of Toll-like Receptors on Cardiac Functional and Electrical Disturbances in Mouse Hearts. Int J Mol Sci 2024; 25:8653. [PMID: 39201339 PMCID: PMC11354419 DOI: 10.3390/ijms25168653] [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: 03/29/2024] [Revised: 08/02/2024] [Accepted: 08/04/2024] [Indexed: 09/02/2024] Open
Abstract
In polymicrobial sepsis, the extracellular histones, mainly released from activated neutrophils, significantly contribute to cardiac dysfunction (septic cardiomyopathy), as demonstrated in our previous studies using Echo-Doppler measurements. This study aims to elucidate the roles of extracellular histones and their interactions with Toll-like receptors (TLRs) in cardiac dysfunction. Through ex vivo assessments of ECG, left ventricle (LV) function parameters, and in vivo Echo-Doppler studies in mice perfused with extracellular histones, we aim to provide comprehensive insights into the mechanisms underlying sepsis-induced cardiac dysfunction. Langendorff-perfused hearts from both wild-type and TLR2, TLR3, or TLR4 knockout (KO) mice were examined. Paced mouse hearts were perfused with histones to assess contractility and relaxation. Echo-Doppler studies evaluated cardiac dysfunction after intravenous histone injection. Histone perfusion caused defects in contractility and relaxation, with TLR2 and TLR3 KO mice being partially protected. Specifically, TLR2 KO mice exhibited the greatest reduction in Echo-Doppler abnormalities, while TLR4 KO exacerbated cardiac dysfunction. Among individual histones, H1 induced the most pronounced abnormalities in cardiac function, apoptosis of cardiomyocytes, and LDH release. Our data highlight significant interactions between histones and TLRs, providing insights into histones especially H1 as potential therapeutic targets for septic cardiomyopathy. Further studies are needed to explore specific histone-TLR interactions and their mechanisms.
Collapse
Affiliation(s)
- Randall Loaiza
- Center for Arrhythmia Research, University of Michigan, Ann Arbor, MI 48109, USA; (R.L.); (J.J.); (H.H.V.)
- CENIBiot Laboratory, The National Center of High Technology (CeNAT-CONARE), San José 10109, Costa Rica
| | - Fatemeh Fattahi
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA;
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; (M.K.); (J.J.G.); (F.S.Z.)
| | - Miriam Kalbitz
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; (M.K.); (J.J.G.); (F.S.Z.)
- Department of Orthopaedic Trauma, Hand, Plastic and Reconstructive Surgery, University Hospital of Ulm, 89081 Ulm, Germany
- Military Medical City Hospital, Doha 486441, Qatar
| | - Jamison J. Grailer
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; (M.K.); (J.J.G.); (F.S.Z.)
- Integrated Biology R&D, Bioassay Development, Promega Corporation, Madison, WI 53711, USA
| | - Mark W. Russell
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI 48109, USA;
| | - Jose Jalife
- Center for Arrhythmia Research, University of Michigan, Ann Arbor, MI 48109, USA; (R.L.); (J.J.); (H.H.V.)
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
| | - Hector H. Valdivia
- Center for Arrhythmia Research, University of Michigan, Ann Arbor, MI 48109, USA; (R.L.); (J.J.); (H.H.V.)
- Department of Medicine, Cardiovascular Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Firas S. Zetoune
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; (M.K.); (J.J.G.); (F.S.Z.)
| | - Peter A. Ward
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; (M.K.); (J.J.G.); (F.S.Z.)
| |
Collapse
|
3
|
Zhong T, Chen S, Deng K, Guan J, Zhang J, Lu F, Shichen M, Lv R, Liu Z, Liu Y, Chang P, Liu Z. Magnesium alleviates extracellular histone-induced apoptosis and defective bacterial phagocytosis in macrophages by regulating intracellular calcium signal. Int Immunopharmacol 2024; 132:111870. [PMID: 38547771 DOI: 10.1016/j.intimp.2024.111870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 02/18/2024] [Accepted: 03/12/2024] [Indexed: 05/01/2024]
Abstract
Extracellular histones have been determined as important mediators of sepsis, which induce excessive inflammatory responses in macrophages and impair innate immunity. Magnesium (Mg2+), one of the essential nutrients of the human body, contributes to the proper regulation of immune function. However, no reports indicate whether extracellular histones affect survival and bacterial phagocytosis in macrophages and whether Mg2+ is protective against histone-induced macrophage damage. Our clinical data revealed a negative correlation between circulating histone and monocyte levels in septic patients, and in vitro experiments confirmed that histones induced mitochondria-associated apoptosis and defective bacterial phagocytosis in macrophages. Interestingly, our clinical data also indicated an association between lower serum Mg2+ levels and reduced monocyte levels in septic patients. Moreover, in vitro experiments demonstrated that Mg2+ attenuated histone-induced apoptosis and defective bacterial phagocytosis in macrophages through the PLC/IP3R/STIM-mediated calcium signaling pathway. Importantly, further animal experiments proved that Mg2+ significantly improved survival and attenuated histone-mediated lung injury and macrophage damage in histone-stimulated mice. Additionally, in a cecal ligation and puncture (CLP) + histone-induced injury mouse model, Mg2+ inhibited histone-mediated apoptosis and defective phagocytosis in macrophages and further reduced bacterial load. Overall, these results suggest that Mg2+ supplementation may be a promising treatment for extracellular histone-mediated macrophage damage in sepsis.
Collapse
Affiliation(s)
- Tao Zhong
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Sainan Chen
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Ke Deng
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jianbin Guan
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jiaqi Zhang
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Furong Lu
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Maoyou Shichen
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Ronggui Lv
- Department of Intensive Care Unit, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China
| | - Zhifeng Liu
- Department of Medicine Intensive Care Units, General Hospital of Southern Theatre Command of PLA, Guangzhou, Guangdong, China.
| | - Yong Liu
- Department of Intensive Care Unit, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China.
| | - Ping Chang
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| | - Zhanguo Liu
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
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
|
7
|
Liao YE, Liu J, Arnold K. Heparan sulfates and heparan sulfate binding proteins in sepsis. Front Mol Biosci 2023; 10:1146685. [PMID: 36865384 PMCID: PMC9971734 DOI: 10.3389/fmolb.2023.1146685] [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: 01/17/2023] [Accepted: 01/31/2023] [Indexed: 02/16/2023] Open
Abstract
Heparan sulfates (HSs) are the main components in the glycocalyx which covers endothelial cells and modulates vascular homeostasis through interactions with multiple Heparan sulfate binding proteins (HSBPs). During sepsis, heparanase increases and induces HS shedding. The process causes glycocalyx degradation, exacerbating inflammation and coagulation in sepsis. The circulating heparan sulfate fragments may serve as a host defense system by neutralizing dysregulated Heparan sulfate binding proteins or pro-inflammatory molecules in certain circumstances. Understanding heparan sulfates and heparan sulfate binding proteins in health and sepsis is critical to decipher the dysregulated host response in sepsis and advance drug development. In this review, we will overview the current understanding of HS in glycocalyx under septic condition and the dysfunctional heparan sulfate binding proteins as potential drug targets, particularly, high mobility group box 1 (HMGB1) and histones. Moreover, several drug candidates based on heparan sulfates or related to heparan sulfates, such as heparanase inhibitors or heparin-binding protein (HBP), will be discussed regarding their recent advances. By applying chemical or chemoenzymatic approaches, the structure-function relationship between heparan sulfates and heparan sulfate binding proteins is recently revealed with structurally defined heparan sulfates. Such homogenous heparan sulfates may further facilitate the investigation of the role of heparan sulfates in sepsis and the development of carbohydrate-based therapy.
Collapse
Affiliation(s)
- Yi-En Liao
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, United States
| | - Jian Liu
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, United States
| | | |
Collapse
|
8
|
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
|
9
|
Yin L, Tang Y, Lin X, Jiang B. Progress in the mechanism of mitochondrial dysfunction in septic cardiomyopathy. ALL LIFE 2022. [DOI: 10.1080/26895293.2022.2156622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Leijing Yin
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, People’s Republic of China
- Sepsis Translational Medicine Key Lab of Hunan Province, Hunan, People’s Republic of China
- National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan, People’s Republic of China
| | - Yuting Tang
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, People’s Republic of China
- Sepsis Translational Medicine Key Lab of Hunan Province, Hunan, People’s Republic of China
- National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan, People’s Republic of China
| | - Xiaofang Lin
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, People’s Republic of China
- Sepsis Translational Medicine Key Lab of Hunan Province, Hunan, People’s Republic of China
- National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan, People’s Republic of China
| | - Bimei Jiang
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, People’s Republic of China
- Sepsis Translational Medicine Key Lab of Hunan Province, Hunan, People’s Republic of China
- National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan, People’s Republic of China
| |
Collapse
|
10
|
Extracellular histones trigger oxidative stress-dependent induction of the NF-kB/CAM pathway via TLR4 in endothelial cells. J Physiol Biochem 2022:10.1007/s13105-022-00935-z. [DOI: 10.1007/s13105-022-00935-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 11/24/2022] [Indexed: 12/12/2022]
Abstract
Abstract
Extracellular histones have been reported to aggravate different pathophysiological processes by increasing vascular permeability, coagulopathy, and inflammation. In the present study, we elucidate how extracellular histones (10–100 µg/mL) concentration dependently increase cytosolic reactive oxygen species (ROS) production using human umbilical vein endothelial cells (HUVECs). Furthermore, we identify cyclooxygenase (COX) and NADPH oxidase (NOX) activity as sources of ROS production in extracellular histone-treated HUVEC. This COX/NOX-mediated ROS production is also involved in enhanced NF-kB activity and cell adhesion molecules (VCAM1 and ICAM1) expression in histone-treated HUVEC. Finally, by using different toll-like receptor (TLR) antagonists, we demonstrate the role of TLR4 in CAMs overexpression triggered by extracellular histones in endothelial cells. In conclusion, our data suggest that through TLR4 signaling, extracellular histones increase endothelial cell activation, a mechanism involving increased COX- and NOX-mediated ROS. These findings increase our understanding on how extracellular histones enhance systemic inflammatory responses in diseases in which histone release occurs as part of the pathological processes.
Collapse
|
11
|
Role of neutrophil extracellular traps in inflammatory evolution in severe acute pancreatitis. Chin Med J (Engl) 2022; 135:2773-2784. [PMID: 36729096 PMCID: PMC9945416 DOI: 10.1097/cm9.0000000000002359] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Indexed: 02/03/2023] Open
Abstract
ABSTRACT Severe acute pancreatitis (SAP) is a life-threatening acute abdominal disease with two peaks of death: the first in the early stage, characterized by systemic inflammatory response-associated organ failure; and the second in the late stage, characterized by infectious complications. Neutrophils are the main immune cells participating in the whole process of SAP. In addition to the traditional recognition of neutrophils as the origination of chemokine and cytokine cascades or phagocytosis and degranulation of pathogens, neutrophil extracellular traps (NETs) also play an important roles in inflammatory reactions. We reviewed the role of NETs in the occurrence and development of SAP and its fatal complications, including multiple organs injury, infected pancreatic necrosis, and thrombosis. This review provides novel insights into the involvement of NETs throughout the entire process of SAP, showing that targeting NETs might be a promising strategy in SAP treatment. However, precision therapeutic options targeting NETs in different situations require further investigation.
Collapse
|
12
|
Listeria monocytogenes Infection Alters the Content and Function of Extracellular Vesicles Produced by Trophoblast Stem Cells. Infect Immun 2022; 90:e0034722. [PMID: 36154271 DOI: 10.1128/iai.00347-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Placental immunity is critical for fetal health during pregnancy, as invading pathogens spread from the parental blood to the fetus through this organ. However, inflammatory responses in the placenta can adversely affect both the fetus and the pregnant person, and the balance between protective placental immune response and detrimental inflammation is poorly understood. Extracellular vesicles (EVs) are membrane-enclosed vesicles that play a critical role in placental immunity. EVs produced by placental trophoblasts mediate immune tolerance to the fetus and to the placenta itself, but these EVs can also activate detrimental inflammatory responses. The regulation of these effects is not well characterized, and the role of trophoblast EVs (tEVs) in the response to infection has yet to be defined. The Gram-positive bacterial pathogen Listeria monocytogenes infects the placenta, serving as a model to study tEV function in this context. We investigated the effect of L. monocytogenes infection on the production and function of tEVs, using a trophoblast stem cell (TSC) model. We found that tEVs from infected TSCs can induce the production of the proinflammatory cytokine tumor necrosis factor alpha (TNF-α) in recipient cells. Surprisingly, this tEV treatment could confer increased susceptibility to subsequent L. monocytogenes infection, which has not been reported previously as an effect of EVs. Proteomic analysis and RNA sequencing revealed that tEVs from infected TSCs had altered cargo compared with those from uninfected TSCs. However, no L. monocytogenes proteins were detected in tEVs from infected TSCs. Together, these results suggest an immunomodulatory role for tEVs during prenatal infection.
Collapse
|
13
|
Lackner I, Weber B, Pressmar J, Odwarka A, Lam C, Haffner-Luntzer M, Marcucio R, Miclau T, Kalbitz M. Cardiac alterations following experimental hip fracture - inflammaging as independent risk factor. Front Immunol 2022; 13:895888. [PMID: 36131923 PMCID: PMC9484325 DOI: 10.3389/fimmu.2022.895888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 08/08/2022] [Indexed: 12/04/2022] Open
Abstract
Background Cardiac injuries following trauma are associated with a worse clinical outcome. So-called trauma-induced secondary cardiac injuries have been recently described after experimental long bone fracture even in absence of direct heart damage. With the progressive aging of our society, the number of elderly trauma victims rises and therefore the incidence of hip fractures increases. Hip fractures were previously shown to be associated with adverse cardiac events in elderly individuals, which have mainly been attributed to pre-conditioned cardiac diseases. The aim of the present study was to investigate the effect of hip fractures on the heart in healthy young and middle-aged mice. Materials and Methods Young (12-week-old) and middle-aged (52-week-old) female C57BL/6 mice either received an intramedullary stabilized proximal femur fracture or sham treatment. The observation time points included 6 and 24 h. Systemic levels of pro-inflammatory mediators as well as local inflammation and alterations in myocardial structure, metabolism and calcium homeostasis in left ventricular tissue was analyzed following hip fracture by multiplex analysis, RT-qPCR and immunohistochemistry. Results After hip fracture young and middle-aged mice showed increased systemic IL-6 and KC levels, which were significantly elevated in the middle-aged animals. Furthermore, the middle-aged mice showed enhanced myocardial expression of HMGB1, TLR2/4, TNF, IL1β and NLRP3 as well as considerable alterations in the myocardial expression of glucose- and fatty acid transporters (HFABP, GLUT4), calcium homeostasis proteins (SERCA) and cardiac structure proteins (desmin, troponin I) compared to the young animals following hip fracture. Conclusion Young and middle-aged mice showed local myocardial alterations, which might predispose for the development of secondary cardiac injury following hip fracture. Age and the age-associated phenomenon of ‘inflammaging’ seemed to be an independent risk factor aggravating and accelerating cardiac alterations following hip fracture.
Collapse
Affiliation(s)
- Ina Lackner
- Department of Trauma and Orthopedic Surgery, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
- Department of Traumatology, Hand, Plastic, and Reconstructive Surgery, University Medical Center Ulm, Ulm, Germany
| | - Birte Weber
- Department of Traumatology, Hand, Plastic, and Reconstructive Surgery, University Medical Center Ulm, Ulm, Germany
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, Zuckerberg San Francisco General Hospital, University of California, San Francisco, San Francisco, CA, United States
- Department of Trauma, Hand and Reconstructive Surgery, Goethe University of Frankfurt, Frankfurt, Germany
| | - Jochen Pressmar
- Department of Trauma and Orthopedic Surgery, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
- Department of Traumatology, Hand, Plastic, and Reconstructive Surgery, University Medical Center Ulm, Ulm, Germany
| | - Anna Odwarka
- Department of Traumatology, Hand, Plastic, and Reconstructive Surgery, University Medical Center Ulm, Ulm, Germany
| | - Charles Lam
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, Zuckerberg San Francisco General Hospital, University of California, San Francisco, San Francisco, CA, United States
| | - Melanie Haffner-Luntzer
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, Zuckerberg San Francisco General Hospital, University of California, San Francisco, San Francisco, CA, United States
- Institute of Orthopaedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Ralph Marcucio
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, Zuckerberg San Francisco General Hospital, University of California, San Francisco, San Francisco, CA, United States
| | - Theodore Miclau
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, Zuckerberg San Francisco General Hospital, University of California, San Francisco, San Francisco, CA, United States
| | - Miriam Kalbitz
- Department of Trauma and Orthopedic Surgery, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
- Department of Traumatology, Hand, Plastic, and Reconstructive Surgery, University Medical Center Ulm, Ulm, Germany
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, Zuckerberg San Francisco General Hospital, University of California, San Francisco, San Francisco, CA, United States
- *Correspondence: Miriam Kalbitz,
| |
Collapse
|
14
|
Xue W, Pang J, Liu J, Wang H, Guo H, Chen Y. Septic cardiomyopathy: characteristics, evaluation, and mechanism. EMERGENCY AND CRITICAL CARE MEDICINE 2022; 2:135-147. [DOI: 10.1097/ec9.0000000000000060] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
Abstract
Sepsis is a common clinical disease; if there is no early active treatment, it is likely to develop into multiple organ dysfunction syndrome and even cause death. Septic cardiomyopathy is a complication of sepsis-related cardiovascular failure, characterized by reversible left ventricular dilatation and decreased ventricular systolic and/or diastolic function. At present, echocardiography and biomarkers are often used to screen septic cardiomyopathy in clinics. Although there is still a lack of clear diagnostic criteria for septic cardiomyopathy, according to existing studies, the pathogenesis of several septic cardiomyopathy has been clarified, such as immune response caused by infection and mitochondrial dysfunction. This review summarizes the characteristics, pathophysiology, and diagnosis of septic cardiomyopathy and focuses on the mechanisms of infection immunity and mitochondrial dysfunction.
Collapse
Affiliation(s)
| | | | - Jiao Liu
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | | | | |
Collapse
|
15
|
Celani LMS, Egito EST, Azevedo ÍM, Oliveira CN, Dourado D, Medeiros AC. Treatment of colitis by oral negatively charged nanostructured curcumin in rats. Acta Cir Bras 2022; 37:e370602. [PMID: 35976279 PMCID: PMC9377652 DOI: 10.1590/acb370602] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 04/08/2022] [Accepted: 05/03/2022] [Indexed: 12/05/2022] Open
Abstract
PURPOSE To examine the effects of a negatively charged nanostructured curcumin microemulsion in experimental ulcerative colitis (UC) in rats. METHODS Four percent acetic acid was used to induce UC. The animals were treated for seven days and randomly assigned to four groups: normal control (NC), colitis/normal saline (COL/NS), colitis/curcumin (COL/CUR), and colitis/mesalazine (COL/MES). The nanostructured curcumin was formulated with a negative zeta potential (-16.70 ± 1.66 mV). Dosage of the pro-inflammatory cytokines tumor necrosis factor-α (TNF-α), interleukin 1-β (IL-1β), interleukin 6 (IL-6), and antioxidant enzymes (catalase, superoxide dismutase, and glutathione peroxidase), macro and microscopic evaluation of the colon tissue were analyzed. RESULTS The COL/CUR group had a higher level of antioxidant enzymes compared to the COL/MESgroup. The levels of TNF-α, IL-1β and IL-6 were significantly lower in the colonic tissue of the COL/CUR group rats, when compared to the COL/NS and COL/MES groups (p < 0.001). The presence of ulcers in the colonic mucosa in rats of the COL/NSgroup was significantly higher than in the COL/MES group (p < 0.001). In the NC and COL/CUR groups, there were no ulcers in the colonic mucosa. CONCLUSIONS The nanostructured microemulsion of curcumin, used orally, positively influenced the results of the treatment of UC in rats. The data also suggests that nanostructured curcumin with negative zeta potential is a promising phytopharmaceutical oral delivery system for UC therapy. Further research needs to be done to better understand the mechanisms of the negatively charged nanostructured curcumin microemulsion in UC therapy.
Collapse
Affiliation(s)
- Lívia Medeiros Soares Celani
- Fellow master degree. Universidade Federal do Rio Grande do Norte – Postgraduate Program in Health Sciences – Natal (RN), Brazil
| | - Eryvaldo Sócrates Tabosa Egito
- PhD, full professor, chairman. Universidade Federal do Rio Grande do Norte – Laboratory of Dispersed Systems – Natal (RN), Brazil
| | | | - Cláudia Nunes Oliveira
- PhD. Universidade Federal do Rio Grande do Norte – Pathology Department – Health Sciences – Natal (RN), Brazil
| | - Douglas Dourado
- Fellow PhD degree. Universidade Federal do Rio Grande do Norte – Postgraduate Program in Health Sciences – Natal (RN), Brazil
| | - Aldo Cunha Medeiros
- PhD, full professor, chairman. Universidade Federal do Rio Grande do Norte – Nucleus of Experimental Surgery – Natal (RN), Brazil
| |
Collapse
|
16
|
Shah M, He Z, Rauf A, Beikoghli Kalkhoran S, Heiestad CM, Stensløkken KO, Parish CR, Soehnlein O, Arjun S, Davidson SM, Yellon D. Extracellular histones are a target in myocardial ischaemia-reperfusion injury. Cardiovasc Res 2022; 118:1115-1125. [PMID: 33878183 PMCID: PMC8930072 DOI: 10.1093/cvr/cvab139] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 10/16/2020] [Accepted: 04/17/2021] [Indexed: 12/14/2022] Open
Abstract
AIMS Acute myocardial infarction causes lethal cardiomyocyte injury during ischaemia and reperfusion (I/R). Histones have been described as important Danger Associated Molecular Proteins (DAMPs) in sepsis. The objective of this study was to establish whether extracellular histone release contributes to myocardial infarction. METHODS AND RESULTS Isolated, perfused rat hearts were subject to I/R. Nucleosomes and histone-H4 release was detected early during reperfusion. Sodium-β-O-Methyl cellobioside sulfate (mCBS), a newly developed histone-neutralizing compound, significantly reduced infarct size whilst also reducing the detectable levels of histones. Histones were directly toxic to primary adult rat cardiomyocytes in vitro. This was prevented by mCBS or HIPe, a recently described, histone-H4 neutralizing peptide, but not by an inhibitor of TLR4, a receptor previously reported to be involved in DAMP-mediated cytotoxicity. Furthermore, TLR4-reporter HEK293 cells revealed that cytotoxicity of histone H4 was independent of TLR4 and NF-κB. In an in vivo rat model of I/R, HIPe significantly reduced infarct size. CONCLUSION Histones released from the myocardium are cytotoxic to cardiomyocytes, via a TLR4-independent mechanism. The targeting of extracellular histones provides a novel opportunity to limit cardiomyocyte death during I/R injury of the myocardium.
Collapse
Affiliation(s)
- Mohammed Shah
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Zhenhe He
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Ali Rauf
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Siavash Beikoghli Kalkhoran
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Christina Mathisen Heiestad
- Section of Physiology, Department of Molecular Medicine, Institute for Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Kåre-Olav Stensløkken
- Section of Physiology, Department of Molecular Medicine, Institute for Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Christopher R Parish
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Oliver Soehnlein
- Institute for Cardiovascular Prevention (IPEK), LMU Munich Hospital, Pettenkoferstrasse 8a, D-80336 Munich, Germany
- Institute for Experimental Pathology (ExPat), Center for Molecular Biology of Inflammation, WWU Münster, Von-Esmarch-Strasse 56 48149 Münster, Germany
- Department of Physiology and Pharmacology (FyFa), Karolinska Institutet, Stockholm, Sweden
| | - Sapna Arjun
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Derek Yellon
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| |
Collapse
|
17
|
Wang L, Wang Z, Liu X, Zhang Y, Wang M, Liang X, Li G. Effects of extracellular histones on left ventricular diastolic function and potential mechanisms in mice with sepsis. Am J Transl Res 2022; 14:150-165. [PMID: 35173835 PMCID: PMC8829607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 10/14/2021] [Indexed: 06/14/2023]
Abstract
OBJECTIVE Extracellular histone (EH) is involved in the development of septic myocardial injury (SMI). In this study, we explored whether EH could induce left ventricular diastolic dysfunction (LVDD) in sepsis, and investigated the potential mechanisms through in vivo and in vitro experiments using animal models. METHODS The ratio between E-wave and A-wave (E/A ratio), left ventricular end diastolic volume, and isovolumic relaxation time (IVRT) were measured in cecal ligation and perforation (CLP)- and EH-treated male C57BL/6J mice using echocardiography. The protein and mRNA levels of apoptosis-related proteins (cleaved caspase-3, Bcl-2, and Bax) and cardiac troponin T (cTnT) in the left ventricular tissue/cardiomyocytes were measured using enzyme-linked immunosorbent assay, qRT-PCR, and western blotting. Cardiomyocyte apoptosis was detected by flow cytometry. RESULTS CLP mice presented with LVDD, which was accompanied by increased circulating histones, cTnT and Bax protein levels. Circulating histones were correlated with cTnT, Bax, IVRT, and E/A ratio in CLP mice. Intraperitoneal injection of EH resulted in LVDD in mice. EH induced cardiomyocyte apoptosis, and histone neutralizing agents improved SMI and protected mice against CLP- and EH-induced death. CONCLUSION EH is involved in septic LVDD, and this alteration might be associated with EH-induced apoptosis. EH may serve as a potential therapeutic target for SMI.
Collapse
Affiliation(s)
- Lijun Wang
- Department of Cardiology, Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical UniversityTianjin 300211, People’s Republic of China
- Department of Emergency Medicine, Tianjin Medical University General HospitalTianjin 300052, People’s Republic of China
| | - Ziyi Wang
- Department of Emergency Medicine, Tianjin Medical University General HospitalTianjin 300052, People’s Republic of China
- School of Clinical Medicine, Tsinghua UniversityBeijing 100084, People’s Republic of China
| | - Xing Liu
- Department of Cardiology, Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical UniversityTianjin 300211, People’s Republic of China
| | - Yue Zhang
- Department of Cardiology, Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical UniversityTianjin 300211, People’s Republic of China
| | - Manman Wang
- Department of Cardiology, Affiliated Hospital of Jining Medical UniversityJining 272000, Shandong, People’s Republic of China
| | - Xue Liang
- Department of Cardiology, Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical UniversityTianjin 300211, People’s Republic of China
| | - Guangping Li
- Department of Cardiology, Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical UniversityTianjin 300211, People’s Republic of China
| |
Collapse
|
18
|
Tian Y, Li P, Wu Z, Deng Q, Pan B, Stringer KA, Alam HB, Standiford TJ, Li Y. Citrullinated Histone H3 Mediates Sepsis-Induced Lung Injury Through Activating Caspase-1 Dependent Inflammasome Pathway. Front Immunol 2021; 12:761345. [PMID: 34950139 PMCID: PMC8688857 DOI: 10.3389/fimmu.2021.761345] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 11/17/2021] [Indexed: 12/25/2022] Open
Abstract
Sepsis is a life-threatening organ dysfunction caused by dysregulated host response to infection that often results in acute lung injury (ALI)/acute respiratory distress syndrome (ARDS). An emerging mechanism of sepsis-induced ARDS involves neutrophils/macrophages undergoing cell death, releasing nuclear histones to cause tissue damage that exacerbates pulmonary injury. While published studies focus on unmodified histones, little is known about the role of citrullinated histone H3 (CitH3) in the pathogenesis of sepsis and ALI. In this study, we found that levels of CitH3 were elevated in the patients with sepsis-induced ARDS and correlated to PaO2/FiO2 in septic patients. Systematic administration of CitH3 peptide in mice provoked Caspase-1 activation in the lung tissue and caused ALI. Neutralization of CitH3 with monoclonal antibody improved survival and attenuated ALI in a mouse sepsis model. Furthermore, we demonstrated that CitH3 induces ALI through activating Caspase-1 dependent inflammasome in bone marrow derived macrophages and bone marrow derived dendritic cells. Our study suggests that CitH3 is an important mediator of inflammation and mortality during sepsis-induced ALI.
Collapse
Affiliation(s)
- Yuzi Tian
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, China
- Department of Surgery, University of Michigan Health System, Ann Arbor, MI, United States
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Central South University, Changsha, China
| | - Patrick Li
- Department of Surgery, University of Michigan Health System, Ann Arbor, MI, United States
- Department of Internal Medicine, New York University (NYU) Langone Health, New York, NY, United States
| | - Zhenyu Wu
- Department of Surgery, University of Michigan Health System, Ann Arbor, MI, United States
- Department of Infectious Disease, Second Xiangya Hospital, Central South University, Changsha, China
| | - Qiufang Deng
- Department of Surgery, University of Michigan Health System, Ann Arbor, MI, United States
| | - Baihong Pan
- Department of Surgery, University of Michigan Health System, Ann Arbor, MI, United States
| | - Kathleen A. Stringer
- Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, MI, United States
- Division of Pulmonary and Critical Care Medicine, University of Michigan Medical Center, Ann Arbor, MI, United States
| | - Hasan B. Alam
- Department of Surgery, University of Michigan Health System, Ann Arbor, MI, United States
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Theodore J. Standiford
- Division of Pulmonary and Critical Care Medicine, University of Michigan Medical Center, Ann Arbor, MI, United States
| | - Yongqing Li
- Department of Surgery, University of Michigan Health System, Ann Arbor, MI, United States
| |
Collapse
|
19
|
Busch K, Kny M, Huang N, Klassert TE, Stock M, Hahn A, Graeger S, Todiras M, Schmidt S, Chamling B, Willenbrock M, Groß S, Biedenweg D, Heuser A, Scheidereit C, Butter C, Felix SB, Otto O, Luft FC, Slevogt H, Fielitz J. Inhibition of the NLRP3/IL-1β axis protects against sepsis-induced cardiomyopathy. J Cachexia Sarcopenia Muscle 2021; 12:1653-1668. [PMID: 34472725 PMCID: PMC8718055 DOI: 10.1002/jcsm.12763] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 06/03/2021] [Accepted: 07/09/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Septic cardiomyopathy worsens the prognosis of critically ill patients. Clinical data suggest that interleukin-1β (IL-1β), activated by the NLRP3 inflammasome, compromises cardiac function. Whether or not deleting Nlrp3 would prevent cardiac atrophy and improve diastolic cardiac function in sepsis was unclear. Here, we investigated the role of NLRP3/IL-1β in sepsis-induced cardiomyopathy and cardiac atrophy. METHODS Male Nlrp3 knockout (KO) and wild-type (WT) mice were exposed to polymicrobial sepsis by caecal ligation and puncture (CLP) surgery (KO, n = 27; WT, n = 33) to induce septic cardiomyopathy. Sham-treated mice served as controls (KO, n = 11; WT, n = 16). Heart weights and morphology, echocardiography and analyses of gene and protein expression were used to evaluate septic cardiomyopathy and cardiac atrophy. IL-1β effects on primary and immortalized cardiomyocytes were investigated by morphological and molecular analyses. IonOptix and real-time deformability cytometry (RT-DC) analysis were used to investigate functional and mechanical effects of IL-1β on cardiomyocytes. RESULTS Heart morphology and echocardiography revealed preserved systolic (stroke volume: WT sham vs. WT CLP: 33.1 ± 7.2 μL vs. 24.6 ± 8.7 μL, P < 0.05; KO sham vs. KO CLP: 28.3 ± 8.1 μL vs. 29.9 ± 9.9 μL, n.s.; P < 0.05 vs. WT CLP) and diastolic (peak E wave velocity: WT sham vs. WT CLP: 750 ± 132 vs. 522 ± 200 mm/s, P < 0.001; KO sham vs. KO CLP: 709 ± 152 vs. 639 ± 165 mm/s, n.s.; P < 0.05 vs. WT CLP) cardiac function and attenuated cardiac (heart weight-tibia length ratio: WT CLP vs. WT sham: -26.6%, P < 0.05; KO CLP vs. KO sham: -3.3%, n.s.; P < 0.05 vs. WT CLP) and cardiomyocyte atrophy in KO mice during sepsis. IonOptix measurements showed that IL-1β decreased contractility (cell shortening: IL-1β: -15.4 ± 2.3%, P < 0.001 vs. vehicle, IL-1RA: -6.1 ± 3.3%, P < 0.05 vs. IL-1β) and relaxation of adult rat ventricular cardiomyocytes (time-to-50% relengthening: IL-1β: 2071 ± 225 ms, P < 0.001 vs. vehicle, IL-1RA: 564 ± 247 ms, P < 0.001 vs. IL-1β), which was attenuated by an IL-1 receptor antagonist (IL-1RA). RT-DC analysis indicated that IL-1β reduced cardiomyocyte size (P < 0.001) and deformation (P < 0.05). RNA sequencing showed that genes involved in NF-κB signalling, autophagy and lysosomal protein degradation were enriched in hearts of septic WT but not in septic KO mice. Western blotting and qPCR disclosed that IL-1β activated NF-κB and its target genes, caused atrophy and decreased myosin protein in myocytes, which was accompanied by an increased autophagy gene expression. These effects were attenuated by IL-1RA. CONCLUSIONS IL-1β causes atrophy, impairs contractility and relaxation and decreases deformation of cardiomyocytes. Because NLRP3/IL-1β pathway inhibition attenuates cardiac atrophy and cardiomyopathy in sepsis, it could be useful to prevent septic cardiomyopathy.
Collapse
Affiliation(s)
- Katharina Busch
- Experimental and Clinical Research Center (ECRC), Charité-Universitätsmedizin Berlin, Max Delbrück Center (MDC) for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Melanie Kny
- Experimental and Clinical Research Center (ECRC), Charité-Universitätsmedizin Berlin, Max Delbrück Center (MDC) for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Nora Huang
- Experimental and Clinical Research Center (ECRC), Charité-Universitätsmedizin Berlin, Max Delbrück Center (MDC) for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Department of Cardiology, Heart Center Brandenburg and Medical School Brandenburg (MHB), Bernau, Germany
| | - Tilman E Klassert
- ZIK Septomics, Host Septomics, Jena, Germany.,Jena University Hospital, Integrated Research and Treatment Center - Center for Sepsis Control and Care (CSCC), Jena, Germany
| | - Magdalena Stock
- ZIK Septomics, Host Septomics, Jena, Germany.,Jena University Hospital, Integrated Research and Treatment Center - Center for Sepsis Control and Care (CSCC), Jena, Germany
| | - Alexander Hahn
- Experimental and Clinical Research Center (ECRC), Charité-Universitätsmedizin Berlin, Max Delbrück Center (MDC) for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Sebastian Graeger
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Mihail Todiras
- Laboratory of Molecular Biology of Peptide Hormones, Max Delbrück Center for Molecular Medicine, Berlin, Germany.,Nicolae Testemiţanu State University of Medicine and Pharmacy, Chișinău, Moldova
| | - Sibylle Schmidt
- Experimental and Clinical Research Center (ECRC), Charité-Universitätsmedizin Berlin, Max Delbrück Center (MDC) for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Bishwas Chamling
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany.,Department of Internal Medicine B, Molecular Cardiology, University Medicine Greifswald, Greifswald, Germany
| | - Michael Willenbrock
- Signal Transduction in Development and Cancer, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Stefan Groß
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany.,Department of Internal Medicine B, Molecular Cardiology, University Medicine Greifswald, Greifswald, Germany
| | - Doreen Biedenweg
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany.,Centre for Innovation Competence - Humoral Immune Response in Cardiovascular Diseases, University of Greifswald, Greifswald, Germany
| | - Arnd Heuser
- Animal Phenotyping Facility, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Claus Scheidereit
- Signal Transduction in Development and Cancer, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Christian Butter
- Department of Cardiology, Heart Center Brandenburg and Medical School Brandenburg (MHB), Bernau, Germany
| | - Stephan B Felix
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany.,Department of Internal Medicine B, Molecular Cardiology, University Medicine Greifswald, Greifswald, Germany
| | - Oliver Otto
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany.,Centre for Innovation Competence - Humoral Immune Response in Cardiovascular Diseases, University of Greifswald, Greifswald, Germany
| | - Friedrich C Luft
- Experimental and Clinical Research Center (ECRC), Charité-Universitätsmedizin Berlin, Max Delbrück Center (MDC) for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Hortense Slevogt
- ZIK Septomics, Host Septomics, Jena, Germany.,Jena University Hospital, Integrated Research and Treatment Center - Center for Sepsis Control and Care (CSCC), Jena, Germany
| | - Jens Fielitz
- Experimental and Clinical Research Center (ECRC), Charité-Universitätsmedizin Berlin, Max Delbrück Center (MDC) for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany.,Department of Internal Medicine B, Molecular Cardiology, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
20
|
Biswas I, Panicker SR, Cai XS, Giri H, Rezaie AR. Extracellular Histones Bind Vascular Glycosaminoglycans and Inhibit the Anti-Inflammatory Function of Antithrombin. Cell Physiol Biochem 2021; 55:605-617. [PMID: 34655467 DOI: 10.33594/000000438] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/07/2021] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND/AIMS Binding of histones to molecular pattern recognition receptors on endothelial cells and leukocytes provokes proinflammatory responses and promotes activation of coagulation. Histones also bind therapeutic heparins, thereby neutralizing their anticoagulant functions. The aim of this study was to test the hypothesis that histones can interact with the antithrombin (AT)-binding vascular glycosaminoglycans (GAGs) to induce inflammation and inhibit the anti-inflammatory function of AT. METHODS We evaluated the heparin-binding function of histones by an AT-dependent protease-inhibition assay. Furthermore, we treated endothelial cells with histones in the absence and presence of AT and monitored cellular phenotypes employing established signaling assays. RESULTS Histones neutralized AT-dependent anticoagulant function of heparin in both purified protease-inhibition and plasma-based assays. Histones also disrupted endothelial cell barrier-permeability function by a GAG-dependent mechanism as evidenced by the GAG-antagonist, surfen, abrogating their disruptive effects. Further studies revealed histones and AT compete for overlapping binding-sites on GAGs, thus increasing concentrations of one protein abrogated effects of the other. Histones elicited proapoptotic effects by inducing nuclear localization of PKC-δ in endothelial cells and barrier-disruptive effects by destabilizing VE-cadherin, which were inhibited by AT, but not by a D-helix mutant of AT incapable of interacting with GAGs. Finally, histones induced release of Weibel-Palade body contents, VWF and angiopoietin-2, and promoted expression of cell adhesion molecules on endothelial cells, which were all downregulated by AT but not by D-helix mutant of AT. CONCLUSION We conclude that histones and AT compete for overlapping binding sites on vascular GAGs to modulate coagulation and inflammation.
Collapse
Affiliation(s)
- Indranil Biswas
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Sumith R Panicker
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Xiaofeng S Cai
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Hemant Giri
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Alireza R Rezaie
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA, .,Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
21
|
The Early Diagnostic Efficacy of Serum Histone H3 in Rabbit Urosepsis Model. BIOMED RESEARCH INTERNATIONAL 2021; 2021:9969344. [PMID: 34327242 PMCID: PMC8310443 DOI: 10.1155/2021/9969344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 06/09/2021] [Accepted: 07/05/2021] [Indexed: 11/25/2022]
Abstract
Objective We want to explore the changing law of circulating histones in the acute stage of urosepsis and to find more sensitive and specific biomarkers for diagnosing urosepsis as early as possible. Methods Twenty healthy male New Zealand rabbits were randomly divided into 4 groups (N = 5): the control group, sham group, model group of LPS 600 μg/kg, and model group of LPS 1000 μg/kg. Heart rate (HR), respiration rate (RR), rectal temperature (T), and mean arterial pressure (MAP) were examined at 1, 3, 6, 12, and 24 hours after operation. Besides, peripheral blood cell counts (RBC, WBC, PLT, and Hb) and C reaction protein (CRP) were tested at 1, 3, and 6 hours after operation, while the levels of histone H3, MMP-9, TIMP-1, and procalcitonin (PCT) in the serum were tested at 1, 3, and 6 hours after operation by ELISA. The heart, left lung, liver, and left kidney were harvested for HE stain and observed to research the pathological change of these tissues. Results (1) The general status of rabbits: rabbits in the control and sham groups came out in 2 h after operation and regain to drink and eat in 12-24 h after operation. State of the rabbits in the control group was better than that in the sham group. Rabbits in the model groups were languid after operation and stopped to drink and eat. (2) Vital signs of rabbits: there was no statistic difference in HR (P = 0.238) and RR (P = 0.813) among all groups. MAP of the model groups decreased at 3 h postoperative, but transient (P < 0.001). The T of the LPS 1000 group decreased at 6 h postoperative (P = 0.003). (3) The change of biomarkers: H3 level of the LPS groups in the serum increased at 1 h postoperative (P < 0.01); MMP-9 of the LPS 1000 group increased at 1 h postoperative (P < 0.01); WBC of the model groups decreased at 3 h postoperative (P < 0.05); PLT of the LPS 1000 group is significantly increased at 1 h postoperative (P < 0.05); no statistic difference was found in CRP, PCT, and TIMP-1 among all groups. (4) Pathological sections: no abnormal performance was found in the control and sham groups. Glomerulus of the model groups was out of shape and necrosis with obvious renal tubule expansion. Pulmonary pathology showed alveolar septum diffuse increased and inflammatory infiltrate. Change of the LPS 1000 group was more serious than that of the LPS 600 group. Conclusions Ligating the ureter after an injection of 1000 μg/kg LPS into the ureter of the rabbit can establish the animal model of urosepsis. Histone H3 increase immediately at 1 h postoperative and are promised to be biomarkers of urosepsis, which are more effective than WBC, CRP, and PCT.
Collapse
|
22
|
Mannes M, Schmidt CQ, Nilsson B, Ekdahl KN, Huber-Lang M. Complement as driver of systemic inflammation and organ failure in trauma, burn, and sepsis. Semin Immunopathol 2021; 43:773-788. [PMID: 34191093 PMCID: PMC8243057 DOI: 10.1007/s00281-021-00872-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 05/23/2021] [Indexed: 02/08/2023]
Abstract
Complement is one of the most ancient defense systems. It gets strongly activated immediately after acute injuries like trauma, burn, or sepsis and helps to initiate regeneration. However, uncontrolled complement activation contributes to disease progression instead of supporting healing. Such effects are perceptible not only at the site of injury but also systemically, leading to systemic activation of other intravascular cascade systems eventually causing dysfunction of several vital organs. Understanding the complement pathomechanism and its interplay with other systems is a strict requirement for exploring novel therapeutic intervention routes. Ex vivo models exploring the cross-talk with other systems are rather limited, which complicates the determination of the exact pathophysiological roles that complement has in trauma, burn, and sepsis. Literature reporting on these three conditions is often controversial regarding the importance, distribution, and temporal occurrence of complement activation products further hampering the deduction of defined pathophysiological pathways driven by complement. Nevertheless, many in vitro experiments and animal models have shown beneficial effects of complement inhibition at different levels of the cascade. In the future, not only inhibition but also a complement reconstitution therapy should be considered in prospective studies to expedite how meaningful complement-targeted interventions need to be tailored to prevent complement augmented multi-organ failure after trauma, burn, and sepsis. This review summarizes clinically relevant studies investigating the role of complement in the acute diseases trauma, burn, and sepsis with important implications for clinical translation.
Collapse
Affiliation(s)
- Marco Mannes
- Institute of Clinical and Experimental Trauma-Immunology, University Hospital of Ulm, Helmholtzstr. 8/2, 89081, Ulm, Germany
| | - Christoph Q Schmidt
- Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm University, Ulm, Germany
| | - Bo Nilsson
- Department of Immunology, Genetics and Pathology (IGP), Rudbeck Laboratory C5:3, Uppsala University, Uppsala, Sweden
| | - Kristina N Ekdahl
- Department of Immunology, Genetics and Pathology (IGP), Rudbeck Laboratory C5:3, Uppsala University, Uppsala, Sweden.,Linnaeus Center of Biomaterials Chemistry, Linnaeus University, Kalmar, Sweden
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma-Immunology, University Hospital of Ulm, Helmholtzstr. 8/2, 89081, Ulm, Germany.
| |
Collapse
|
23
|
Early myocardial damage (EMD) and valvular dysfunction after femur fracture in pigs. Sci Rep 2021; 11:8503. [PMID: 33875675 PMCID: PMC8055677 DOI: 10.1038/s41598-021-86151-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 10/01/2020] [Indexed: 12/27/2022] Open
Abstract
Musculoskeletal injuries are the most common reason for surgery in severely injured patients. In addition to direct cardiac damage after physical trauma, there is rising evidence that trauma induces secondary cardiac structural and functional damage. Previous research associates hip fractures with the appearance of coronary heart disease: As 25% of elderly patients developed a major adverse cardiac event after hip fracture. 20 male pigs underwent femur fracture with operative stabilization via nailing (unreamed, reamed, RIA I and a new RIA II; each group n = 5). Blood samples were collected 6 h after trauma and the concentration of troponin I and heart-type fatty acid binding protein (HFABP) as biomarkers for EMD were measured. At baseline and 6 h after trauma, transesophageal ECHO (TOE) was performed; and invasive arterial and left ventricular blood pressure were measured to evaluate the cardiac function after femur fracture. A systemic elevation of troponin I and HFABP indicate an early myocardial damage after femur fracture in pigs. Furthermore, various changes in systolic (ejection fraction and cardiac output) and diastolic (left ventricular end-diastolic pressure, mitral valve deceleration time and E/A ratio) parameters illustrate the functional impairment of the heart. These findings were accompanied by the development of valvular dysfunction (pulmonary and tricuspid valve). To the best of our knowledge, we described for the first time the development of functional impairment of the heart in the context of EMD after long bone fracture in pigs. Next to troponin and HFABP elevation, alterations in the systolic and diastolic function occurred and were accompanied by pulmonary and tricuspid valvular insufficiency. Regarding EMD, none of the fracture stabilization techniques (unreamed nailing, reaming, RIA I and RIA II) was superior.
Collapse
|
24
|
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: 46] [Impact Index Per Article: 11.5] [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
|
25
|
Yehya N, Fazelinia H, Lawrence GG, Spruce LA, Mai MV, Worthen GS, Christie JD. Plasma Nucleosomes Are Associated With Mortality in Pediatric Acute Respiratory Distress Syndrome. Crit Care Med 2021; 49:1149-1158. [PMID: 33729723 PMCID: PMC8217097 DOI: 10.1097/ccm.0000000000004923] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVES Circulating nucleosomes and their component histones have been implicated as pathogenic in sepsis and acute respiratory distress syndrome in adults. However, their role in pediatric acute respiratory distress syndrome is unknown. DESIGN We performed a prospective cohort study in children with acute respiratory distress syndrome, with plasma collection within 24 hours of acute respiratory distress syndrome onset. We associated nucleosome levels with severity of acute respiratory distress syndrome and with nonpulmonary organ failures and tested for association of nucleosomes with PICU mortality and ventilator-free days at 28 days in univariate and multivariable analyses. We also performed proteomics of DNA-bound plasma proteins in a matched case-control study of septic children with and without acute respiratory distress syndrome in order to identify specific histone proteins elevated in acute respiratory distress syndrome. SETTING Large academic tertiary-care PICU. PATIENTS Intubated children meeting Berlin criteria for acute respiratory distress syndrome. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS We enrolled 333 children with acute respiratory distress syndrome, with 69 nonsurvivors (21%). Plasma nucleosomes were correlated with acute respiratory distress syndrome severity and with the number of nonpulmonary organ failures at acute respiratory distress syndrome onset. Nucleosomes were higher (p < 0.001) in nonsurvivors (0.40 [interquartile range, 0.20-0.71] arbitrary units) relative to survivors (0.10 [interquartile range, 0.04-0.25] arbitrary units). Nucleosomes were associated with PICU mortality in multivariable analysis (adjusted odds ratio 1.84 per 1 sd increase; 95% CI, 1.38-2.45; p < 0.001). Nucleosomes were also associated with a lower probability of being extubated alive by day 28 after multivariable adjustment (adjusted subdistribution hazard ratio, 0.74; 95% CI, 0.63-0.88; p = 0.001). Proteomic analysis demonstrated higher levels of the core nucleosome histones H2A, H2B, H3, and H4 in septic children with acute respiratory distress syndrome, relative to septic children without acute respiratory distress syndrome. CONCLUSIONS Plasma nucleosomes are associated with acute respiratory distress syndrome severity, nonpulmonary organ failures, and worse outcomes in pediatric acute respiratory distress syndrome.
Collapse
Affiliation(s)
- Nadir Yehya
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA
| | - Hossein Fazelinia
- Proteomics Core, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - Gladys G Lawrence
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA
| | - Lynn A Spruce
- Proteomics Core, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - Mark V Mai
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA
| | - G Scott Worthen
- Division of Neonatology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Jason D Christie
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine and Center for Translational Lung Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| |
Collapse
|
26
|
Drennan SE, Burge KY, Szyld EG, Eckert JV, Mir AM, Gormley AK, Schwartz RM, Daves SM, Thompson JL, Burkhart HM, Chaaban H. Clinical and Laboratory Predictors for the Development of Low Cardiac Output Syndrome in Infants Undergoing Cardiopulmonary Bypass: A Pilot Study. J Clin Med 2021; 10:jcm10040712. [PMID: 33670210 PMCID: PMC7916966 DOI: 10.3390/jcm10040712] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 02/02/2021] [Accepted: 02/08/2021] [Indexed: 01/31/2023] Open
Abstract
Cardiac surgery employing cardiopulmonary bypass exposes infants to a high risk of morbidity and mortality. The objective of this study was to assess the utility of clinical and laboratory variables to predict the development of low cardiac output syndrome, a frequent complication following cardiac surgery in infants. We performed a prospective observational study in the pediatric cardiovascular ICU in an academic children’s hospital. Thirty-one patients with congenital heart disease were included. Serum levels of nucleosomes and a panel of 20 cytokines were measured at six time points in the perioperative period. Cardiopulmonary bypass patients were characterized by increased levels of interleukin-10, -6, and -1α upon admission to the ICU compared to non-bypass cardiac patients. Patients developing low cardiac output syndrome endured longer aortic cross-clamp time and required greater inotropic support at 12 h postoperatively compared to bypass patients not developing the condition. Higher preoperative interleukin-10 levels and 24 h postoperative interleukin-8 levels were associated with low cardiac output syndrome. Receiver operating characteristic curve analysis demonstrated a moderate capability of aortic cross-clamp duration to predict low cardiac output syndrome but not IL-8. In conclusion, low cardiac output syndrome was best predicted in our patient population by the surgical metric of aortic cross-clamp duration.
Collapse
Affiliation(s)
- Sarah E. Drennan
- Section of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (S.E.D.); (K.Y.B.); (E.G.S.); (J.V.E.)
| | - Kathryn Y. Burge
- Section of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (S.E.D.); (K.Y.B.); (E.G.S.); (J.V.E.)
| | - Edgardo G. Szyld
- Section of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (S.E.D.); (K.Y.B.); (E.G.S.); (J.V.E.)
| | - Jeffrey V. Eckert
- Section of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (S.E.D.); (K.Y.B.); (E.G.S.); (J.V.E.)
| | - Arshid M. Mir
- Section of Cardiology, Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
| | - Andrew K. Gormley
- Section of Pediatric Critical Care, Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
| | - Randall M. Schwartz
- Department of Anesthesiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (R.M.S.); (S.M.D.)
| | - Suanne M. Daves
- Department of Anesthesiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (R.M.S.); (S.M.D.)
| | - Jess L. Thompson
- Department of Cardiovascular Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (J.L.T.); (H.M.B.)
| | - Harold M. Burkhart
- Department of Cardiovascular Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (J.L.T.); (H.M.B.)
| | - Hala Chaaban
- Section of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (S.E.D.); (K.Y.B.); (E.G.S.); (J.V.E.)
- Correspondence:
| |
Collapse
|
27
|
Chen DD, Wang HW, Cai XJ. Transcription factor Sp1 ameliorates sepsis-induced myocardial injury via ZFAS1/Notch signaling in H9C2 cells. Cytokine 2021; 140:155426. [PMID: 33517197 DOI: 10.1016/j.cyto.2021.155426] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 12/24/2020] [Accepted: 01/06/2021] [Indexed: 12/11/2022]
Abstract
PURPOSE To investigate whether Sp1 can ameliorate sepsis-induced myocardial injury and explore the potential molecular mechanism. METHODS The embryonic cardiomyocyte cell line H9C2 and primary cultured mouse neonatal cardiomyocytes (CMNCs) were treated with LPS or phosphate-buffered saline (PBS). A mouse model of LPS-induced sepsis was established using male C57BL/6J mice and their cardiomyocytes were collected. Real-time reverse transcription-PCR (qRT-PCR) assay was used to detect the expression levels of Sp1 and ZFAS1 in cardiomyocytes. Western blotting analysis was used to assess the protein expression levels of Sp1, apoptosis-associated proteins and Notch signaling pathway related proteins. Luciferase assay was used to detect the interaction between Sp1 and ZFAS1. Cell transfection was used to generate H9C2 cells with overexpressed or knocked down of Sp1 or ZFAS1. MTT assay and flow cytometry analysis were used to test the cell proliferation and cell apoptosis ratio. RESULTS Our data revealed that the expressions of ZFAS1 and Sp1 were significantly reduced in LPS-treated H9C2 cells and primary CMNCs. The downregulation of ZFAS1 and Sp1 were also found in cardiomyocytes obtained from LPS-challenged mice. LPS induced H9C2 cell apoptosis and depressed cell proliferation was ameliorated by ZFAS1 overexpression and aggravated by ZFAS1 knockdown. Mechanistically, Luciferase assay indicated that Sp1 could bind to ZFAS1, and positively regulated ZFAS1 expression. Moreover, Notch signaling pathway participates in H9C2 cell apoptosis mediated by Sp1. CONCLUSION The present study demonstrates that Sp1 regulates LPS-induced cardiomyocyte apoptosis via ZFAS1/Notch signaling pathway, which may serve as therapeutic targets for sepsis-induced myocardial injury.
Collapse
Affiliation(s)
- Dan-Dan Chen
- Department of Critical Care Medicine, Haikou Hospital, Xiangya Medical College, Central South University, Haikou 570208, PR China
| | - Hong-Wu Wang
- Department of Critical Care Medicine, Haikou Hospital, Xiangya Medical College, Central South University, Haikou 570208, PR China
| | - Xing-Jun Cai
- Department of Respiratory and Critical Care Medicine, Hainan General Hospital, Haikou 570311, PR China.
| |
Collapse
|
28
|
Weber B, Lackner I, Gebhard F, Miclau T, Kalbitz M. Trauma, a Matter of the Heart-Molecular Mechanism of Post-Traumatic Cardiac Dysfunction. Int J Mol Sci 2021; 22:E737. [PMID: 33450984 PMCID: PMC7828409 DOI: 10.3390/ijms22020737] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 01/07/2021] [Accepted: 01/09/2021] [Indexed: 12/18/2022] Open
Abstract
Trauma remains a leading global cause of mortality, particularly in the young population. In the United States, approximately 30,000 patients with blunt cardiac trauma were recorded annually. Cardiac damage is a predictor for poor outcome after multiple trauma, with a poor prognosis and prolonged in-hospitalization. Systemic elevation of cardiac troponins was correlated with survival, injury severity score, and catecholamine consumption of patients after multiple trauma. The clinical features of the so-called "commotio cordis" are dysrhythmias, including ventricular fibrillation and sudden cardiac arrest as well as wall motion disorders. In trauma patients with inappropriate hypotension and inadequate response to fluid resuscitation, cardiac injury should be considered. Therefore, a combination of echocardiography (ECG) measurements, echocardiography, and systemic appearance of cardiomyocyte damage markers such as troponin appears to be an appropriate diagnostic approach to detect cardiac dysfunction after trauma. However, the mechanisms of post-traumatic cardiac dysfunction are still actively being investigated. This review aims to discuss cardiac damage following trauma, focusing on mechanisms of post-traumatic cardiac dysfunction associated with inflammation and complement activation. Herein, a causal relationship of cardiac dysfunction to traumatic brain injury, blunt chest trauma, multiple trauma, burn injury, psychosocial stress, fracture, and hemorrhagic shock are illustrated and therapeutic options are discussed.
Collapse
Affiliation(s)
- Birte Weber
- Department of Traumatology, Hand-, Plastic-, and Reconstructive Surgery, Center of Surgery, University of Ulm, 86081 Ulm, Germany; (B.W.); (I.L.); (F.G.)
| | - Ina Lackner
- Department of Traumatology, Hand-, Plastic-, and Reconstructive Surgery, Center of Surgery, University of Ulm, 86081 Ulm, Germany; (B.W.); (I.L.); (F.G.)
| | - Florian Gebhard
- Department of Traumatology, Hand-, Plastic-, and Reconstructive Surgery, Center of Surgery, University of Ulm, 86081 Ulm, Germany; (B.W.); (I.L.); (F.G.)
| | - Theodore Miclau
- Orthopaedic Trauma Institute, Department of Orthopaedic Surgery, University of California, 2550 23rd Street, San Francisco, CA 94110, USA;
| | - Miriam Kalbitz
- Department of Traumatology, Hand-, Plastic-, and Reconstructive Surgery, Center of Surgery, University of Ulm, 86081 Ulm, Germany; (B.W.); (I.L.); (F.G.)
| |
Collapse
|
29
|
Weber B, Lackner I, Braun CK, Kalbitz M, Huber-Lang M, Pressmar J. Laboratory Markers in the Management of Pediatric Polytrauma: Current Role and Areas of Future Research. Front Pediatr 2021; 9:622753. [PMID: 33816396 PMCID: PMC8010656 DOI: 10.3389/fped.2021.622753] [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: 10/29/2020] [Accepted: 02/19/2021] [Indexed: 11/13/2022] Open
Abstract
Severe trauma is the most common cause of mortality in children and is associated with a high socioeconomic burden. The most frequently injured organs in children are the head and thorax, followed by the extremities and by abdominal injuries. The efficient and early assessment and management of these injuries is essential to improve patients' outcome. Physical examination as well as imaging techniques like ultrasound, X-ray and computer tomography are crucial for a valid early diagnosis. Furthermore, laboratory analyses constitute additional helpful tools for the detection and monitoring of pediatric injuries. Specific inflammatory markers correlate with post-traumatic complications, including the development of multiple organ failure. Other laboratory parameters, including lactate concentration, coagulation parameters and markers of organ injury, represent further clinical tools to identify trauma-induced disorders. In this review, we outline and evaluate specific biomarkers for inflammation, acid-base balance, blood coagulation and organ damage following pediatric polytrauma. The early use of relevant laboratory markers may assist decision making on imaging tools, thus contributing to minimize radiation-induced long-term consequences, while improving the outcome of children with multiple trauma.
Collapse
Affiliation(s)
- Birte Weber
- Department of Traumatology, Hand-, Plastic- and Reconstructive Surgery, Center of Surgery, University of Ulm, Ulm, Germany
| | - Ina Lackner
- Department of Traumatology, Hand-, Plastic- and Reconstructive Surgery, Center of Surgery, University of Ulm, Ulm, Germany
| | - Christian Karl Braun
- Institute of Clinical and Experimental Trauma-Immunology, University Hospital of Ulm, Ulm, Germany.,Department of Pediatrics, University Medical Center Ulm, Ulm, Germany
| | - Miriam Kalbitz
- Department of Traumatology, Hand-, Plastic- and Reconstructive Surgery, Center of Surgery, University of Ulm, Ulm, Germany
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma-Immunology, University Hospital of Ulm, Ulm, Germany
| | - Jochen Pressmar
- Department of Traumatology, Hand-, Plastic- and Reconstructive Surgery, Center of Surgery, University of Ulm, Ulm, Germany
| |
Collapse
|
30
|
Zetoune FS, Ward PA. Role of Complement and Histones in Sepsis. Front Med (Lausanne) 2020; 7:616957. [PMID: 33425963 PMCID: PMC7785970 DOI: 10.3389/fmed.2020.616957] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 11/30/2020] [Indexed: 12/17/2022] Open
Abstract
The wide use of the mouse model of polymicrobial sepsis has provided important evidence for events occurring in infectious sepsis involving septic mice and septic humans. Nearly 100 clinical trials in humans with sepsis have been completed, yet there is no FDA-approved drug. Our studies of polymicrobial sepsis have highlighted the role of complement activation products (especially C5a anaphylatoxin and its receptors C5aR1 and C5aR2) in adverse effects of sepsis. During sepsis, the appearance of these complement products is followed by appearance of extracellular histones in plasma, which have powerful proinflammatory and prothrombotic activities that cause cell injury and multiorgan dysfunction in septic mice. Similar responses occur in septic humans. Histone appearance in plasma is related to complement activation and appearance of C5a and its interaction with its receptors. Development of the cardiomyopathy of sepsis also depends on C5a, C5a receptors and histones. Neutralization of C5a with antibody or absence of C5aR1 blocks appearance of extracellular histones and cell and organ failure in sepsis. Survival rates in septic mice are greatly improved after blockade of C5a with antibody. We also review the various strategies in sepsis that greatly reduce the development of life-threatening events of sepsis.
Collapse
Affiliation(s)
- Firas S Zetoune
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Peter A Ward
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
31
|
Lackner I, Weber B, Miclau T, Holzwarth N, Baur M, Gebhard F, Teuben M, Halvachizadeh S, Cinelli P, Pfeifer R, Lipiski M, Cesarovic N, Haffner-Luntzer M, Pape HC, Kalbitz M. Reaming of femoral fractures with different reaming irrigator aspirator systems shows distinct effects on cardiac function after experimental polytrauma. J Orthop Res 2020; 38:2608-2618. [PMID: 32827323 DOI: 10.1002/jor.24830] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 08/11/2020] [Accepted: 08/18/2020] [Indexed: 02/04/2023]
Abstract
Cardiac injuries are recorded after multiple trauma and are associated with a poor patient outcome. Reaming prior to locked intramedullary nailing is a frequently used technique to stabilize femoral diaphysis fractures. However, in polytraumatized patients, complications such as fat emboli and acute respiratory distress syndrome have been associated with reaming. The reaming irrigator aspirator (RIA) system provides concomitant irrigation and suction of the intramedullary contents, and should, therefore, reduce reaming-associated complications. The aim of the study was to investigate cardiac function after multiple trauma with regard to two different RIA devices (RIAI vs RIAII). 15 male pigs were included in the study. Pigs received either sham treatment or multiple trauma (chest trauma, femur fracture, liver laceration, and hemorrhagic shock), followed by intramedullary nailing after reaming with either the RIAI or RIAII system (RIAII: reduced diameter of the reamer, improved control of irrigation and suction). Cardiac function was assessed by transesophageal echocardiography and systemic inflammation as well as local cardiac damage examined. Pigs of both treatment groups showed impaired cardiac function, valvular insufficiency, and cardiac damage. Systemic inflammation and local cardiac alterations were observed which might contribute to early myocardial damage in vivo. Multiple trauma including long-bone fracture and subsequent intramedullary reaming induces cardiac dysfunction and valvular insufficiency, which might be linked to both mechanical cardiac injury and increased systemic inflammation. 6 hours after trauma there are less differences between RIAI and RIAII treatment with regard to post-traumatic cardiac consequences in multiple injured pigs, indicating no beneficial effect of RIAII over RIAI.
Collapse
Affiliation(s)
- Ina Lackner
- Department of Traumatology, Hand-, Plastic- and Reconstructive Surgery, University Hospital Ulm, Ulm, Germany
| | - Birte Weber
- Department of Traumatology, Hand-, Plastic- and Reconstructive Surgery, University Hospital Ulm, Ulm, Germany
| | - Theodore Miclau
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco, California
| | - Nina Holzwarth
- Department of Traumatology, Hand-, Plastic- and Reconstructive Surgery, University Hospital Ulm, Ulm, Germany
| | - Meike Baur
- Department of Traumatology, Hand-, Plastic- and Reconstructive Surgery, University Hospital Ulm, Ulm, Germany
| | - Florian Gebhard
- Department of Traumatology, Hand-, Plastic- and Reconstructive Surgery, University Hospital Ulm, Ulm, Germany
| | - Michel Teuben
- Department of Trauma, University Hospital of Zurich, Zurich, Switzerland
| | | | - Paolo Cinelli
- Department of Trauma, University Hospital of Zurich, Zurich, Switzerland
| | - Roman Pfeifer
- Department of Trauma, University Hospital of Zurich, Zurich, Switzerland
| | - Miriam Lipiski
- Department of Surgical Research, University Hospital of Zurich, Zurich, Switzerland
| | - Nikola Cesarovic
- Department of Surgical Research, University Hospital of Zurich, Zurich, Switzerland
| | | | | | - Miriam Kalbitz
- Department of Traumatology, Hand-, Plastic- and Reconstructive Surgery, University Hospital Ulm, Ulm, Germany
| | | |
Collapse
|
32
|
Villalba N, Baby S, Cha BJ, Yuan SY. Site-specific opening of the blood-brain barrier by extracellular histones. J Neuroinflammation 2020; 17:281. [PMID: 32962721 PMCID: PMC7510151 DOI: 10.1186/s12974-020-01950-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 09/08/2020] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Increased extracellular histones in the bloodstream are known as a biomarker for vascular dysfunction associated with severe trauma or sepsis. There is limited information regarding the pathogenic role of circulating histones in neuroinflammation and cerebrovascular endothelial injury. Particularly, it remains unclear whether histones affect the blood-brain barrier (BBB) permeability function. METHODS The direct effects of unfractionated histones on endothelial barrier properties were first assessed in brain microvascular endothelial cell monolayers by measuring transendothelial electrical resistance and solute flux. This was followed by in vivo mouse experiments, where BBB function was assessed by quantifying brain tissue accumulation of intravenously injected tracers of different molecular sizes, and comparison was made in mice receiving a sublethal dose of histones versus sterile saline. In parallel, the endothelial barrier ultrastructure was examined in histone- and saline-injected animals under transmission electron microscopy, corresponding to the expression of tight junction and adherens junction proteins. RESULTS Histones increased paracellular permeability to sodium fluorescein and reduced barrier resistance at 100 μg/mL; these responses were accompanied by discontinuous staining of the tight junction proteins claudin-5 and zona ocludens-1. Interestingly, the effects of histones did not seem to result from cytotoxicity, as evidenced by negative propidium iodide staining. In vivo, histones increased the paracellular permeability of the BBB to small tracers of < 1-kDa, whereas tracers larger than 3-kDa remained impermeable across brain microvessels. Further analysis of different brain regions showed that histone-induced tracer leakage and loss of tight junction protein expression mainly occurred in the hippocampus, but not in the cerebral cortex. Consistently, opening of tight junctions was found in hippocampal capillaries from histone-injected animals. Protein expression levels of GFAP and iBA1 remained unchanged in histone-injected mice indicating that histones did not affect reactive gliosis. Moreover, cell membrane surface charge alterations are involved in histone-induced barrier dysfunction and tight junction disruption. CONCLUSIONS Extracellular histones cause a reversible, region-specific increase in BBB permeability to small molecules by disrupting tight junctions in the hippocampus. We suggest that circulating histones may contribute to cerebrovascular injury or brain dysfunction by altering BBB structure and function.
Collapse
Affiliation(s)
- Nuria Villalba
- Department of Molecular Pharmacology & Physiology, University of South Florida, Morsani College of Medicine, Tampa, FL, USA
| | - Sheon Baby
- Department of Molecular Pharmacology & Physiology, University of South Florida, Morsani College of Medicine, Tampa, FL, USA
| | - Byeong J Cha
- Department of Molecular Pharmacology & Physiology, University of South Florida, Morsani College of Medicine, Tampa, FL, USA
| | - Sarah Y Yuan
- Department of Molecular Pharmacology & Physiology, University of South Florida, Morsani College of Medicine, Tampa, FL, USA.
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, USA.
| |
Collapse
|
33
|
Fattahi F, Grailer JJ, Parlett M, Lu H, Malan EA, Abe E, Russell MW, Frydrych LM, Delano MJ, Zetoune FS, Ward PA. Requirement of Complement C6 for Intact Innate Immune Responses in Mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 205:251-260. [PMID: 32444389 DOI: 10.4049/jimmunol.1900801] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 04/01/2020] [Indexed: 12/11/2022]
Abstract
Over the first days of polymicrobial sepsis, there is robust activation of the innate immune system, causing the appearance of proinflammatory cytokines and chemokines, along with the appearance of extracellular histones, which are highly proinflammatory and prothrombotic. In the current study, we studied different innate immune responses in mice with knockout (KO) of complement protein 6 (C6). Polymorphonuclear neutrophils (PMNs) from these KO mice had defective innate immune responses, including defective expression of surface adhesion molecules, generation of superoxide anion, and appearance of reactive oxygen species and histone release after activation of PMNs, along with defective phagocytosis. In addition, in C6-/- mice, the NLRP3 inflammasome was defective both in PMNs and in macrophages. When these KO mice were subjected to polymicrobial sepsis, their survival was improved, associated with reduced levels in the plasma of proinflammatory cytokines and chemokines and lower levels of histones in plasma. In addition, sepsis-induced cardiac dysfunction was attenuated in these KO mice. In a model of acute lung injury induced by LPS, C6-/- mice showed reduced PMN buildup and less lung epithelial/endothelial cell dysfunction (edema and hemorrhage). These data indicate that C6-/- mice have reduced innate immune responses that result in less organ injury and improved survival after polymicrobial sepsis.
Collapse
Affiliation(s)
- Fatemeh Fattahi
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Jamison J Grailer
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Michella Parlett
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Hope Lu
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Elizabeth A Malan
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Elizabeth Abe
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Mark W Russell
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109; and
| | - Lynn M Frydrych
- Department of Surgery, University of Michigan School Medical School, Ann Arbor, MI 48109
| | - Matthew J Delano
- Department of Surgery, University of Michigan School Medical School, Ann Arbor, MI 48109
| | - Firas S Zetoune
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Peter A Ward
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109;
| |
Collapse
|
34
|
Structural alterations and inflammation in the heart after multiple trauma followed by reamed versus non-reamed femoral nailing. PLoS One 2020; 15:e0235220. [PMID: 32584885 PMCID: PMC7316303 DOI: 10.1371/journal.pone.0235220] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 06/10/2020] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Approximately 30,000 patients with blunt cardiac trauma are recorded each year in the United States. Blunt cardiac injuries after trauma are associated with a longer hospital stay and a poor overall outcome. Organ damage after trauma is linked to increased systemic release of pro-inflammatory cytokines and damage-associated molecular patterns. However, the interplay between polytrauma and local cardiac injury is unclear. Additionally, the impact of surgical intervention on this process is currently unknown. This study aimed to determine local cardiac immunological and structural alterations after multiple trauma. Furthermore, the impact of the chosen fracture stabilization strategy (reamed versus non-reamed femoral nailing) on cardiac alterations was studied. EXPERIMENTAL APPROACH 15 male pigs were either exposed to multiple trauma (blunt chest trauma, laparotomy, liver laceration, femur fracture and haemorrhagic shock) or sham conditions. Blood samples as well as cardiac tissue were analysed 4 h and 6 h after trauma. Additionally, murine HL-1 cells were exposed to a defined polytrauma-cocktail, mimicking the pro-inflammatory conditions after multiple trauma in vitro. RESULTS After multiple trauma, cardiac structural changes were observed in the left ventricle. More specifically, alterations in the alpha-actinin and desmin protein expression were found. Cardiac structural alterations were accompanied by enhanced local nitrosative stress, increased local inflammation and elevated systemic levels of the high-mobility group box 1 protein. Furthermore, cardiac alterations were observed predominantly in pigs that were treated by non-reamed intramedullary reaming. The polytrauma-cocktail impaired the viability of HL-1 cells in vitro, which was accompanied by a release of troponin I and HFABP. DISCUSSION Multiple trauma induced cardiac structural alterations in vivo, which might contribute to the development of early myocardial damage (EMD). This study also revealed that reamed femoral nailing (reamed) is associated with more prominent immunological cardiac alterations compared to nailing without reaming (non-reamed). This suggests that the choice of the initial fracture treatment strategy might be crucial for the overall outcome as well as for any post-traumatic cardiac consequences.
Collapse
|
35
|
Toll-Like Receptor-Mediated Cardiac Injury during Experimental Sepsis. Mediators Inflamm 2020; 2020:6051983. [PMID: 32410859 PMCID: PMC7199613 DOI: 10.1155/2020/6051983] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 12/20/2019] [Indexed: 12/19/2022] Open
Abstract
Sepsis is associated with global cardiac dysfunction and with high mortality rate. The development of septic cardiomyopathy is due to complex interactions of damage-associated molecular patters, cytokines, and complement activation products. The aim of this study was to define the effects of sepsis on cardiac structure, gap junction, and tight junction (TJ) proteins. Sepsis was induced by cecal ligation and puncture in male C57BL/6 mice. After a period of 24 h, the expression of cardiac structure, gap junction, and TJ proteins was determined. Murine HL-1 cells were stimulated with LPS, and mRNA expression of cardiac structure and gap junction proteins, intracellular reactive oxygen species, and troponin I release was analyzed. Furthermore, pyrogenic receptor subtype 7 (P2X7) expression and troponin I release of human cardiomyocytes (iPS) were determined after LPS exposure. In vivo, protein expression of connexin43 and α-actinin was decreased after the onset of polymicrobial sepsis, whereas in HL-1 cells, mRNA expression of connexin43, α-actinin, and desmin was increased in the presence of LPS. Expression of TJ proteins was not affected in vivo during sepsis. Although the presence of LPS and nigericin resulted in a significant troponin I release from HL-1 cells. Sepsis affected cardiac structure and gap junction proteins in mice, potentially contributing to compromised cardiac function.
Collapse
|
36
|
Effects of Circulating HMGB-1 and Histones on Cardiomyocytes-Hemadsorption of These DAMPs as Therapeutic Strategy after Multiple Trauma. J Clin Med 2020; 9:jcm9051421. [PMID: 32403440 PMCID: PMC7291040 DOI: 10.3390/jcm9051421] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 04/29/2020] [Accepted: 05/05/2020] [Indexed: 02/06/2023] Open
Abstract
Background and purpose: The aim of the study was to determine the effects of post-traumatically released High Mobility Group Box-1 protein (HMGB1) and extracellular histones on cardiomyocytes (CM). We also evaluated a therapeutic option to capture circulating histones after trauma, using a hemadsorption filter to treat CM dysfunction. Experimental Approach: We evaluated cell viability, calcium handling and mitochondrial respiration of human cardiomyocytes in the presence of HMGB-1 and extracellular histones. In a translational approach, a hemadsorption filter was applied to either directly eliminate extracellular histones or to remove them from blood samples obtained from multiple injured patients. Key results: Incubation of human CM with HMGB-1 or histones is associated with changes in calcium handling, a reduction of cell viability and a substantial reduction of the mitochondrial respiratory capacity. Filtrating plasma from injured patients with a hemadsorption filter reduces histone concentration ex vivo and in vitro, depending on dosage. Conclusion and implications: Danger associated molecular patterns such as HMGB-1 and extracellular histones impair human CM in vitro. A hemadsorption filter could be a therapeutic option to reduce high concentrations of histones.
Collapse
|
37
|
Abstract
There is abundant evidence that infectious sepsis both in humans and mice with polymicrobial sepsis results in robust activation of complement. Major complement activation products involved in sepsis include C5a anaphylatoxin and its receptors (C5aR1 and C5aR2) and, perhaps, the terminal complement activation product, C5b-9. These products (and others) also cause dysfunction of the innate immune system, with exaggerated early proinflammatory responses, followed by decline of the innate immune system, leading to immunosuppression and multiorgan dysfunction. Generation of C5a during sepsis also leads to activation of neutrophils and macrophages and ultimate appearance of extracellular histones, which have powerful proinflammatory and prothrombotic activities. The distal complement activation product, C5b-9, triggers intracellular Ca fluxes in epithelial and endothelial cells. Histones activate the NLRP3 inflammasome, products of which can damage cells. C5a also activates MAPKs and Akt signaling pathways in cardiomyocytes, causing buildup of [Ca]i, defective action potentials and substantial cell dysfunction, resulting in cardiac and other organ dysfunction. Cardiac dysfunction can be quantitated by ECHO-Doppler parameters. In vivo interventions that block these complement-dependent products responsible for organ dysfunction in sepsis reduce the intensity of sepsis. The obvious targets in sepsis are C5a and its receptors, histones, and perhaps the MAPK pathways. Blockade of C5 has been considered in sepsis, but the FDA-approved antibody (eculizumab) is known to compromise defenses against neisseria and pneumonococcal bacteria, and requires immunization before the mAb to C5 can be used clinically. Small molecular blocking agents for C5aRs are currently in development and may be therapeutically effective for treatment of sepsis.
Collapse
|
38
|
Frydman GH, Tessier SN, Wong KHK, Vanderburg CR, Fox JG, Toner M, Tompkins RG, Irimia D. Megakaryocytes contain extranuclear histones and may be a source of platelet-associated histones during sepsis. Sci Rep 2020; 10:4621. [PMID: 32165642 PMCID: PMC7067782 DOI: 10.1038/s41598-020-61309-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 02/24/2020] [Indexed: 12/31/2022] Open
Abstract
Histones are typically located within the intracellular compartment, and more specifically, within the nucleus. When histones are located within the extracellular compartment, they change roles and become damage-associated molecular patterns (DAMPs), promoting inflammation and coagulation. Patients with sepsis have increased levels of extracellular histones, which have been shown to correlate with poor prognosis and the development of sepsis-related sequelae, such as end-organ damage. Until now, neutrophils were assumed to be the primary source of circulating histones during sepsis. In this paper, we show that megakaryocytes contain extranuclear histones and transfer histones to their platelet progeny. Upon examination of isolated platelets from patients with sepsis, we identified that patients with sepsis have increased amounts of platelet-associated histones (PAHs), which appear to be correlated with the type of infection. Taken together, these results suggest that megakaryocytes and platelets may be a source of circulating histones during sepsis and should be further explored.
Collapse
Affiliation(s)
- Galit H Frydman
- Division of Comparative Medicine and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America. .,BioMEMS Resource Center, Center for Engineering in Medicine, and Center for Surgery, Innovation and Bioengineering, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, United States of America.
| | - Shannon N Tessier
- BioMEMS Resource Center, Center for Engineering in Medicine, and Center for Surgery, Innovation and Bioengineering, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Keith H K Wong
- BioMEMS Resource Center, Center for Engineering in Medicine, and Center for Surgery, Innovation and Bioengineering, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Charles R Vanderburg
- Harvard Neurodiscovery Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - James G Fox
- Division of Comparative Medicine and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Mehmet Toner
- BioMEMS Resource Center, Center for Engineering in Medicine, and Center for Surgery, Innovation and Bioengineering, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Ronald G Tompkins
- BioMEMS Resource Center, Center for Engineering in Medicine, and Center for Surgery, Innovation and Bioengineering, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Daniel Irimia
- BioMEMS Resource Center, Center for Engineering in Medicine, and Center for Surgery, Innovation and Bioengineering, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, United States of America.
| |
Collapse
|
39
|
Lin H, Wang W, Lee M, Meng Q, Ren H. Current Status of Septic Cardiomyopathy: Basic Science and Clinical Progress. Front Pharmacol 2020; 11:210. [PMID: 32194424 PMCID: PMC7062914 DOI: 10.3389/fphar.2020.00210] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 02/14/2020] [Indexed: 12/27/2022] Open
Abstract
Septic cardiomyopathy (SCM) is a complication that is sepsis-associated cardiovascular failure. In the last few decades, there is progress in diagnosis and treatment despite the lack of consistent diagnostic criteria. According to current studies, several hypotheses about pathogenic mechanisms have been revealed to elucidate the pathophysiological characteristics of SCM. The objective of this manuscript is to review literature from the past 5 years to provide an overview of current knowledge on pathogenesis, diagnosis and treatment in SCM.
Collapse
Affiliation(s)
- Huan Lin
- Department of Intensive Care Unit, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Wenting Wang
- Department of Intensive Care Unit, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | | | - Qinghe Meng
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Hongsheng Ren
- Department of Intensive Care Unit, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| |
Collapse
|
40
|
Abstract
Despite an increase in the rates of survival in patients suffering myocardial infarction, as yet there is no therapy specifically targeting ischaemia and reperfusion injury of the myocardium. With a greater understanding of immune activation during infarction, more potential treatment targets are now being identified. The innate immune system is believed to play an important role in the myocardium after ischaemia-driven cardiomyocyte death. The release of intracellular contents including DNA into the extracellular space during necrosis and cell rupture is now believed to create a pro-inflammatory milieu which propagates the inflammatory process. DNA and DNA fragments have been shown to activate the innate immune system by acting as Danger-Associated Molecular Patterns (DAMPs), which act as ligands on toll-like receptors (TLRs). Stimulation of TLRs, in turn, can activate intracellular cell death pathways such as pyroptosis. Here, we review the role of DNA fragments during ischaemia and reperfusion, and assess their potential as a target in the quest to preserve cardiomyocyte viability following myocardial infarction.
Collapse
Affiliation(s)
- Mohammed Shah
- The Hatter Cardiovascular Institute, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, 67 Chenies Mews, London, WC1E 6HX, UK.
| |
Collapse
|
41
|
Lackner I, Weber B, Baur M, Fois G, Gebhard F, Pfeifer R, Cinelli P, Halvachizadeh S, Lipiski M, Cesarovic N, Schrezenmeier H, Huber-Lang M, Pape HC, Kalbitz M. Complement Activation and Organ Damage After Trauma-Differential Immune Response Based on Surgical Treatment Strategy. Front Immunol 2020; 11:64. [PMID: 32117238 PMCID: PMC7025487 DOI: 10.3389/fimmu.2020.00064] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 01/10/2020] [Indexed: 12/27/2022] Open
Abstract
Background: The complement system is part of the innate immunity, is activated immediately after trauma and is associated with adult respiratory distress syndrome, acute lung injury, multiple organ failure, and with death of multiply injured patients. The aim of the study was to investigate the complement activation in multiply injured pigs as well as its effects on the heart in vivo and in vitro. Moreover, the impact of reamed vs. non-reamed intramedullary nailing was examined with regard to the complement activation after multiple trauma in pigs. Materials and Methods: Male pigs received multiple trauma, followed by femoral nailing with/without prior conventional reaming. Systemic complement hemolytic activity (CH-50 and AH-50) as well as the local cardiac expression of C3a receptor, C5a receptors1/2, and the deposition of the fragments C3b/iC3b/C3c was determined in vivo after trauma. Human cardiomyocytes were exposed to C3a or C5a and analyzed regarding calcium signaling and mitochondrial respiration. Results: Systemic complement activation increased within 6 h after trauma and was mediated via the classical and the alternative pathway. Furthermore, complement activation correlated with invasiveness of fracture treatment. The expression of receptors for complement activation were altered locally in vivo in left ventricles. C3a and C5a acted detrimentally on human cardiomyocytes by affecting their functionality and their mitochondrial respiration in vitro. Conclusion: After multiple trauma, an early activation of the complement system is triggered, affecting the heart in vivo as well as in vitro, leading to complement-induced cardiac dysfunction. The intensity of complement activation after multiple trauma might correlate with the invasiveness of fracture treatment. Reaming of the femoral canal might contribute to an enhanced “second hit” response after trauma. Consequently, the choice of fracture treatment might imply the clinical outcome of the critically injured patients and might be therefore crucial for their survival.
Collapse
Affiliation(s)
- Ina Lackner
- Department of Traumatology, Hand-, Plastic- and Reconstructive Surgery, University of Ulm, Ulm, Germany
| | - Birte Weber
- Department of Traumatology, Hand-, Plastic- and Reconstructive Surgery, University of Ulm, Ulm, Germany
| | - Meike Baur
- Department of Traumatology, Hand-, Plastic- and Reconstructive Surgery, University of Ulm, Ulm, Germany
| | - Giorgio Fois
- Institute of General Physiology, University of Ulm, Ulm, Germany
| | - Florian Gebhard
- Department of Traumatology, Hand-, Plastic- and Reconstructive Surgery, University of Ulm, Ulm, Germany
| | - Roman Pfeifer
- Department of Trauma, University Hospital of Zurich, Zurich, Switzerland
| | - Paolo Cinelli
- Department of Trauma, University Hospital of Zurich, Zurich, Switzerland
| | | | - Miriam Lipiski
- Department of Surgical Research, University Hospital of Zurich, Zurich, Switzerland
| | - Nikola Cesarovic
- Department of Surgical Research, University Hospital of Zurich, Zurich, Switzerland
| | - Hubert Schrezenmeier
- Institute of Transfusion Medicine, University of Ulm and Institute of Clinical Transfusion Medicine and Immunogenetics Ulm, Ulm, Germany.,German Red Cross Blood Transfusion Service Baden-Württemberg - Hessen and University Hospital Ulm, Ulm, Germany
| | - Markus Huber-Lang
- Institute for Clinical- and Experimental Trauma-Immunology, University of Ulm, Ulm, Germany
| | | | - Miriam Kalbitz
- Department of Traumatology, Hand-, Plastic- and Reconstructive Surgery, University of Ulm, Ulm, Germany
| |
Collapse
|
42
|
Unfractionated heparin attenuates histone-mediated cytotoxicity in vitro and prevents intestinal microcirculatory dysfunction in histone-infused rats. J Trauma Acute Care Surg 2020; 87:614-622. [PMID: 31454337 DOI: 10.1097/ta.0000000000002387] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND Extracellular histones are major mediators of organ dysfunction and death in sepsis, and they may cause microcirculatory dysfunction. Heparins have beneficial effects in sepsis and have been reported to bind to histones and neutralize their cytotoxicity. The aim of this study was to investigate the impact of histones on intestinal microcirculation and the intestinal endothelium and to discuss the protective effect of unfractionated heparin (UFH) on the endothelial cytotoxicity and microcirculatory dysfunction induced by histones. METHODS Anesthetized rats were infused with 30 mg/kg calf thymus histones, and UFH was administered intravenously at a concentration of 100 IU/kg per hour. The intestinal microcirculation was visualized and measured with incident dark field microscope. Plasma von Willebrand factor (vWF) and soluble thrombomodulin were detected, and structural changes in the rat intestinal microvascular endothelium were examined. The effects of histones and UFH on cell survival rates, vWF release and calcium influx were investigated in human intestinal microvascular endothelial cells (HIMECs). RESULTS Histone infusion caused severe intestinal microcirculatory dysfunction in the absence of obvious hemodynamic changes, and UFH protected intestinal microcirculation in histone-infused rats. Concentrations of the plasma endothelial injury markers vWF and soluble thrombomodulin were elevated, and structural abnormalities were found in the intestinal microvascular endothelium in the histone-infused rats. These events were attenuated by UFH. In vitro, UFH significantly reduced the histone-induced cytotoxicity of HIMECs, reduced the release of vWF from the cytoplasm into the culture medium, and inhibited calcium influx into HIMECs. CONCLUSION Histones induce intestinal microcirculatory dysfunction followed by direct injury to the endothelial cells; UFH protects the intestinal microcirculation partly by antagonizing the endothelial toxicity of histones.
Collapse
|
43
|
Tissue damage in the heart after cardiac arrest induced by asphyxia and hemorrhage in newborn pigs. Pediatr Res 2019; 86:709-718. [PMID: 31336381 DOI: 10.1038/s41390-019-0505-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 07/03/2019] [Accepted: 07/09/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND Asphyxia of newborns is a severe and frequent challenge of the peri- and postnatal period. METHODS Forty-four neonatal piglets underwent asphyxia and hemorrhage (AH), followed by resuscitation with blood or crystalloid transfusion. In this study, 15 piglets (blood n = 9, NaCl n = 6, mean age 31 h) were randomly chosen. Four hours after return of spontaneous circulation, heart tissue and blood were collected. Analyses of heart fatty acid binding protein (HFABP), cardiac troponin I (TnI) levels, and activation of the complement system were performed. Histological staining for connexin 43 (Cx43) and complement C5a receptor 1 (C5aR1) was performed. RESULTS Following AH, systemic elevation of cardiac TnI and HFABP revealed cardiac damage in both groups. Systemic activation of the complement system and the appearance of extracellular histones in plasma of the blood transfusion group were observed. The Cx43 was translocated from the intercalated discs to the cytosol after AH. Cardiac glycogen concentration was reduced in both groups. A significant reduction of C5aR1 in the left ventricle and a significant elevation of the heart injury score were investigated after blood transfusion. CONCLUSION AH leads to alteration of the heart, particularly in Cx43 and glycogen reserves, as well as local inflammation.
Collapse
|
44
|
Lu NF, Jiang L, Zhu B, Yang DG, Zheng RQ, Shao J, Yuan J, Xi XM. Elevated Plasma Histone H4 Levels Are an Important Risk Factor in the Development of Septic Cardiomyopathy. Balkan Med J 2019; 37:72-78. [PMID: 31674172 PMCID: PMC7094183 DOI: 10.4274/balkanmedj.galenos.2019.2019.8.40] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Background: Myocardial impairment is a major complication and an important prognostic predictor of sepsis. Therefore, early and accurate diagnosis as well as timely management of septic cardiomyopathy is critical to achieve favorable outcomes. Aims: To investigate the risk factors of septic cardiomyopathy. Study Design: Cross-sectional study. Methods: This study performed between May 2016 and June 2018 recruited 93 septic patients from the intensive care unit. All patients received standardized treatments. Septic patients were divided into two groups: non cardiomyopathy (n=45) and septic cardiomyopathy group (n=48). Blood samples were collected and transthoracic echocardiography was performed within 24 hours of intensive care unit admission. Septic patients with one ultrasound abnormality but no history of heart disease were diagnosed as having septic cardiomyopathy. Plasma histones, cardiac troponin I, and N-terminal pro-brain natriuretic peptide were measured using ELISA. Sequential Organ Failure Assessment scores, vasopressor use, and the outcomes of intensive care unit stay were analyzed. Spearman rank analysis was used to determine the correlation between plasma histone H4 and other parameters. Binary logistic regression and receiver operating characteristic curve analysis were used to determine the risk factors for septic cardiomyopathy. Results: Compared with the non-cardiomyopathy group, the septic cardiomyopathy group had significantly higher plasma H4 and cardiac troponin I levels, a higher Sequential Organ Failure Assessment score, more frequent vasopressor use, and a higher mortality rate (p<0.05). Plasma histone H4 levels positively correlated with cardiac troponin I (r=0.577, p<0.001), N-terminal pro-brain natriuretic peptide (r=0.349, p=0.001), and Sequential Organ Failure Assessment scores (r=0.469, p<0.001). Binary logistic regression and receiver operating characteristic curve analyses revealed that elevated plasma histone H4 levels and vasopressor use were important risk factors for septic cardiomyopathy (p<0.05). Conclusion: Elevated plasma histone H4 levels could be used to predict septic cardiomyopathy in patients with sepsis.
Collapse
Affiliation(s)
- Nian-Fang Lu
- Clinic of Critical Care Medicine, Beijing Electric Power Hospital, Beijing, China
| | - Li Jiang
- Department of Critical Care Medicine, Capital Medical University Fuxing Hospital, Beijing, China
| | - Bo Zhu
- Department of Critical Care Medicine, Capital Medical University Fuxing Hospital, Beijing, China
| | - De-Gang Yang
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, School of Rehabilitation Medicine, Capital Medical University, Beijing, China
| | - Rui-Qiang Zheng
- Clinic of Critical Care Medicine, Subei People’s Hospital of Jiangsu Province, Jiangsu, China
| | - Jun Shao
- Clinic of Critical Care Medicine, Subei People’s Hospital of Jiangsu Province, Jiangsu, China
| | - Jing Yuan
- Clinic of Cardiac Function Tests, Subei People’s Hospital of Jiangsu Province, Jiangsu, China
| | - Xiu-Ming Xi
- Department of Critical Care Medicine, Capital Medical University Fuxing Hospital, Beijing, China
| |
Collapse
|
45
|
The Role of ALDH2 in Sepsis and the To-Be-Discovered Mechanisms. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1193:175-194. [PMID: 31368104 DOI: 10.1007/978-981-13-6260-6_10] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Sepsis, defined as life-threatening tissue damage and organ dysfunction caused by a dysregulated host response to infection, is a critical disease which imposes global health burden. Sepsis-induced organ dysfunction, including circulatory and cardiac dysfunction, hepatic dysfunction, renal dysfunction, etc., contributes to high mortality and long-term disability of sepsis patients. Altered inflammatory response, ROS and reactive aldehyde stress, mitochondrial dysfunction, and programmed cell death pathways (necrosis, apoptosis, and autophagy) have been demonstrated to play crucial roles in septic organ dysfunction. Unfortunately, except for infection control and supportive therapies, no specific therapy exists for sepsis. New specific therapeutic targets are highly warranted. Emerging studies suggested a role of potential therapeutic target of ALDH2, a tetrameric enzyme located in mitochondria to detoxify aldehydes, in septic organ dysfunction. In this article, we will review the presentations and pathophysiology of septic organ dysfunction, as well as summarize and discuss the recent insights regarding ALDH2 in sepsis.
Collapse
|
46
|
Lackner I, Weber B, Baur M, Haffner-Luntzer M, Eiseler T, Fois G, Gebhard F, Relja B, Marzi I, Pfeifer R, Halvachizadeh S, Lipiski M, Cesarovic N, Pape HC, Kalbitz M. Midkine Is Elevated After Multiple Trauma and Acts Directly on Human Cardiomyocytes by Altering Their Functionality and Metabolism. Front Immunol 2019; 10:1920. [PMID: 31552013 PMCID: PMC6736577 DOI: 10.3389/fimmu.2019.01920] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 07/29/2019] [Indexed: 12/12/2022] Open
Abstract
Background and Purpose: Post-traumatic cardiac dysfunction often occurs in multiply injured patients (ISS ≥ 16). Next to direct cardiac injury, post-traumatic cardiac dysfunction is mostly induced by the release of inflammatory biomarkers. One of these is the heparin-binding factor Midkine, which is elevated in humans after fracture, burn injury and traumatic spinal cord injury. Midkine is associated with cardiac pathologies but the exact role of Midkine in the development of those diseases is ambiguous. The systemic profile of Midkine after multiple trauma, its effects on cardiomyocytes and the association with post-traumatic cardiac dysfunction, remain unknown. Experimental Approach: Midkine levels were investigated in blood plasma of multiply injured humans and pigs. Furthermore, human cardiomyocytes (iPS) were cultured in presence/absence of Midkine and analyzed regarding viability, apoptosis, calcium handling, metabolic alterations, and oxidative stress. Finally, the Midkine filtration capacity of the therapeutic blood absorption column CytoSorb ®300 was tested with recombinant Midkine or plasma from multiply injured patients. Key Results: Midkine levels were significantly increased in blood plasma of multiply injured humans and pigs. Midkine acts on human cardiomyocytes, altering their mitochondrial respiration and calcium handling in vitro. CytoSorb®300 filtration reduced Midkine concentration ex vivo and in vitro depending on the dosage. Conclusion and Implications: Midkine is elevated in human and porcine plasma after multiple trauma, affecting the functionality and metabolism of human cardiomyocytes in vitro. Further examinations are required to determine whether the application of CytoSorb®300 filtration in patients after multiple trauma is a promising therapeutic approach to prevent post-traumatic cardiac disfunction.
Collapse
Affiliation(s)
- Ina Lackner
- Department of Traumatology, Hand, Plastic, and Reconstructive Surgery, Center of Surgery, University of Ulm, Ulm, Germany
| | - Birte Weber
- Department of Traumatology, Hand, Plastic, and Reconstructive Surgery, Center of Surgery, University of Ulm, Ulm, Germany
| | - Meike Baur
- Department of Traumatology, Hand, Plastic, and Reconstructive Surgery, Center of Surgery, University of Ulm, Ulm, Germany
| | | | - Tim Eiseler
- Department of Internal Medicine I, University of Ulm, Ulm, Germany
| | - Giorgio Fois
- Institute of General Physiology, University of Ulm, Ulm, Germany
| | - Florian Gebhard
- Department of Traumatology, Hand, Plastic, and Reconstructive Surgery, Center of Surgery, University of Ulm, Ulm, Germany
| | - Borna Relja
- Department of Trauma, Hand and Reconstructive Surgery, Goethe University Frankfurt, Frankfurt, Germany
| | - Ingo Marzi
- Department of Trauma, Hand and Reconstructive Surgery, Goethe University Frankfurt, Frankfurt, Germany
| | - Roman Pfeifer
- Department of Trauma, University Hospital of Zurich, Zurich, Switzerland
| | | | - Miriam Lipiski
- Department of Surgical Research, University Hospital of Zurich, Zurich, Switzerland
| | - Nikola Cesarovic
- Department of Surgical Research, University Hospital of Zurich, Zurich, Switzerland
| | | | - Miriam Kalbitz
- Department of Traumatology, Hand, Plastic, and Reconstructive Surgery, Center of Surgery, University of Ulm, Ulm, Germany
| | | |
Collapse
|
47
|
Ward PA, Fattahi F. New strategies for treatment of infectious sepsis. J Leukoc Biol 2019; 106:187-192. [PMID: 30821872 DOI: 10.1002/jlb.4mir1118-425r] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 01/11/2019] [Accepted: 01/29/2019] [Indexed: 12/12/2022] Open
Abstract
In this mini review, we describe the molecular mechanisms in polymicrobial sepsis that lead to a series of adverse events including activation of inflammatory and prothrombotic pathways, a faulty innate immune system, and multiorgan dysfunction. Complement activation is a well-established feature of sepsis, especially involving generation of C5a and C5b-9, along with engagement of relevant receptors for C5a. Activation of neutrophils by C5a leads to extrusion of DNA, forming neutrophil extracellular traps that contain myeloperoxidase and oxidases, along with extracellular histones. Generation of the distal complement activation product, C5b-9 (known as the membrane attack complex, MAC), also occurs in sepsis. C5b-9 activates the NLRP3 inflammasome, which damages mitochondria, together with appearance in plasma of IL-1β and IL-18. Histones are strongly proinflammatory as well as being prothrombotic, leading to activation of platelets and development of venous thrombosis. Multiorgan dysfunction is also a feature of sepsis. It is well known that septic cardiomyopathy, which if severe, can lead to death. This complication in sepsis is linked to reduced levels in cardiomyocytes of three critical proteins (SERCA2, NCX, Na+ /K+ -ATPase). The reductions in these three key proteins are complement- and histone-dependent. Dysfunction of these ATPases is linked to the cardiomyopathy of sepsis. These data suggest novel targets in the setting of sepsis in humans.
Collapse
Affiliation(s)
- Peter A Ward
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Fatemeh Fattahi
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
48
|
Braun CK, Schaffer A, Weber B, Huber-Lang M, Kalbitz M, Preßmar J. The Prognostic Value of Troponin in Pediatric Polytrauma. Front Pediatr 2019; 7:477. [PMID: 31824896 PMCID: PMC6879657 DOI: 10.3389/fped.2019.00477] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 10/30/2019] [Indexed: 12/17/2022] Open
Abstract
Introduction: Severe trauma accounts for a great number of deaths among children and adolescents. The diagnostic value of troponin serum levels of severely injured patients has been reported for adults, but data on pediatric polytrauma (PT) are scarce. Therefore, we conducted a retrospective monocentered study analyzing the prognostic value of troponin T (TnT) in pediatric trauma patients at the time point of hospital admission. Methods: Data of 88 polytraumatized pediatric patients admitted to the emergency room of the University Hospital of Ulm, Germany, between 2007 and 2016 were analyzed retrospectively. The data source was the written and digital patient records. Interleukin-6 (IL-6), creatine kinase activity (CK activity), and lactate and TnT levels were measured by a certified clinical diagnostic laboratory; and patients were stratified for the Injury Severity Score (ISS). The prognostic value for lung contusion, organ dysfunction, and fatal outcome was statistically explored. The study was approved by the independent ethical committee of the University of Ulm (#44/18). Results: TnT levels were significantly increased in patients after severe PT compared with mild or moderate trauma severity as assessed by ISS values. Patients with TnT levels above the cutoff showed significantly increased levels of IL-6 and CK activity and a significantly prolonged stay in the intensive care unit. However, TnT levels did not correlate with absolute ISS values. TnT levels were significantly increased in patients with chest trauma and lung contusion. The incidence of lung contusion was associated with elevation of TnT. So was the onset of organ dysfunction, defined as a Sequential Organ Failure Assessment (SOFA) score ≥ 2 and fatal outcome, with a significant enhancement of plasma levels in children with organ dysfunction and in non-survivors. Conclusion: These descriptive data suggest that evaluation of TnT on admission of multiply injured children may help in predicting severity of injury and mortality in the clinical course after trauma and thus may be a useful addition to established prognostic parameters in the future.
Collapse
Affiliation(s)
- Christian Karl Braun
- Institute of Clinical and Experimental Trauma-Immunology, University Hospital of Ulm, Ulm, Germany
| | - Annika Schaffer
- Department of Traumatology, Hand-, Plastic-, and Reconstructive Surgery, Center of Surgery, University Hospital of Ulm, Ulm, Germany
| | - Birte Weber
- Department of Traumatology, Hand-, Plastic-, and Reconstructive Surgery, Center of Surgery, University Hospital of Ulm, Ulm, Germany
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma-Immunology, University Hospital of Ulm, Ulm, Germany
| | - Miriam Kalbitz
- Department of Traumatology, Hand-, Plastic-, and Reconstructive Surgery, Center of Surgery, University Hospital of Ulm, Ulm, Germany
| | - Jochen Preßmar
- Department of Traumatology, Hand-, Plastic-, and Reconstructive Surgery, Center of Surgery, University Hospital of Ulm, Ulm, Germany
| |
Collapse
|
49
|
On the origin of proteins in human drusen: The meet, greet and stick hypothesis. Prog Retin Eye Res 2018; 70:55-84. [PMID: 30572124 DOI: 10.1016/j.preteyeres.2018.12.003] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 12/11/2018] [Accepted: 12/12/2018] [Indexed: 12/12/2022]
Abstract
Retinal drusen formation is not only a clinical hallmark for the development of age-related macular degeneration (AMD) but also for other disorders, such as Alzheimer's disease and renal diseases. The initiation and growth of drusen is poorly understood. Attention has focused on lipids and minerals, but relatively little is known about the origin of drusen-associated proteins and how they are retained in the space between the basal lamina of the retinal pigment epithelium and the inner collagenous layer space (sub-RPE-BL space). While some authors suggested that drusen proteins are mainly derived from cellular debris from processed photoreceptor outer segments and the RPE, others suggest a choroidal cell or blood origin. Here, we reviewed and supplemented the existing literature on the molecular composition of the retina/choroid complex, to gain a more complete understanding of the sources of proteins in drusen. These "drusenomics" studies showed that a considerable proportion of currently identified drusen proteins is uniquely originating from the blood. A smaller, but still large fraction of drusen proteins comes from both blood and/or RPE. Only a small proportion of drusen proteins is uniquely derived from the photoreceptors or choroid. We next evaluated how drusen components may "meet, greet and stick" to each other and/or to structures like hydroxyapatite spherules to form macroscopic deposits in the sub-RPE-BL space. Finally, we discuss implications of our findings with respect to the previously proposed homology between drusenogenesis in AMD and plaque formation in atherosclerosis.
Collapse
|
50
|
Wang Z, Hu W, Lu C, Ma Z, Jiang S, Gu C, Acuña-Castroviejo D, Yang Y. Targeting NLRP3 (Nucleotide-Binding Domain, Leucine-Rich–Containing Family, Pyrin Domain–Containing-3) Inflammasome in Cardiovascular Disorders. Arterioscler Thromb Vasc Biol 2018; 38:2765-2779. [PMID: 30571177 DOI: 10.1161/atvbaha.118.311916] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Inflammation is an important innate immune response to infection or tissue damage. Inflammasomes are involved in the onset and development of inflammation. The NLRP3 (nucleotide-binding domain, leucine-rich–containing family, pyrin domain–containing-3) inflammasome is the best-characterized inflammasome. Recent evidence has indicated the importance of the NLRP3 inflammasome in the pathophysiology of cardiovascular disorders. To further understand the roles of the NLRP3 inflammasome in the cardiovascular system, we provide a comprehensive overview and discuss the remaining questions. First, a summary of NLRP3 inflammasome in the cardiovascular system is introduced. Then, the associations between NLRP3 inflammasome and cardiovascular disorders are presented. Finally, we discuss existing problems and potential directions with this issue. The information compiled here summarizes recent progress, thus potentially aiding in the understanding of the NLRP3 inflammasome in cardiovascular disorders, designing experimental and clinical research about the NLRP3 inflammasome, and promoting therapeutics for cardiovascular disorders.
Collapse
Affiliation(s)
- Zheng Wang
- From the Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, Taibai, Xi’an, China (Z.W., C.L., Y.Y.)
- Department of Cardiothoracic Surgery, Wuhan General Hospital of The People’s Liberation Army, China (Z.W.)
| | - Wei Hu
- Department of Immunology (W.H.), The Fourth Military Medical University, Xi’an, China
| | - Chenxi Lu
- From the Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, Taibai, Xi’an, China (Z.W., C.L., Y.Y.)
| | - Zhiqiang Ma
- Department of Thoracic Surgery, Tangdu Hospital (Z.M.), The Fourth Military Medical University, Xi’an, China
| | - Shuai Jiang
- Department of Aerospace Medicine (S.J.), The Fourth Military Medical University, Xi’an, China
| | - Chunhu Gu
- Department of Cardiovascular Surgery, Xijing Hospital (C.G.), The Fourth Military Medical University, Xi’an, China
| | - Darío Acuña-Castroviejo
- Biomedical Research Center, Health Sciences Technology Park, University of Granada, Spain (D.A.-C.)
| | - Yang Yang
- From the Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, Taibai, Xi’an, China (Z.W., C.L., Y.Y.)
| |
Collapse
|