1
|
Culkin MC, Bele P, Georges AP, Santos P, Niziolek G, Kaplan LJ, Smith DH, Pascual JL. Dose-Dependent Tranexamic Acid Blunting of Penumbral Leukocyte Mobilization and Blood-Brain Barrier Permeability Following Traumatic Brain Injury: An In Vivo Murine Study. Neurocrit Care 2024; 41:469-478. [PMID: 38443709 DOI: 10.1007/s12028-024-01952-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 01/29/2024] [Indexed: 03/07/2024]
Abstract
BACKGROUND Early posttraumatic brain injury (TBI) tranexamic acid (TXA) may reduce blood-brain barrier (BBB) permeability, but it is unclear if this effect is fixed regardless of dose. We hypothesized that post-TBI TXA demonstrates a dose-dependent reduction of in vivo penumbral leukocyte mobilization, BBB microvascular permeability, and enhancement of neuroclinical recovery. METHODS CD1 male mice (n = 40) were randomly assigned to TBI by controlled cortical impact (injury [I]) or sham TBI (S), followed by intravenous bolus of either saline (placebo [P]) or TXA (15, 30, or 60 mg/kg). At 48 h, in vivo pial intravital microscopy visualized live penumbral BBB microvascular leukocytes and albumin leakage. Neuroclinical recovery was assessed by Garcia Neurological Test scores and animal weight changes at 24 h and 48 h after injury. RESULTS I + TXA60 reduced live penumbral leukocyte rolling compared with I + P (p < 0.001) and both lower TXA doses (p = 0.017 vs. I + TXA15, p = 0.012 vs. I + TXA30). Leukocyte adhesion was infrequent and similar across groups. Only I + TXA60 significantly reduced BBB permeability compared with that in the I + P (p = 0.004) group. All TXA doses improved Garcia Test scores relative to I + P at both 24 h and 48 h (p < 0.001 vs. I + P for all at both time points). Mean 24-h body weight loss was greatest in the I + P (- 8.7 ± 1.3%) group and lowest in the I + TXA15 (- 4.4 ± 1.0%, p = 0.051 vs. I + P) group. CONCLUSIONS Only higher TXA dosing definitively abrogates penumbral leukocyte mobilization, preserving BBB integrity post TBI. Some neuroclinical recovery is observed, even with lower TXA dosing. Better outcomes with higher dose TXA after TBI may occur secondary to blunting of leukocyte-mediated penumbral cerebrovascular inflammation.
Collapse
Affiliation(s)
- Matthew C Culkin
- Division of Traumatology, Surgical Critical Care and Emergency Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, SICU Administration Office - 5 Founders Pavilion, 3400 Spruce Street, Philadelphia, PA, 19104, USA
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Priyanka Bele
- Division of Traumatology, Surgical Critical Care and Emergency Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, SICU Administration Office - 5 Founders Pavilion, 3400 Spruce Street, Philadelphia, PA, 19104, USA
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Anastasia P Georges
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Patricia Santos
- Division of Traumatology, Surgical Critical Care and Emergency Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, SICU Administration Office - 5 Founders Pavilion, 3400 Spruce Street, Philadelphia, PA, 19104, USA
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Grace Niziolek
- Division of Traumatology, Surgical Critical Care and Emergency Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, SICU Administration Office - 5 Founders Pavilion, 3400 Spruce Street, Philadelphia, PA, 19104, USA
| | - Lewis J Kaplan
- Division of Traumatology, Surgical Critical Care and Emergency Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, SICU Administration Office - 5 Founders Pavilion, 3400 Spruce Street, Philadelphia, PA, 19104, USA
| | - Douglas H Smith
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jose L Pascual
- Division of Traumatology, Surgical Critical Care and Emergency Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, SICU Administration Office - 5 Founders Pavilion, 3400 Spruce Street, Philadelphia, PA, 19104, USA.
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
2
|
Prokazyuk A, Tlemissov A, Zhanaspayev M, Aubakirova S, Mussabekov A. Development and validation of a machine learning-based model to assess probability of systemic inflammatory response syndrome in patients with severe multiple traumas. BMC Med Inform Decis Mak 2024; 24:235. [PMID: 39192291 DOI: 10.1186/s12911-024-02640-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: 05/09/2024] [Accepted: 08/20/2024] [Indexed: 08/29/2024] Open
Abstract
BACKGROUND Systemic inflammatory response syndrome (SIRS) is a predictor of serious infectious complications, organ failure, and death in patients with severe polytrauma and is one of the reasons for delaying early total surgical treatment. To determine the risk of SIRS within 24 h after hospitalization, we developed six machine learning models. MATERIALS AND METHODS Using retrospective data about the patient, the nature of the injury, the results of general and standard biochemical blood tests, and coagulation tests, six models were developed: decision tree, random forest, logistic regression, support vector and gradient boosting classifiers, logistic regressor, and neural network. The effectiveness of the models was assessed through internal and external validation. RESULTS Among the 439 selected patients with severe polytrauma in 230 (52.4%), SIRS was diagnosed within the first 24 h of hospitalization. The SIRS group was more strongly associated with class II bleeding (39.5% vs. 60.5%; OR 1.81 [95% CI: 1.23-2.65]; P = 0.0023), long-term vasopressor use (68.4% vs. 31.6%; OR 5.51 [95% CI: 2.37-5.23]; P < 0.0001), risk of acute coagulopathy (67.8% vs. 32.2%; OR 2.4 [95% CI: 1.55-3.77]; P < 0.0001), and greater risk of pneumonia (59.5% vs. 40.5%; OR 1.74 [95% CI: 1.19-2.54]; P = 0.0042), longer ICU length of stay (5 ± 6.3 vs. 2.7 ± 4.3 days; P < 0.0001) and mortality rate (64.5% vs. 35.5%; OR 10.87 [95% CI: 6.3-19.89]; P = 0.0391). Of all the models, the random forest classifier showed the best predictive ability in the internal (AUROC 0.89; 95% CI: 0.83-0.96) and external validation (AUROC 0.83; 95% CI: 0.75-0.91) datasets. CONCLUSIONS The developed model made it possible to accurately predict the risk of developing SIRS in the early period after injury, allowing clinical specialists to predict patient management tactics and calculate medication and staffing needs for the patient. LEVEL OF EVIDENCE Level 3. TRIAL REGISTRATION The study was retrospectively registered in the ClinicalTrials.gov database of the National Library of Medicine (NCT06323096).
Collapse
Affiliation(s)
- Alexander Prokazyuk
- University Hospital of Non-Commercial Joint-Stock Company "Semey Medical University", 1a, Ivan Sechenov str, Semey city, 071400, Republic of Kazakhstan.
| | - Aidos Tlemissov
- Center of habilitation and rehabilitation of persons with disabilities of the Abai region, 109, Karagaily, Semey city, 071400, Republic of Kazakhstan
| | - Marat Zhanaspayev
- Non-Commercial Joint-Stock Company "Semey Medical University", 103, Abai Kunanbayev str, Semey city, 071400, Republic of Kazakhstan
| | - Sabina Aubakirova
- Non-Commercial Joint-Stock Company "Semey Medical University", 103, Abai Kunanbayev str, Semey city, 071400, Republic of Kazakhstan
| | - Arman Mussabekov
- Non-Commercial Joint-Stock Company "Semey Medical University", 103, Abai Kunanbayev str, Semey city, 071400, Republic of Kazakhstan
| |
Collapse
|
3
|
Rios KE, Alamneh Y, Werner LM, Leung C, Pavlovic R, Abu-Taleb R, Thanapaul RJRS, Lee S, Hull D, Czintos C, Su W, Getnet D, Antonic V, Bobrov AG. Optimization of a Lethal, Combat-Relevant Model of Sterile Inflammation in Mice for Drug Candidate Screening. Mil Med 2024; 189:694-701. [PMID: 39160880 DOI: 10.1093/milmed/usae233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 03/09/2024] [Accepted: 04/19/2024] [Indexed: 08/21/2024] Open
Abstract
INTRODUCTION Extensive trauma, commonly seen in wounded military Service Members, often leads to a severe sterile inflammation termed systemic inflammatory response syndrome (SIRS), which can progress to multiple organ dysfunction syndrome (MODS) and death. MODS is a serious threat to wounded Service Members, historically causing 10% of all deaths in trauma admissions at a forward deployed combat hospital. The importance of this problem will be exacerbated in large-scale combat operations, in which evacuation will be delayed and care of complex injuries at lower echelons of care may be prolonged. The main goal of this study was to optimize an existing mouse model of lethal SIRS/MODS as a therapeutic screening platform for the evaluation of immunomodulatory drugs. MATERIALS AND METHODS Male C57BL/6 mice were euthanized, and the bones and muscles were collected and blended into a paste termed tissue-bone matrix (TBX). The TBX at 12.5%-20% relative to body weight of each recipient mouse was implanted into subcutaneous pouches created on the dorsum of anesthetized animals. Mice were observed for clinical scores for up to 48 hours postimplantation and euthanized at the preset point of moribundity. To test effects of anesthetics on TBX-induced mortality, animals received isoflurane or ketamine/xylazine (K/X). In a separate set of studies, mice received TBX followed by intraperitoneal injection with 20 mg/kg or 40 mg/kg Eritoran or a placebo carrier. All Eritoran studies were performed in a blinded fashion. RESULTS We observed that K/X anesthesia significantly increased the lethality of the implanted TBX in comparison to inhaled anesthetics. Although all the mice anesthetized with isoflurane and implanted with 12.5% TBX survived for 24 hours, 60% of mice anesthetized with K/X were moribund by 24 hours postimplantation. To mimic more closely the timing of lethal SIRS/MODS following polytrauma in human patients, we extended observation to 48 hours. We performed TBX dose-response studies and found that as low as 15%, 17.5%, and 20% TBX caused moribundity/mortality in 50%, 80%, and 100% mice, respectively, over a 48-hour time period. With 17.5% TBX, we tested if moribundity/mortality could be rescued by anti-inflammatory drug Eritoran, a toll-like receptor 4 antagonist. Neither 20 mg/kg nor 40 mg/kg doses of Eritoran were found to be effective in this model. CONCLUSIONS We optimized a TBX mouse model of SIRS/MODS for the purpose of evaluating novel therapeutic interventions to prevent trauma-related pathophysiologies in wounded Service Members. Negative effects of K/X on lethality of TBX should be further evaluated, particularly in the light of widespread use of ketamine in treatment of pain. By mimicking muscle crush, bone fracture, and necrosis, the TBX model has pleiotropic effects on physiology and immunology that make it uniquely valuable as a screening tool for the evaluation of novel therapeutics against trauma-induced SIRS/MODS.
Collapse
Affiliation(s)
- Kariana E Rios
- Wound Infections Department, Bacterial Diseases Branch, Center for Infectious Diseases Research, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Oak Ridge Institute for Science and Education, Oak Ridge, TN 37830, USA
| | - Yonas Alamneh
- Wound Infections Department, Bacterial Diseases Branch, Center for Infectious Diseases Research, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Lacie M Werner
- Wound Infections Department, Bacterial Diseases Branch, Center for Infectious Diseases Research, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Clara Leung
- Wound Infections Department, Bacterial Diseases Branch, Center for Infectious Diseases Research, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Radmila Pavlovic
- Wound Infections Department, Bacterial Diseases Branch, Center for Infectious Diseases Research, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Rania Abu-Taleb
- Wound Infections Department, Bacterial Diseases Branch, Center for Infectious Diseases Research, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Rex J R S Thanapaul
- Wound Infections Department, Bacterial Diseases Branch, Center for Infectious Diseases Research, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- NRC Research Associateship Programs, National Academies of Sciences, Engineering, and Medicine, Washington, DC 20001, USA
| | - Sunjoo Lee
- Wound Infections Department, Bacterial Diseases Branch, Center for Infectious Diseases Research, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Dawn Hull
- Wound Infections Department, Bacterial Diseases Branch, Center for Infectious Diseases Research, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Christine Czintos
- Wound Infections Department, Bacterial Diseases Branch, Center for Infectious Diseases Research, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Oak Ridge Institute for Science and Education, Oak Ridge, TN 37830, USA
| | - Wanwen Su
- Wound Infections Department, Bacterial Diseases Branch, Center for Infectious Diseases Research, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Derese Getnet
- Wound Infections Department, Bacterial Diseases Branch, Center for Infectious Diseases Research, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Vlado Antonic
- Wound Infections Department, Bacterial Diseases Branch, Center for Infectious Diseases Research, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Alexander G Bobrov
- Wound Infections Department, Bacterial Diseases Branch, Center for Infectious Diseases Research, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| |
Collapse
|
4
|
Ullah H, Arbab S, Tian Y, Chen Y, Liu CQ, Li Q, Li K. Crosstalk between gut microbiota and host immune system and its response to traumatic injury. Front Immunol 2024; 15:1413485. [PMID: 39144142 PMCID: PMC11321976 DOI: 10.3389/fimmu.2024.1413485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 06/04/2024] [Indexed: 08/16/2024] Open
Abstract
Millions of microorganisms make up the complex microbial ecosystem found in the human gut. The immune system's interaction with the gut microbiota is essential for preventing inflammation and maintaining intestinal homeostasis. Numerous metabolic products that can cross-talk between immune cells and the gut epithelium are metabolized by the gut microbiota. Traumatic injury elicits a great and multifaceted immune response in the minutes after the initial offense, containing simultaneous pro- and anti-inflammatory responses. The development of innovative therapies that improve patient outcomes depends on the gut microbiota and immunological responses to trauma. The altered makeup of gut microbes, or gut dysbiosis, can also dysregulate immunological responses, resulting in inflammation. Major human diseases may become more common as a result of chronic dysbiosis and the translocation of bacteria and the products of their metabolism beyond the mucosal barrier. In this review, we briefly summarize the interactions between the gut microbiota and the immune system and human disease and their therapeutic probiotic formulations. We also discuss the immune response to traumatic injury.
Collapse
Affiliation(s)
- Hanif Ullah
- Medicine and Engineering Interdisciplinary Research Laboratory of Nursing & Materials/Nursing Key Laboratory of Sichuan Province, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, Sichuan, China
| | - Safia Arbab
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Yali Tian
- Medicine and Engineering Interdisciplinary Research Laboratory of Nursing & Materials/Nursing Key Laboratory of Sichuan Province, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, Sichuan, China
| | - Yuwen Chen
- Medicine and Engineering Interdisciplinary Research Laboratory of Nursing & Materials/Nursing Key Laboratory of Sichuan Province, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, Sichuan, China
| | - Chang-qing Liu
- Medicine and Engineering Interdisciplinary Research Laboratory of Nursing & Materials/Nursing Key Laboratory of Sichuan Province, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, Sichuan, China
| | - Qijie Li
- Medicine and Engineering Interdisciplinary Research Laboratory of Nursing & Materials/Nursing Key Laboratory of Sichuan Province, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, Sichuan, China
| | - Ka Li
- Medicine and Engineering Interdisciplinary Research Laboratory of Nursing & Materials/Nursing Key Laboratory of Sichuan Province, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
5
|
Banderwal R, Kadian M, Garg S, Kumar A. 'Comprehensive review of emerging drug targets in traumatic brain injury (TBI): challenges and future scope. Inflammopharmacology 2024:10.1007/s10787-024-01524-w. [PMID: 39023681 DOI: 10.1007/s10787-024-01524-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 02/12/2024] [Indexed: 07/20/2024]
Abstract
Traumatic brain injury (TBI) is a complex brain problem that causes significant morbidity and mortality among people of all age groups. The complex pathophysiology, varied symptoms, and inadequate treatment further precipitate the problem. Further, TBI produces several psychiatric problems and other related complications in post-TBI survival patients, which are often treated symptomatically or inadequately. Several approaches, including neuroprotective agents targeting several pathways of oxidative stress, neuroinflammation, cytokines, immune system GABA, glutamatergic, microglia, and astrocytes, are being tried by researchers to develop effective treatments or magic bullets to manage the condition effectively. The problem of TBI is therefore treated as a challenge among pharmaceutical scientists or researchers to develop drugs for the effective management of this problem. The goal of the present comprehensive review is to provide an overview of the several pharmacological targets, processes, and cellular pathways that researchers are focusing on, along with an update on their current state.
Collapse
Affiliation(s)
- Rittu Banderwal
- Pharmacology Division, University Institute of Pharmaceutical Sciences (UIPS), UGC- Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, 160014, India
| | - Monika Kadian
- Pharmacology Division, University Institute of Pharmaceutical Sciences (UIPS), UGC- Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, 160014, India
| | - Sukant Garg
- Department of General Pathology, Dr HS Judge Institute of Dental Sciences and Hospital, Panjab University, Chandigarh, 160014, India
| | - Anil Kumar
- Pharmacology Division, University Institute of Pharmaceutical Sciences (UIPS), UGC- Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, 160014, India.
| |
Collapse
|
6
|
Roos J, Zinngrebe J, Huber-Lang M, Lupu L, Schmidt MA, Strobel H, Westhoff MA, Stifel U, Gebhard F, Wabitsch M, Mollnes TE, Debatin KM, Halbgebauer R, Fischer-Posovszky P. Trauma-associated extracellular histones mediate inflammation via a MYD88-IRAK1-ERK signaling axis and induce lytic cell death in human adipocytes. Cell Death Dis 2024; 15:285. [PMID: 38653969 PMCID: PMC11039744 DOI: 10.1038/s41419-024-06676-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 04/09/2024] [Accepted: 04/11/2024] [Indexed: 04/25/2024]
Abstract
Despite advances in the treatment and care of severe physical injuries, trauma remains one of the main reasons for disability-adjusted life years worldwide. Trauma patients often suffer from disturbances in energy utilization and metabolic dysfunction, including hyperglycemia and increased insulin resistance. White adipose tissue plays an essential role in the regulation of energy homeostasis and is frequently implicated in traumatic injury due to its ubiquitous body distribution but remains poorly studied. Initial triggers of the trauma response are mainly damage-associated molecular patterns (DAMPs) such as histones. We hypothesized that DAMP-induced adipose tissue inflammation contributes to metabolic dysfunction in trauma patients. Therefore, we investigated whether histone release during traumatic injury affects adipose tissue. Making use of a murine polytrauma model with hemorrhagic shock, we found increased serum levels of histones accompanied by an inflammatory response in white adipose tissue. In vitro, extracellular histones induced an inflammatory response in human adipocytes. On the molecular level, this inflammatory response was mediated via a MYD88-IRAK1-ERK signaling axis as demonstrated by pharmacological and genetic inhibition. Histones also induced lytic cell death executed independently of caspases and RIPK1 activity. Importantly, we detected increased histone levels in the bloodstream of patients after polytrauma. Such patients might benefit from a therapy consisting of activated protein C and the FDA-approved ERK inhibitor trametinib, as this combination effectively prevented histone-mediated effects on both, inflammatory gene activation and cell death in adipocytes. Preventing adipose tissue inflammation and adipocyte death in patients with polytrauma could help minimize posttraumatic metabolic dysfunction.
Collapse
Affiliation(s)
- Julian Roos
- Department of Pediatrics and Adolescent Medicine, University Medical Center, Ulm, Germany
| | - Julia Zinngrebe
- Department of Pediatrics and Adolescent Medicine, University Medical Center, Ulm, Germany
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma Immunology, University Medical Center, Ulm, Germany
| | - Ludmila Lupu
- Institute of Clinical and Experimental Trauma Immunology, University Medical Center, Ulm, Germany
| | - Miriam A Schmidt
- Department of Pediatrics and Adolescent Medicine, University Medical Center, Ulm, Germany
| | - Hannah Strobel
- Department of Pediatrics and Adolescent Medicine, University Medical Center, Ulm, Germany
| | - Mike-Andrew Westhoff
- Department of Pediatrics and Adolescent Medicine, University Medical Center, Ulm, Germany
| | - Ulrich Stifel
- Department of Pediatrics and Adolescent Medicine, University Medical Center, Ulm, Germany
| | - Florian Gebhard
- Department of Orthopedic Trauma, Hand, and Reconstructive Surgery, University Medical Center, Ulm, Germany
| | - Martin Wabitsch
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, University Medical Center, Ulm, Germany
| | - Tom Eirik Mollnes
- Department of Immunology, Oslo University Hospital and University of Oslo, Oslo, Norway
- Research Laboratory, Nordland Hospital Trust, Bodo, Norway
| | - Klaus-Michael Debatin
- Department of Pediatrics and Adolescent Medicine, University Medical Center, Ulm, Germany
| | - Rebecca Halbgebauer
- Institute of Clinical and Experimental Trauma Immunology, University Medical Center, Ulm, Germany
| | | |
Collapse
|
7
|
Hu Z, Li J, Zhang F, Jacob A, Wang P. A NOVEL OLIGONUCLEOTIDE MRNA MIMIC ATTENUATES HEMORRHAGE-INDUCED ACUTE LUNG INJURY. Shock 2024; 61:630-637. [PMID: 38300836 PMCID: PMC11009070 DOI: 10.1097/shk.0000000000002309] [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] [Indexed: 02/03/2024]
Abstract
ABSTRACT Hemorrhagic shock (HS) is accompanied by a pronounced activation of the inflammatory response in which acute lung injury (ALI) is one of the most frequent consequences. Among the pivotal orchestrators of this inflammatory cascade, extracellular cold-inducible RNA-binding protein (eCIRP) emerges as a noteworthy focal point, rendering it as a promising target for the management of inflammation and tissue injury. Recently, we have reported that oligonucleotide poly(A) mRNA mimic termed A 12 selectively binds to the RNA binding region of eCIRP and inhibits eCIRP binding to its receptor TLR4. Furthermore, in vivo administration of eCIRP induces lung injury in healthy mice and that mouse deficient in CIRP showed protection from inflammation-associated lung injury. We hypothesize that A 12 inhibits systemic inflammation and ALI in HS. To test the impacts of A 12 on systemic and lung inflammation, extent of inflammatory cellular infiltration and resultant lung damage were evaluated in a mouse model of HS. Male mice were subjected to controlled hemorrhage with a mean arterial pressure of 30 mm Hg for 90 min and then resuscitated with Ringer's lactate solution containing phosphate-buffered saline (vehicle) or A 12 at a dose of 4 nmol/g body weight (treatment). The infusion volume was twice that of the shed blood. At 4 h after resuscitation, mice were euthanized, and blood and lung tissues were harvested. Blood and tissue markers of inflammation and injury were evaluated. Serum markers of injury (lactate dehydrogenase, alanine transaminase, and blood urea nitrogen) and inflammation (TNF-α, IL-6) were increased after HS and A 12 treatment significantly decreased their levels. A 12 treatment also decreased lung levels of TNF-α, MIP-2, and KC mRNA expressions. Lung histological injury score, neutrophil infiltration (Ly6G staining and myeloperoxidase activity), and lung apoptosis were significantly attenuated after A 12 treatment. Our study suggests that the capacity of A 12 in attenuating HS-induced ALI and may provide novel perspectives in developing efficacious pharmaceutics for improving hemorrhage prognosis.
Collapse
Affiliation(s)
- Zhijian Hu
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, New York, United States
| | - Jingsong Li
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, New York, United States
| | - Fangming Zhang
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, New York, United States
| | - Asha Jacob
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, New York, United States
- Departments of Surgery and Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, New York, United States
| | - Ping Wang
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, New York, United States
- Departments of Surgery and Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, New York, United States
| |
Collapse
|
8
|
Komaru Y, Ning L, Lama C, Suresh A, Kefaloyianni E, Miller MJ, Herrlich A. Sterile kidney tissue injury induces neutrophil swarming in lung alveolar capillaries. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.27.582396. [PMID: 38464306 PMCID: PMC10925262 DOI: 10.1101/2024.02.27.582396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Sterile tissue injury, such as by acute kidney injury, is common in the clinic and frequently associated with respiratory compromise and hypoxemia. We previously described signaling components released by the injured kidney that drive a remote inflammatory response in the lung. How this caused the resultant hypoxemia remained unclear. Here, we report that sterile kidney tissue injury induces rapid intravascular "neutrophil train" formation in lung capillaries, a novel form of neutrophil swarming. Rapid swarming is enhanced by decreased deformability of circulating neutrophils that impedes their lung capillary passage. Classical lung monocytes are required for neutrophil train formation and release CXCL2 to attract and retain stiffened neutrophils in lung capillaries which reduces capillary perfusion. We thus discovered a novel feature of kidney-lung crosstalk after sterile kidney tissue injury, capillary perfusion deficits that lead to reduced oxygenation despite proper alveolar function and ventilation, unlike in infectious inflammatory lung processes, such as bacterial pneumonia.
Collapse
|
9
|
Zhang W, Jin C, Zhang S, Wu L, Li B, Shi M. Gut lymph purification alleviates acute lung injury induced by intestinal ischemia-reperfusion in rats by removing danger-associated molecular patterns from gut lymph. Heliyon 2024; 10:e25711. [PMID: 38371985 PMCID: PMC10873747 DOI: 10.1016/j.heliyon.2024.e25711] [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: 09/14/2023] [Revised: 01/25/2024] [Accepted: 02/01/2024] [Indexed: 02/20/2024] Open
Abstract
Background The potential effect of removing danger-associated molecular patterns (DAMPs) from gut lymph on reducing acute lung injury (ALI) induced by gut ischemia-reperfusion injury (GIRI) is uncertain. This study aimed to investigate whether gut lymph purification (GLP) could improve GIRI-induced acute lung injury in rats by clearing danger-associated molecular patterns. Materials and methods Rats were divided into four groups: Sham, GIRI, GIRI + gut lymph drainage (GLD), and GIRI + GLP. After successful modeling, lung tissue samples were collected from rats for hematoxylin-eosin (HE) staining and detection of apoptotic indexes. We detected the DAMPs levels in blood and lymph samples. We observed the microstructure of AEC Ⅱ and measured the expression levels of apoptosis indexes. Results The GIRI group showed destruction of alveolar structure, thickened alveolar walls, and inflammatory cell infiltration. This was accompanied by significantly increased levels of high mobility group protein-1 (HMGB-1) and Interleukin-6 (IL-6), while reduced levels of heat shock protein 70 (HSP 70) and Interleukin-10 (IL-10) in both lymph and serum. In contrast, the lung tissue damage in the GIRI + GLP group was significantly improved compared to the GIRI group. This was evidenced by a reduction in the expression levels of HMGB-1 and IL-6 in both lymph and serum and an increase in HSP 70 and IL-10 levels. Additionally, organelle structure of AEC II was significantly improved in the GIRI + GLP group compared to the GIRI group. Conclusions GLP inhibits inflammation and cell apoptosis in GIRI-induced ALI by blocking the link between DAMPs and mononuclear phagocytes, reducing the severity of ALI.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Critical Care Medicine, The People's Hospital of Leshan, Leshan City, Sichuan Province, 614008, China
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Can Jin
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
- Zunyi Medical University, Zunyi, Guizhou, China
| | | | - Linlin Wu
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
- Zunyi Medical University, Zunyi, Guizhou, China
| | - Bohan Li
- Zunyi Medical University, Zunyi, Guizhou, China
| | - Meimei Shi
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. School of Medicine, Northwest University. Xi'an, 710069, Shanxi, China
| |
Collapse
|
10
|
Xian L, Cheng S, Chen W, Zhong C, Hu Z, Deng X. Systematic analysis of MASP-1 serves as a novel immune-related biomarker in sepsis and trauma followed by preliminary experimental validation. Front Med (Lausanne) 2024; 11:1320811. [PMID: 38384415 PMCID: PMC10879275 DOI: 10.3389/fmed.2024.1320811] [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: 10/13/2023] [Accepted: 01/08/2024] [Indexed: 02/23/2024] Open
Abstract
Background Dysregulated immune response in trauma and sepsis leads to the abnormal activation of the complement and coagulation systems. Mannose-binding lectin (MBL)-associated serine protease-1 (MASP-1) activates the lectin pathway of the complement system and mediates proinflammatory and procoagulant reactions. However, the potential effects of MASP-1 in trauma and sepsis have not yet been explored. Methods We obtained five sepsis, two trauma, and one sepsis and trauma RNA-sequencing dataset from the Gene Expression Omnibus (GEO) database and conducted a comprehensive evaluation of the expression pattern, biological functions, and diagnostic value of MASP-1 in trauma and sepsis. Additionally, we investigated the association between MASP-1 expression and clinicopathological characteristics of trauma and sepsis. Furthermore, we collected clinical specimens to preliminarily validate the expression level and diagnostic efficacy of MASP-1 as well as the correlation of MASP-1 with clinical features of trauma and sepsis. Subsequently, we conducted a correlation analysis among MASP-1, immune cell infiltration, and immune and molecular pathways. Finally, we mechanistically analyzed the relationship among MASP-1, specific immune cells, and pivotal molecular pathways. Results MASP-1 expression was significantly upregulated in the trauma/sepsis samples compared to the control samples in the GEO datasets. MASP-1 exhibited excellent diagnostic values (AUC > 0.7) in multiple datasets and at multiple time points and could efficiently distinguish trauma/sepsis samples from the control samples. Moreover, MASP-1 expression was significantly positively correlated with the severity of the disease (APACHE-II, CRP, and neutrophil levels). These results were further validated by real-time quantitative polymerase chain reaction and enzyme-linked immunosorbent assay. Functional enrichment analysis revealed that MASP-1 primarily promotes trauma and sepsis via the immune-related signaling pathway. MASP-1 was significantly correlated with the infiltration of specific immune cells (such as B cells, CD8 T cells, neutrophils, macrophages, and infiltrating lymphocytes) and immune and molecular pathways (such as checkpoint, HLA, IL6/JAK/STAT3 signaling, necrosis, T-cell co-inhibition, and T-cell co-stimulation). Finally, analysis of the transcription and single-cell data revealed that MASP-1 was specifically expressed in T cells, and further correlation analysis revealed a close correlation between MASP-1 expression, proportion of CD8 T cells, and IL6/JAK/STAT3 signaling scores. Conclusion Our results suggest that MASP-1 can serve as an immune-related biomarker for the diagnosis and disease severity of trauma and sepsis. It may activate the IL6 JAK-STAT3 signaling pathway and promote CD8 T-cell depletion to trigger traumatic sepsis.
Collapse
Affiliation(s)
- Lina Xian
- Department of Intensive Care Unit, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Shaowen Cheng
- Department of Wound Repair, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Wei Chen
- Department of Intensive Care Unit, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Changhui Zhong
- Department of Intensive Care Unit, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Zhihua Hu
- Department of Intensive Care Unit, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Xiaoyan Deng
- Department of Intensive Care Unit, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| |
Collapse
|
11
|
Gando S, Levi M, Toh CH. Trauma-induced innate immune activation and disseminated intravascular coagulation. J Thromb Haemost 2024; 22:337-351. [PMID: 37816463 DOI: 10.1016/j.jtha.2023.09.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 09/25/2023] [Accepted: 09/26/2023] [Indexed: 10/12/2023]
Abstract
Dysregulated innate immunity participates in the pathomechanisms of disseminated intravascular coagulation (DIC) in trauma-induced coagulopathy. Accidental and regulated cell deaths and neutrophil extracellular traps release damage-associated molecular patterns (DAMPs), such as histones, nuclear and mitochondrial DNA, and high-mobility group box 1, into circulation immediately after trauma. DAMP-induced inflammation activation releases tissue factor-bearing procoagulant extracellular vesicles through gasdermin D-mediated pore formation and plasma membrane rupture by regulated cell death. DAMPs also evoke systemic inflammation, platelet, coagulation activation, and impaired fibrinolysis associated with endothelial injury, leading to the dysfunction of anticoagulation systems, which are the main pathophysiological mechanisms of DIC. All these processes induce systemic thrombin generation in vivo, not restricted to the injury sites immediately after trauma. Thrombin generation at the site of injury stops bleeding and maintains homeostasis. However, DIC associated with endothelial injury generates massive thrombin, enhancing protease-activated, receptor-mediated bidirectional interplays between inflammation and coagulation, aggravating the diverse actions of thrombin and disturbing homeostasis. Insufficiently regulated thrombin causes disseminated microvascular thrombosis, resulting in tissue hypoxia due to reduced oxygen delivery, and mitochondrial dysfunction due to DAMPs causes tissue dysoxia. In addition, DAMP-induced calcium influx and overload, as well as neutrophil activation, play a role in endothelial cell injury. Tissue hypoxia and cytotoxicity result in multiple organ dysfunction in DIC after trauma. Controls against dysregulated innate immunity evoking systemic inflammation, thrombin generation, and cytotoxicity are key issues in improving the prognosis of DIC in trauma-induced coagulopathy.
Collapse
Affiliation(s)
- Satoshi Gando
- Department of Acute and Critical Care Medicine, Sapporo Higashi Tokushukai Hospital, Sapporo, Japan; Division of Acute and Critical Care Medicine, Department of Anesthesiology and Critical Care Medicine, Hokkaido University Faculty of Medicine, Sapporo, Japan.
| | - Marcel Levi
- Department of Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands; Department of Medicine, University College London Hospitals NHS Foundation Trust, and Cardio-Metabolic Program - NIHR UCLH/UCL BRC London, London, United Kingdom
| | - Cheng-Hock Toh
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, United Kingdom; Liverpool University Hospitals NHS Foundation Trust, Liverpool, United Kingdom
| |
Collapse
|
12
|
Yamaga S, Aziz M, Murao A, Brenner M, Wang P. DAMPs and radiation injury. Front Immunol 2024; 15:1353990. [PMID: 38333215 PMCID: PMC10850293 DOI: 10.3389/fimmu.2024.1353990] [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: 12/11/2023] [Accepted: 01/15/2024] [Indexed: 02/10/2024] Open
Abstract
The heightened risk of ionizing radiation exposure, stemming from radiation accidents and potential acts of terrorism, has spurred growing interests in devising effective countermeasures against radiation injury. High-dose ionizing radiation exposure triggers acute radiation syndrome (ARS), manifesting as hematopoietic, gastrointestinal, and neurovascular ARS. Hematopoietic ARS typically presents with neutropenia and thrombocytopenia, while gastrointestinal ARS results in intestinal mucosal injury, often culminating in lethal sepsis and gastrointestinal bleeding. This deleterious impact can be attributed to radiation-induced DNA damage and oxidative stress, leading to various forms of cell death, such as apoptosis, necrosis and ferroptosis. Damage-associated molecular patterns (DAMPs) are intrinsic molecules released by cells undergoing injury or in the process of dying, either through passive or active pathways. These molecules then interact with pattern recognition receptors, triggering inflammatory responses. Such a cascade of events ultimately results in further tissue and organ damage, contributing to the elevated mortality rate. Notably, infection and sepsis often develop in ARS cases, further increasing the release of DAMPs. Given that lethal sepsis stands as a major contributor to the mortality in ARS, DAMPs hold the potential to function as mediators, exacerbating radiation-induced organ injury and consequently worsening overall survival. This review describes the intricate mechanisms underlying radiation-induced release of DAMPs. Furthermore, it discusses the detrimental effects of DAMPs on the immune system and explores potential DAMP-targeting therapeutic strategies to alleviate radiation-induced injury.
Collapse
Affiliation(s)
- Satoshi Yamaga
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Monowar Aziz
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
- Departments of Surgery and Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
| | - Atsushi Murao
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Max Brenner
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
- Departments of Surgery and Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
| | - Ping Wang
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
- Departments of Surgery and Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
| |
Collapse
|
13
|
Zhou M, Zhang H, Xu X, Chen H, Qi B. Association between circulating cell-free mitochondrial DNA and inflammation factors in noninfectious diseases: A systematic review. PLoS One 2024; 19:e0289338. [PMID: 38241222 PMCID: PMC10798522 DOI: 10.1371/journal.pone.0289338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 07/18/2023] [Indexed: 01/21/2024] Open
Abstract
OBJECTIVE This study aimed to assess the correlation between the circulating cell-free mitochondria DNA and inflammation factors in noninfectious disease by meta-analysis of data from eligible studies. MATERIALS AND METHODS Through a comprehensive searching of pubmed, embase, web of science, cochrane from establishment of the database to October 31, 2022, studies were selected that investigated the association of circulating cell free mitochondria DNA with inflammatory factors in non-infectious diseases. Studies that met the inclusion criteria and were published in English or Chinese were included. Data of each correlation coefficients were extracted from the paper and 95% confidence intervals were calculated. Sensitivity and heterogeneity tests were carried out for each data. RESULTS A total of 660 articles were retrieved and 22 were included in this meta-analysis, including 2600 patients. A fixed effects model was employed to examine ISS and IL-8, others were analyzed using random effects models. The correlation coefficient between mtDNA and ISS score was 0.37 (95%CI = [0.232;0.494]), p<0.0001, heterogeneity I2 = 46%, p = 0.11). The correlation coefficients between mtDNA and inflammatory factors are as follows: TNFα, 0.405 [(95%CI = [0.253;0.538], p<0.0001, heterogeneity I2 = 77%, p = 0.0001]. IL-6, 0.469 [(95%CI = [0.296;0.612]), p = 0.0001, heterogeneity I2 = 93%, p<0.0001]. CRP, 0.333[(95%CI = [0.149;0.494]), p = 0.005, heterogeneity I2 = 85%, p<0.0001]. IL-8, 0.343[(95%CI = [0.233;0.524]), p = 0.001, heterogeneity I2 = 50%, p = 0.09]. PCT, 0.333 [(95%CI = [0.06;0.64]), p = 0.09,heterogeneity I2 = 64%,p = 0.06]. There were no significant publication bias for TNFα, IL-6 and CRP. CONSLUSION Circulating cell free mtDNA was moderate positively correlated with the expression of inflammatory factors and the degree of trauma.
Collapse
Affiliation(s)
- Min Zhou
- Department of Orthopeadics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Hao Zhang
- Department of Orthopeadics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xin Xu
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Hairen Chen
- Department of Orthopeadics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Baiwen Qi
- Department of Orthopeadics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
14
|
Frelich M, Vodička V, Jor O, Burša F, Sklienka P. Predicting Acute Kidney Injury in Trauma Patients: Biomarkers as Early Indicators. Med Sci Monit 2024; 30:e942271. [PMID: 38204152 PMCID: PMC10795410 DOI: 10.12659/msm.942271] [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: 08/23/2023] [Accepted: 10/20/2023] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND Acute kidney injury (AKI) is a common cause of organ failure in patients after major trauma and is associated with increased morbidity and mortality. Early identification of patients at risk enables the implementation of a bundle of supportive care, which reduces the incidence of AKI. The primary objective of our study was to investigate whether the levels of biomarkers on admission predicted the onset of early AKI in patients with serious injuries. MATERIAL AND METHODS This prospective observational study included 98 adult patients of both sexes with a serious injury (injury severity score >16). At admission, blood samples were taken, and creatinine, neutrophil gelatinase-associated lipocalin (NGAL), high mobility group box 1 (HMGB-1), and markers of rhabdomyolysis (creatine kinase, myoglobin) were evaluated. The patients were provided with standard resuscitation care, and the occurrence of AKI was monitored during the first 7 days after admission to the Intensive Care Unit, according to the Kidney Disease Improving Global Outcomes diagnostic criteria. RESULTS AKI occurred in 25 (25.5%) patients, in whom the admission levels of HMGB-1, NGAL, creatinine, and myoglobin were significantly higher than in non-AKI patients (48.3±98.4 vs 113.0±209.4 µg/L, P=0.006; 150.2±349.9 vs 181.4±152.2 µg/L, P=0.004; 83.1±20.8 vs 118.8±32.2 µmol/L, P<0.005; 2734.4±2214.5 vs 4182.3±2477.1 µg/L, P=0.008, respectively). Creatine kinase was 14.5±9.2 µkat/L in non-AKI patients and 13.7±7.9 µkat/L in AKI patients (P=0.916). CONCLUSIONS Admission levels of HMGB-1, NGAL, creatinine, and myoglobin predicted the risk of AKI in severely injured patients.
Collapse
Affiliation(s)
- Michal Frelich
- Department of Anaesthesiology and Intensive Care Medicine, University Hospital of Ostrava, Ostrava, Czech Republic
- Department of Intensive Medicine, Emergency Medicine and Forensic Studies, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Vojtěch Vodička
- Department of Anaesthesiology and Intensive Care Medicine, University Hospital of Ostrava, Ostrava, Czech Republic
| | - Ondřej Jor
- Department of Anaesthesiology and Intensive Care Medicine, University Hospital of Ostrava, Ostrava, Czech Republic
- Department of Intensive Medicine, Emergency Medicine and Forensic Studies, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Filip Burša
- Department of Anaesthesiology and Intensive Care Medicine, University Hospital of Ostrava, Ostrava, Czech Republic
- Department of Intensive Medicine, Emergency Medicine and Forensic Studies, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Peter Sklienka
- Department of Anaesthesiology and Intensive Care Medicine, University Hospital of Ostrava, Ostrava, Czech Republic
- Department of Intensive Medicine, Emergency Medicine and Forensic Studies, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| |
Collapse
|
15
|
Osuka A, Shigeno A, Matsuura H, Onishi S, Yoneda K. Systemic immune response of burns from the acute to chronic phase. Acute Med Surg 2024; 11:e976. [PMID: 38894736 PMCID: PMC11184575 DOI: 10.1002/ams2.976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 05/07/2024] [Accepted: 06/01/2024] [Indexed: 06/21/2024] Open
Abstract
Immune responses that occur following burn injury comprise a series of reactions that are activated in response to damaged autologous tissues, followed by removal of damaged tissues and foreign pathogens such as invading bacteria, and tissue repair. These immune responses are considered to be programmed in living organisms. Developments of modern medicine have led to the saving of burned patients who could not be cured previously; however, the programmed response is no longer able to keep up, and various problems have arisen. This paper describes the mechanism of immune response specific to burn injury and the emerging concept of persistent inflammation, immunosuppression, and catabolism syndrome.
Collapse
Affiliation(s)
- Akinori Osuka
- Department of Trauma, Critical Care Medicine and Burn CenterJapan Community Health Care Organization Chukyo HospitalNagoyaJapan
- Department of Traumatology and Acute Critical MedicineOsaka University Graduate School of MedicineOsakaJapan
| | - Ayami Shigeno
- Department of Trauma, Critical Care Medicine and Burn CenterJapan Community Health Care Organization Chukyo HospitalNagoyaJapan
| | - Hiroshi Matsuura
- Department of Trauma, Critical Care Medicine and Burn CenterJapan Community Health Care Organization Chukyo HospitalNagoyaJapan
- Osaka Prefectural Nakakawachi Emergency and Critical Care CenterOsakaJapan
| | - Shinya Onishi
- Department of Trauma, Critical Care Medicine and Burn CenterJapan Community Health Care Organization Chukyo HospitalNagoyaJapan
- Department of Traumatology and Acute Critical MedicineOsaka University Graduate School of MedicineOsakaJapan
| | - Kazuhiro Yoneda
- Department of Trauma, Critical Care Medicine and Burn CenterJapan Community Health Care Organization Chukyo HospitalNagoyaJapan
- Department of Traumatology and Acute Critical MedicineOsaka University Graduate School of MedicineOsakaJapan
| |
Collapse
|
16
|
Xiao Y, Fang H, Zhu Y, Zhou J, Dai Z, Wang H, Xia Z, Tu Z, Leong KW. Multifunctional Cationic Hyperbranched Polyaminoglycosides that Target Multiple Mediators for Severe Abdominal Trauma Management. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305273. [PMID: 37997512 PMCID: PMC10767409 DOI: 10.1002/advs.202305273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/12/2023] [Indexed: 11/25/2023]
Abstract
Trauma and its associated complications, including dysregulated inflammatory responses, severe infection, and disseminated intravascular coagulation (DIC), continue to pose lethal threats worldwide. Following injury, cell-free nucleic acids (cfNAs), categorized as damage-associated molecular patterns (DAMPs), are released from dying or dead cells, triggering local and systemic inflammatory responses and coagulation abnormalities that worsen disease progression. Harnessing cfNA scavenging strategies with biomaterials has emerged as a promising approach for treating posttrauma systemic inflammation. In this study, the effectiveness of cationic hyperbranched polyaminoglycosides derived from tobramycin (HPT) and disulfide-included HPT (ss-HPT) in scavenging cfNAs to mitigate posttrauma inflammation and hypercoagulation is investigated. Both cationic polymers demonstrate the ability to suppress DAMP-induced toll-like receptor (TLR) activation, inflammatory cytokine secretion, and hypercoagulation by efficiently scavenging cfNAs. Additionally, HPT and ss-HPT exhibit potent antibacterial efficacy attributed to the presence of tobramycin in their chemical composition. Furthermore, HPT and ss-HPT exhibit favorable modulatory effects on inflammation and therapeutic outcomes in a cecal ligation puncture (CLP) mouse abdominal trauma model. Notably, in vivo studies reveal that ss-HPT displayed high accumulation and retention in injured organs of traumatized mice while maintaining a higher biodegradation rate in healthy mice, contrasting with findings for HPT. Thus, functionalized ss-HPT, a bioreducible polyaminoglycoside, holds promise as an effective option to enhance therapeutic outcomes for trauma patients by alleviating posttrauma inflammation and coagulation complications.
Collapse
Affiliation(s)
- Yongqiang Xiao
- Department of Burn Surgerythe First Affiliated HospitalNaval Medical UniversityShanghai200433P. R. China
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10027USA
- ENT InstituteDepartment of Facial Plastic and Reconstructive SurgeryEye & ENT HospitalFudan UniversityShanghai200031P. R. China
| | - He Fang
- Department of Burn Surgerythe First Affiliated HospitalNaval Medical UniversityShanghai200433P. R. China
| | - Yuefei Zhu
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10027USA
| | - Jie Zhou
- Department of Breast SurgeryAffiliated Cancer Hospital and InstituteGuangzhou Medical UniversityGuangzhou510095P. R. China
| | - Zhanzhan Dai
- Department of Burn Surgerythe First Affiliated HospitalNaval Medical UniversityShanghai200433P. R. China
| | - Hongxia Wang
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10027USA
| | - Zhaofan Xia
- Department of Burn Surgerythe First Affiliated HospitalNaval Medical UniversityShanghai200433P. R. China
| | - Zhaoxu Tu
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10027USA
- The Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510655P. R. China
| | - Kam W. Leong
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10027USA
- Department of Systems BiologyColumbia University Medical CenterNew YorkNY10032USA
| |
Collapse
|
17
|
Maisat W, Hou L, Sandhu S, Sin YC, Kim S, Pelt HV, Chen Y, Emani S, Kong SW, Emani S, Ibla J, Yuki K. Neutrophil extracellular traps formation is associated with postoperative complications in neonates and infants undergoing congenital cardiac surgery. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.21.572768. [PMID: 38187754 PMCID: PMC10769315 DOI: 10.1101/2023.12.21.572768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Pediatric patients with congenital heart diseases (CHD) often undergo surgical repair on cardiopulmonary bypass (CPB). Despite a significant medical and surgical improvement, the mortality of neonates and infants remains high. Damage-associated molecular patterns (DAMPs) are endogenous molecules released from injured/damaged tissues as danger signals. We examined 101 pediatric patients who underwent congenital cardiac surgery on CPB. The mortality rate was 4.0%, and the complication rate was 31.6%. We found that neonates/infants experienced multiple complications most, consistent with the previous knowledge. Neonates and infants in the complication group had received more transfusion intraoperatively than the non-complication arm with lower maximum amplitude (MA) on rewarming CPB thromboelastography (TEG). Despite TEG profiles were comparable at ICU admission between the two groups, the complication arm had higher postoperative chest tube output, requiring more blood transfusion. The complication group showed greater neutrophil extracellular traps (NETs) formation at the end of CPB and postoperatively. Plasma histones and high mobility group box 1 (HMGB1) levels were significantly higher in the complication arm. Both induced NETs in vitro and in vivo . As histones and HMGB1 target Toll-like receptor (TLR)2 and TLR4, their mRNA expression in neutrophils was upregulated in the complication arm. Taken together, NETs play a major role in postoperative complication in pediatric cardiac surgery and would be considered a target for intervention. Key points Neonates and infants showed highest postoperative complications with more upregulation of inflammatory transcriptomes of neutrophils.Neonates and infants with organ dysfunction had more NETs formation with higher plasma histones and HMGB1 levels.
Collapse
|
18
|
Jones MA, Hanison J, Apreutesei R, Allarakia B, Namvar S, Ramaswamy DS, Horner D, Smyth L, Body R, Columb M, Nirmalan M, Nirmalan N. Plasma interleukin responses as predictors of outcome stratification in patients after major trauma: a prospective observational two centre study. Front Immunol 2023; 14:1276171. [PMID: 38077362 PMCID: PMC10702136 DOI: 10.3389/fimmu.2023.1276171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 11/08/2023] [Indexed: 12/18/2023] Open
Abstract
Background and objectives There is a need to develop objective risk stratification tools to define efficient care pathways for trauma patients. Biomarker-based point of care testing may strengthen existing clinical tools currently available for this purpose. The dysregulation of pro- and anti-inflammatory cytokines in the pathogenesis of organ failure is well recognised. This study was carried out to evaluate whether blood concentrations of IL-6, IL-10, and IL-6:IL-10 ratios in the early stages of the illness are significantly different in patients with worsening organ function. Materials and methods In this prospective observational cohort study, plasma concentrations of IL-6 and IL-10 on days 1, 3 and 5 were measured in 91 major trauma patients using a multiplexed cytometric bead array approach. A composite measure of adverse outcome - defined as SOFA ≥ 2 or mortality at 7 days, was the primary outcome. IL-6 and IL-10 concentrations in early samples (days 1, 3 & 5) in patients who developed SOFA ≥ 2 on day 7 were compared against those who did not. Similar composite outcome groups at day 5 and in groups with worsening or improving SOFA scores (ΔSOFA) at days 7 and 5 were undertaken as secondary analyses. Results Stratification on day 7, 44 (48%) patients showed adverse outcomes. These adverse outcomes associated with significantly greater IL-6 concentrations on days 1 and 5 (Day 1: 47.65 [23.24-78.68] Vs 73.69 [39.93 - 118.07] pg/mL, P = 0.040 and Day 5: 12.85 [5.80-19.51] Vs 28.90 [8.78-74.08] pg/mL; P = 0.0019). Similarly, IL-10 levels were significantly greater in the adverse outcome group on days 3 and 5 (Day 3: 2.54 [1.76-3.19] Vs 3.16 [2.68-4.21] pg/mL; P = 0.044 and Day 5: 2.03 [1.65-2.55] Vs 2.90 [2.00-5.06] pg/mL; P <0.001). IL-6 and IL-10 concentrations were also significantly elevated in the adverse outcome groups at day 3 and day 5 when stratified on day 5 outcomes. Both IL-6 and IL-6:IL-10 were found to be significantly elevated on days 1 and 3 when stratified based on ΔSOFA at day 5. This significance was lost when stratified on day 7 scores. Conclusions Early IL-6 and IL-10 concentrations are significantly greater in patients who develop worsening organ functions downstream. These differences may provide an alternate biomarker-based approach to strengthen risk stratification in trauma patients.
Collapse
Affiliation(s)
- Matthew Allan Jones
- Biomedical Research and Innovation Centre, School of Science, Engineering and Environment, University of Salford, Manchester, United Kingdom
| | - James Hanison
- Critical Care Unit, Manchester University National Health Service (NHS) Foundation Trust (MFT), Manchester, United Kingdom
| | - Renata Apreutesei
- Biomedical Research and Innovation Centre, School of Science, Engineering and Environment, University of Salford, Manchester, United Kingdom
| | - Basmah Allarakia
- Biomedical Research and Innovation Centre, School of Science, Engineering and Environment, University of Salford, Manchester, United Kingdom
| | - Sara Namvar
- Biomedical Research and Innovation Centre, School of Science, Engineering and Environment, University of Salford, Manchester, United Kingdom
- Faculty of Biology, Medicine and Health, School of Medical Sciences, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Deepa Shruthi Ramaswamy
- Biomedical Research and Innovation Centre, School of Science, Engineering and Environment, University of Salford, Manchester, United Kingdom
| | - Daniel Horner
- Critical Care Unit, Salford Royal Foundation Trust (SRFT), Salford, United Kingdom
- Division of Infection, Immunity and Respiratory Medicine, University of Manchester, Manchester, United Kingdom
| | - Lucy Smyth
- Biomedical Research and Innovation Centre, School of Science, Engineering and Environment, University of Salford, Manchester, United Kingdom
| | - Richard Body
- Faculty of Biology, Medicine and Health, School of Medical Sciences, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Malachy Columb
- Critical Care Unit, Manchester University National Health Service (NHS) Foundation Trust (MFT), Manchester, United Kingdom
| | - Mahesan Nirmalan
- Faculty of Biology, Medicine and Health, School of Medical Sciences, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Niroshini Nirmalan
- Biomedical Research and Innovation Centre, School of Science, Engineering and Environment, University of Salford, Manchester, United Kingdom
| |
Collapse
|
19
|
Wang YJ, Zheng Y, Cong L, Wang P, Zhao L, Xing L, Liu J, Xu H, Li N, Zhao Y, Shi Q, Liang Q. Lymphatic platelet thrombosis limits bone repair by precluding lymphatic transporting DAMPs. RESEARCH SQUARE 2023:rs.3.rs-3474507. [PMID: 38014223 PMCID: PMC10680927 DOI: 10.21203/rs.3.rs-3474507/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Lymphatic vessels (LVs) interdigitated with blood vessels, travel and form an extensive transport network in the musculoskeletal system. Blood vessels in bone regulate osteogenesis and hematopoiesis, however, whether LVs in bone affect fracture healing is unclear. Here, by near infrared indocyanine green lymphatic imaging (NIR-ICG), we examined lymphatic draining function at the tibial fracture sites and found lymphatic drainage insufficiency (LDI) occurred as early as two weeks after fracture. Sufficient lymphatic drainage facilitates fracture healing. In addition, we identified that lymphatic platelet thrombosis (LPT) blocks the draining lymphoid sinus and LVs, caused LDI and then inhibited fracture healing, which can be rescued by a pharmacological approach. Moreover, unblocked lymphatic drainage decreased neutrophils and increased M2-like macrophages of hematoma niche to support osteoblast (OB) survival and bone marrow-derived mesenchymal stem cell (BMSC) proliferation via transporting damage-associated molecular patterns (DAMPs). These findings demonstrate that LPT limits bone regeneration by blocking lymphatic drainage from transporting DAMPs. Together, these findings represent a novel way forward in the treatment of bone repair.
Collapse
|
20
|
Bijkerk V, Visser J, Jacobs LMC, Keijzer C, Warlé MC. Deep versus moderate neuromuscular blockade during total hip arthroplasty to improve postoperative quality of recovery and immune function: protocol for a randomised controlled study. BMJ Open 2023; 13:e073537. [PMID: 37640469 PMCID: PMC10462972 DOI: 10.1136/bmjopen-2023-073537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 08/16/2023] [Indexed: 08/31/2023] Open
Abstract
INTRODUCTION There is accumulating evidence that deep neuromuscular blockade (NMB) improves intraoperative surgical conditions during laparoscopic surgery. Studies investigating the effects of deep NMB in open surgery are scarce. In theory, by limiting surgical damage through deeper muscle relaxation, postoperative inflammation and concomitant immune suppression can be reduced. Therefore, this study will investigate the effects of deep NMB during total hip arthroplasty, which demands a relatively large exposure of the hip joint through and in between muscles. METHODS AND ANALYSIS This study is a monocentre blinded randomised controlled trial in 100 patients undergoing total hip arthroplasty under general anaesthesia. Patients will be randomised in a 1:1 fashion to an intervention group of intraoperative deep NMB (a post-tetanic count of 1-2) or a control group receiving moderate NMB (a train-of-four count of 1-2). NMB will be achieved by continuous or bolus administration of rocuronium, respectively. The primary endpoint is the quality of recovery at postoperative day 1 measured by the Quality of Recovery-40 Questionnaire, analysed by Analysis of Variance. The secondary endpoint is postoperative innate immune function, measured by ex vivo production capacity of tumour necrosis factor and interleukin-1β on endotoxin stimulation of whole blood. ETHICS AND DISSEMINATION Ethical approval for this study was granted by the Medical Ethics Committee 'METC Oost-Nederland' (reference number 2022-15754). Informed consent will be obtained prior to study participation. Study results will be published in an international peer-reviewed journal. TRIAL REGISTRATION NUMBERS ClinicalTrials.gov Registry (NCT05562999) and EudraCT Registry (2022-002451-19).
Collapse
Affiliation(s)
- Veerle Bijkerk
- Department of Surgery, Radboudumc, Nijmegen, The Netherlands
- Department of Anesthesiology, Radboudumc, Nijmegen, The Netherlands
| | - Jetze Visser
- Department of Orthopedics, Radboudumc, Nijmegen, The Netherlands
| | | | | | - Michiel C Warlé
- Department of Surgery, Radboudumc, Nijmegen, The Netherlands
| |
Collapse
|
21
|
Horner E, Lord JM, Hazeldine J. The immune suppressive properties of damage associated molecular patterns in the setting of sterile traumatic injury. Front Immunol 2023; 14:1239683. [PMID: 37662933 PMCID: PMC10469493 DOI: 10.3389/fimmu.2023.1239683] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 07/31/2023] [Indexed: 09/05/2023] Open
Abstract
Associated with the development of hospital-acquired infections, major traumatic injury results in an immediate and persistent state of systemic immunosuppression, yet the underlying mechanisms are poorly understood. Detected in the circulation in the minutes, days and weeks following injury, damage associated molecular patterns (DAMPs) are a heterogeneous collection of proteins, lipids and DNA renowned for initiating the systemic inflammatory response syndrome. Suggesting additional immunomodulatory roles in the post-trauma immune response, data are emerging implicating DAMPs as potential mediators of post-trauma immune suppression. Discussing the results of in vitro, in vivo and ex vivo studies, the purpose of this review is to summarise the emerging immune tolerising properties of cytosolic, nuclear and mitochondrial-derived DAMPs. Direct inhibition of neutrophil antimicrobial activities, the induction of endotoxin tolerance in monocytes and macrophages, and the recruitment, activation and expansion of myeloid derived suppressor cells and regulatory T cells are examples of some of the immune suppressive properties assigned to DAMPs so far. Crucially, with studies identifying the molecular mechanisms by which DAMPs promote immune suppression, therapeutic strategies that prevent and/or reverse DAMP-induced immunosuppression have been proposed. Approaches currently under consideration include the use of synthetic polymers, or the delivery of plasma proteins, to scavenge circulating DAMPs, or to treat critically-injured patients with antagonists of DAMP receptors. However, as DAMPs share signalling pathways with pathogen associated molecular patterns, and pro-inflammatory responses are essential for tissue regeneration, these approaches need to be carefully considered in order to ensure that modulating DAMP levels and/or their interaction with immune cells does not negatively impact upon anti-microbial defence and the physiological responses of tissue repair and wound healing.
Collapse
Affiliation(s)
- Emily Horner
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Janet M. Lord
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
- National Institute for Health Research Surgical Reconstruction and Microbiology Research Centre, Queen Elizabeth Hospital Birmingham, Birmingham, United Kingdom
| | - Jon Hazeldine
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
- National Institute for Health Research Surgical Reconstruction and Microbiology Research Centre, Queen Elizabeth Hospital Birmingham, Birmingham, United Kingdom
| |
Collapse
|
22
|
Ye J, Hu X, Wang Z, Li R, Gan L, Zhang M, Wang T. The role of mtDAMPs in the trauma-induced systemic inflammatory response syndrome. Front Immunol 2023; 14:1164187. [PMID: 37533869 PMCID: PMC10391641 DOI: 10.3389/fimmu.2023.1164187] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 06/26/2023] [Indexed: 08/04/2023] Open
Abstract
Systemic inflammatory response syndrome (SIRS) is a non-specific exaggerated defense response caused by infectious or non-infectious stressors such as trauma, burn, surgery, ischemia and reperfusion, and malignancy, which can eventually lead to an uncontrolled inflammatory response. In addition to the early mortality due to the "first hits" after trauma, the trauma-induced SIRS and multiple organ dysfunction syndrome (MODS) are the main reasons for the poor prognosis of trauma patients as "second hits". Unlike infection-induced SIRS caused by pathogen-associated molecular patterns (PAMPs), trauma-induced SIRS is mainly mediated by damage-associated molecular patterns (DAMPs) including mitochondrial DAMPs (mtDAMPs). MtDAMPs released after trauma-induced mitochondrial injury, including mitochondrial DNA (mtDNA) and mitochondrial formyl peptides (mtFPs), can activate inflammatory response through multiple inflammatory signaling pathways. This review summarizes the role and mechanism of mtDAMPs in the occurrence and development of trauma-induced SIRS.
Collapse
Affiliation(s)
- Jingjing Ye
- Trauma Center, Peking University People’s Hospital, Key Laboratory of Trauma Treatment and Neural Regeneration (Peking University) Ministry of Education, National Center for Trauma Medicine of China, Beijing, China
| | - Xiaodan Hu
- Trauma Center, Peking University People’s Hospital, Key Laboratory of Trauma Treatment and Neural Regeneration (Peking University) Ministry of Education, National Center for Trauma Medicine of China, Beijing, China
- School of Basic Medicine, Peking University, Beijing, China
| | - Zhiwei Wang
- Orthopedics Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Rui Li
- Trauma Center, Peking University People’s Hospital, Key Laboratory of Trauma Treatment and Neural Regeneration (Peking University) Ministry of Education, National Center for Trauma Medicine of China, Beijing, China
| | - Lebin Gan
- Trauma Center, Peking University People’s Hospital, Key Laboratory of Trauma Treatment and Neural Regeneration (Peking University) Ministry of Education, National Center for Trauma Medicine of China, Beijing, China
| | - Mengwei Zhang
- Trauma Center, Peking University People’s Hospital, Key Laboratory of Trauma Treatment and Neural Regeneration (Peking University) Ministry of Education, National Center for Trauma Medicine of China, Beijing, China
| | - Tianbing Wang
- Trauma Center, Peking University People’s Hospital, Key Laboratory of Trauma Treatment and Neural Regeneration (Peking University) Ministry of Education, National Center for Trauma Medicine of China, Beijing, China
| |
Collapse
|
23
|
Jeffrey M, Denny KJ, Lipman J, Conway Morris A. Differentiating infection, colonisation, and sterile inflammation in critical illness: the emerging role of host-response profiling. Intensive Care Med 2023; 49:760-771. [PMID: 37344680 DOI: 10.1007/s00134-023-07108-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 05/22/2023] [Indexed: 06/23/2023]
Abstract
Infection results when a pathogen produces host tissue damage and elicits an immune response. Critically ill patients experience immune activation secondary to both sterile and infectious insults, with overlapping clinical phenotypes and underlying immunological mechanisms. Patients also undergo a shift in microbiota with the emergence of pathogen-dominant microbiomes. Whilst the combination of inflammation and microbial shift has long challenged intensivists in the identification of true infection, the advent of highly sensitive molecular diagnostics has further confounded the diagnostic dilemma as the number of microbial detections increases. Given the key role of the host immune response in the development and definition of infection, profiling the host response offers the potential to help unravel the conundrum of distinguishing colonisation and sterile inflammation from true infection. This narrative review provides an overview of current approaches to distinguishing colonisation from infection using routinely available techniques and proposes matrices to support decision-making in this setting. In searching for new tools to better discriminate these states, the review turns to the understanding of the underlying pathobiology of the host response to infection. It then reviews the techniques available to assess this response in a clinically applicable context. It will cover techniques including profiling of transcriptome, protein expression, and immune functional assays, detailing the current state of knowledge in diagnostics along with the challenges and opportunities. The ultimate infection diagnostic tool will likely combine an assessment of both host immune response and sensitive pathogen detection to improve patient management and facilitate antimicrobial stewardship.
Collapse
Affiliation(s)
- Mark Jeffrey
- John V Farman Intensive Care Unit, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Division of Anaesthesia, Department of Medicine, Level 4, Addenbrooke's Hospital, University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK
| | - Kerina J Denny
- Department of Intensive Care, Gold Coast University Hospital, Southport, QLD, Australia
- School of Medicine, University of Queensland, Herston, Brisbane, Australia
| | - Jeffrey Lipman
- University of Queensland Centre for Clinical Research, The University of Queensland, Brisbane, Australia
- Jamieson Trauma Institute and Intensive Care Services, Royal Brisbane and Women's Hospital, Brisbane, Australia
- Nimes University Hospital, University of Montpellier, Nimes, France
| | - Andrew Conway Morris
- John V Farman Intensive Care Unit, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
- Division of Anaesthesia, Department of Medicine, Level 4, Addenbrooke's Hospital, University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK.
- Division of Immunology, Department of Pathology, University of Cambridge, Cambridge, UK.
| |
Collapse
|
24
|
Wei YH, Bi RT, Qiu YM, Zhang CL, Li JZ, Li YN, Hu B. The gastrointestinal-brain-microbiota axis: a promising therapeutic target for ischemic stroke. Front Immunol 2023; 14:1141387. [PMID: 37342335 PMCID: PMC10277866 DOI: 10.3389/fimmu.2023.1141387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 05/22/2023] [Indexed: 06/22/2023] Open
Abstract
Ischemic stroke is a highly complex systemic disease characterized by intricate interactions between the brain and gastrointestinal tract. While our current understanding of these interactions primarily stems from experimental models, their relevance to human stroke outcomes is of considerable interest. After stroke, bidirectional communication between the brain and gastrointestinal tract initiates changes in the gastrointestinal microenvironment. These changes involve the activation of gastrointestinal immunity, disruption of the gastrointestinal barrier, and alterations in gastrointestinal microbiota. Importantly, experimental evidence suggests that these alterations facilitate the migration of gastrointestinal immune cells and cytokines across the damaged blood-brain barrier, ultimately infiltrating the ischemic brain. Although the characterization of these phenomena in humans is still limited, recognizing the significance of the brain-gastrointestinal crosstalk after stroke offers potential avenues for therapeutic intervention. By targeting the mutually reinforcing processes between the brain and gastrointestinal tract, it may be possible to improve the prognosis of ischemic stroke. Further investigation is warranted to elucidate the clinical relevance and translational potential of these findings.
Collapse
Affiliation(s)
| | | | | | | | | | - Ya-nan Li
- *Correspondence: Ya-nan Li, ; Bo Hu,
| | - Bo Hu
- *Correspondence: Ya-nan Li, ; Bo Hu,
| |
Collapse
|
25
|
Wang Z, Chen G. Immune regulation in neurovascular units after traumatic brain injury. Neurobiol Dis 2023; 179:106060. [PMID: 36871640 DOI: 10.1016/j.nbd.2023.106060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 02/19/2023] [Accepted: 02/28/2023] [Indexed: 03/07/2023] Open
Abstract
Traumatic brain injury (TBI) is a major cause of death and disability worldwide. Survivors may experience movement disorders, memory loss, and cognitive deficits. However, there is a lack of understanding of the pathophysiology of TBI-mediated neuroinflammation and neurodegeneration. The immune regulation process of TBI involves changes in the peripheral and central nervous system (CNS) immunity, and intracranial blood vessels are essential communication centers. The neurovascular unit (NVU) is responsible for coupling blood flow with brain activity, and comprises endothelial cells, pericytes, astrocyte end-feet, and vast regulatory nerve terminals. A stable NVU is the basis for normal brain function. The concept of the NVU emphasizes that cell-cell interactions between different types of cells are essential for maintaining brain homeostasis. Previous studies have explored the effects of immune system changes after TBI. The NVU can help us further understand the immune regulation process. Herein, we enumerate the paradoxes of primary immune activation and chronic immunosuppression. We describe the changes in immune cells, cytokines/chemokines, and neuroinflammation after TBI. The post-immunomodulatory changes in NVU components are discussed, and research exploring immune changes in the NVU pattern is also described. Finally, we summarize immune regulation therapies and drugs after TBI. Therapies and drugs that focus on immune regulation have shown great potential for neuroprotection. These findings will help us further understand the pathological processes after TBI.
Collapse
Affiliation(s)
- Zongqi Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province 215006, China; Institute of Stroke Research, Soochow University, Suzhou, Jiangsu Province 215006, China
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province 215006, China; Institute of Stroke Research, Soochow University, Suzhou, Jiangsu Province 215006, China.
| |
Collapse
|
26
|
Krämer TJ, Pickart F, Pöttker B, Gölz C, Neulen A, Pantel T, Goetz H, Ritter K, Schäfer MKE, Thal SC. Early DNase-I therapy delays secondary brain damage after traumatic brain injury in adult mice. Sci Rep 2023; 13:4348. [PMID: 36928073 PMCID: PMC10018640 DOI: 10.1038/s41598-023-30421-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 02/22/2023] [Indexed: 03/18/2023] Open
Abstract
Traumatic brain injury (TBI) causes the release of danger-associated molecular patterns (DAMP) from damaged or dead cells, which contribute to secondary brain damage after TBI. Cell-free DNA (cfDNA) is a DAMP known to cause disruption of the blood-brain barrier (BBB), promote procoagulant processes, brain edema, and neuroinflammation. This study tested the hypothesis that administration of deoxyribonuclease-I (DNase-I) has a beneficial effect after TBI. Mice (n = 84) were subjected to controlled cortical impact (CCI) and posttraumatic intraperitoneal injections of low dose (LD) or high dose (HD) of DNase-I or vehicle solution at 30 min and 12 h after CCI. LD was most effective to reduce lesion volume (p = 0.003), brain water content (p < 0.0001) and to stabilize BBB integrity (p = 0.019) 1 day post-injury (dpi). At 6 h post injury LD-treated animals showed less cleavage of fibrin (p = 0.0014), and enhanced perfusion as assessed by micro-computer-tomography (p = 0.027). At 5 dpi the number of Iba1-positive cells (p = 0.037) were reduced, but the number of CD45-positive cells, motoric function and brain lesion volume was not different. Posttraumatic-treatment with DNase-I therefore stabilizes the BBB, reduces the formation of brain edema, immune response, and delays secondary brain damage. DNase-I might be a new approach to extend the treatment window after TBI.
Collapse
Affiliation(s)
- Tobias J Krämer
- Department of Anesthesiology, University Medical Center of Johannes Gutenberg University, Langenbeckstrasse 1, 55131, Mainz, Germany.
- Faculty of Health, University Witten/Herdecke, Witten, Germany.
| | - Florian Pickart
- Department of Anesthesiology, University Medical Center of Johannes Gutenberg University, Langenbeckstrasse 1, 55131, Mainz, Germany
| | - Bruno Pöttker
- Department of Anesthesiology, University Medical Center of Johannes Gutenberg University, Langenbeckstrasse 1, 55131, Mainz, Germany
| | - Christina Gölz
- Department of Anesthesiology, University Medical Center of Johannes Gutenberg University, Langenbeckstrasse 1, 55131, Mainz, Germany
| | - Axel Neulen
- Department of Neurosurgery, University Medical Center of Johannes Gutenberg University, Langenbeckstrasse 1, 55131, Mainz, Germany
| | - Tobias Pantel
- Department of Neurosurgery, University Medical Center of Johannes Gutenberg University, Langenbeckstrasse 1, 55131, Mainz, Germany
| | - Hermann Goetz
- Cell Biology Unit, University Medical Center of Johannes Gutenberg University, Langenbeckstrasse 1, 55131, Mainz, Germany
| | - Katharina Ritter
- Department of Anesthesiology, University Medical Center of Johannes Gutenberg University, Langenbeckstrasse 1, 55131, Mainz, Germany
| | - Michael K E Schäfer
- Department of Anesthesiology, University Medical Center of Johannes Gutenberg University, Langenbeckstrasse 1, 55131, Mainz, Germany
- Focus Program Translational Neurosciences, University Medical Center of Johannes Gutenberg University, Langenbeckstrasse 1, 55131, Mainz, Germany
- Research Center for Immunotherapy, University Medical Center of Johannes Gutenberg University, Langenbeckstrasse 1, 55131, Mainz, Germany
- Center for Molecular Surgical Research, University Medical Center of Johannes Gutenberg University, Langenbeckstrasse 1, 55131, Mainz, Germany
| | - Serge C Thal
- Department of Anesthesiology, University Medical Center of Johannes Gutenberg University, Langenbeckstrasse 1, 55131, Mainz, Germany
- Focus Program Translational Neurosciences, University Medical Center of Johannes Gutenberg University, Langenbeckstrasse 1, 55131, Mainz, Germany
- Center for Molecular Surgical Research, University Medical Center of Johannes Gutenberg University, Langenbeckstrasse 1, 55131, Mainz, Germany
- Department of Anesthesiology, Helios University Hospital Wuppertal, University Witten/Herdecke, Heusnerstrasse 40, 42283, Wuppertal, Germany
| |
Collapse
|
27
|
Wang YR, Cui WQ, Wu HY, Xu XD, Xu XQ. The role of T cells in acute ischemic stroke. Brain Res Bull 2023; 196:20-33. [PMID: 36906042 DOI: 10.1016/j.brainresbull.2023.03.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 03/02/2023] [Accepted: 03/06/2023] [Indexed: 03/11/2023]
Abstract
Acute ischemic stroke (AIS) is associated with high rates of disability and mortality, exerting a substantial impact on overall survival and health-related quality of life. Treatment of AIS remains challenging given that the underlying pathologic mechanisms remain unclear. However, recent research has demonstrated that the immune system plays a key role in the development of AIS. Numerous studies have reported infiltration of T cells into ischemic brain tissue. While some types of T cells can promote the development of inflammatory responses and aggravate ischemic damage in patients with AIS, other T cells appear to exert neuroprotective effects via immunosuppression and other mechanisms. In this review, we discuss the recent findings regarding the infiltration of T cells into ischemic brain tissue, and the mechanisms governing how T cells can facilitate tissue injury or neuroprotection in AIS. Factors influencing the function of T cells, such as intestinal microflora and sex differences, are also discussed. We also explore the recent research on the effect of non-coding RNA on T cells after stroke, as well as the potential for specifically targeting T cells in the treatment of stroke patients.
Collapse
Affiliation(s)
- Yi-Ran Wang
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wen-Qiang Cui
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China; First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Hong-Yun Wu
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China; First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiang-Dong Xu
- Experimental Center, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiang-Qing Xu
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China; First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China.
| |
Collapse
|
28
|
Affiliation(s)
- Justine Devulder
- National Heart & Lung Institute, Imperial College London, London, UK
| |
Collapse
|
29
|
Brittney W, Kozar R, Wei C. EMERGING ROLE OF EXTRACELLULAR RNA IN INNATE IMMUNITY, SEPSIS, AND TRAUMA. Shock 2023; 59:190-199. [PMID: 36730864 PMCID: PMC9957828 DOI: 10.1097/shk.0000000000002032] [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] [Indexed: 02/04/2023]
Abstract
ABSTRACT Sepsis and trauma remain the leading causes of morbidity and mortality. Our understanding of the molecular pathogenesis in the development of multiple organ dysfunction in sepsis and trauma has evolved as more focus is on secondary injury from innate immunity, inflammation, and the potential role of endogenous danger molecules. Studies of the past several decades have generated evidence for extracellular RNAs (exRNAs) as biologically active mediators in health and disease. Here, we review studies on plasma exRNA profiling in mice and humans with sepsis and trauma, the role and mode of action by exRNAs, such as ex-micro(mi)RNAs, in host innate immune response, and their potential implications in various organ injury during sepsis and trauma.
Collapse
Affiliation(s)
- Williams Brittney
- Translational Research Program, Department of Anesthesiology, and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Rosemary Kozar
- Shock Trauma Center and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Chao Wei
- Translational Research Program, Department of Anesthesiology, and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
30
|
Gregorius J, Brenner T. [Pathophysiology of sepsis]. Anasthesiol Intensivmed Notfallmed Schmerzther 2023; 58:13-27. [PMID: 36623527 DOI: 10.1055/a-1813-2057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Up to now, sepsis is one of the most threatening diseases and its therapy remains challenging. Sepsis is currently defined as a severely dysregulated immune response to an infection resulting in organ dysfunction. The pathophysiology is mainly driven by exogenous PAMPs ("pathogen-associated molecular patterns") and endogenous DAMPs ("damage-associated molecular patterns"), which can activate PRRs ("pattern recognition receptors") on different cell types (mainly immune cells), leading to the initiation of manifold downstream pathways and a perpetuation of patients' immune response. Sepsis is neither an exclusive pro- nor an anti-inflammatory disease: both processes take place in parallel, resulting in an individual immunologic disease state depending on the severity of each component at different time points. Septic shock is a complex disorder of the macro- and microcirculation, provoking a severe lack of oxygenation further aggravating sepsis defining organ dysfunctions. An in-depth knowledge of the heterogeneity and the time-dependency of the septic immunopathology will be essential for the design of future sepsis trials and therapy planning in patients with sepsis. The big aim is to achieve a more individualized treatment strategy in patients suffering from sepsis or septic shock.
Collapse
|
31
|
Extracellular CIRP dysregulates macrophage bacterial phagocytosis in sepsis. Cell Mol Immunol 2023; 20:80-93. [PMID: 36471113 PMCID: PMC9794804 DOI: 10.1038/s41423-022-00961-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 11/12/2022] [Indexed: 12/12/2022] Open
Abstract
In sepsis, macrophage bacterial phagocytosis is impaired, but the mechanism is not well elucidated. Extracellular cold-inducible RNA-binding protein (eCIRP) is a damage-associated molecular pattern that causes inflammation. However, whether eCIRP regulates macrophage bacterial phagocytosis is unknown. Here, we reported that the bacterial loads in the blood and peritoneal fluid were decreased in CIRP-/- mice and anti-eCIRP Ab-treated mice after sepsis. Increased eCIRP levels were correlated with decreased bacterial clearance in septic mice. CIRP-/- mice showed a marked increase in survival after sepsis. Recombinant murine CIRP (rmCIRP) significantly decreased the phagocytosis of bacteria by macrophages in vivo and in vitro. rmCIRP decreased the protein expression of actin-binding proteins, ARP2, and p-cofilin in macrophages. rmCIRP significantly downregulated the protein expression of βPIX, a Rac1 activator. We further demonstrated that STAT3 and βPIX formed a complex following rmCIRP treatment, preventing βPIX from activating Rac1. We also found that eCIRP-induced STAT3 phosphorylation was required for eCIRP's action in actin remodeling. Inhibition of STAT3 phosphorylation prevented the formation of the STAT3-βPIX complex, restoring ARP2 and p-cofilin expression and membrane protrusion in rmCIRP-treated macrophages. The STAT3 inhibitor stattic rescued the macrophage phagocytic dysfunction induced by rmCIRP. Thus, we identified a novel mechanism of macrophage phagocytic dysfunction caused by eCIRP, which provides a new therapeutic target to ameliorate sepsis.
Collapse
|
32
|
Ding C, Hong S, Zhang M, Sun Y, Li N, Zhang J, Ma L, Tian L, Ren W, Zhang L, Yao S. Establishment and evaluation of an in vitro blast lung injury model using alveolar epithelial cells. Front Public Health 2022; 10:994670. [PMID: 36620304 PMCID: PMC9816474 DOI: 10.3389/fpubh.2022.994670] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022] Open
Abstract
Background Gas explosion is a fatal disaster commonly occurred in coal mining and often causes systematic physical injuries, of which blast lung injury is the primary one and has not yet been fully investigated due to the absence of disease models. To facilitate studies of this field, we constructed an in vitro blast lung injury model using alveolar epithelial cells. Methods We randomly divided the alveolar epithelial cells into the control group and blast wave group, cells in the blast wave group were stimulated with different strengths of blast wave, and cells in the control group received sham intervention. Based on the standards we set up for a successful blast injury model, the optimal modeling conditions were studied on different frequencies of blast wave, modeling volume, cell incubation duration, and cell density. The changes of cell viability, apoptosis, intracellular oxidative stress, and inflammation were measured. Results We found that cell viability decreased by approximately 50% at 6 h after exposing to 8 bar energy of blast wave, then increased with the extension of culture time and reached to (74.33 ± 9.44) % at 12 h. By applying 1000 ~ 2500 times of shock wave to 1 ~ 5 × 105 cells /ml, the changes of cell viability could well meet the modeling criteria. In parallel, the content of reactive oxide species (ROS), malonaldehyde (MDA), interleukin 18 (IL-18), tumor necrosis factor alpha (TNF-α), and transforming growth factor beta (TGF-β) increased in the blast wave group, while superoxide dismutase (SOD) and Glutathione -S- transferase (GST) decreased, which were highly consistent with that of human beings with gas explosion-induced pulmonary injury. Conclusion An in vitro blast lung injury model is set up using a blast wave physiotherapy under 8 bar, 10 Hz blast wave on (1 ~ 5) ×105 alveolar epithelial cells for 1 000 times. This model is flexible, safe, and stable, and can be used for studies of lung injury caused by gas explosion and blast-associated other external forces.
Collapse
Affiliation(s)
- Chunjie Ding
- School of Public Health, Xinxiang Medical University, Xinxiang, China
| | - Shan Hong
- School of Public Health, Xinxiang Medical University, Xinxiang, China
| | - Miao Zhang
- School of Public Health, Xinxiang Medical University, Xinxiang, China
| | - Yunzhe Sun
- School of Public Health, Xinxiang Medical University, Xinxiang, China
| | - Ning Li
- School of Public Health, North China University of Science and Technology, Tangshan, China
| | - Jing Zhang
- School of Public Health, North China University of Science and Technology, Tangshan, China
| | - Lan Ma
- School of Public Health, Weifang Medical University, Weifang, China
| | - Linqiang Tian
- Institute of Trauma and Orthopedics, Xinxiang Medical University, Xinxiang, China
| | - Wenjie Ren
- Institute of Trauma and Orthopedics, Xinxiang Medical University, Xinxiang, China
| | - Lin Zhang
- Clinical Medical Research Center for Women and Children Diseases, Maternal and Child Health Care Hospital of Shandong Province Affiliated to Qingdao University, Jinan, China,*Correspondence: Lin Zhang ✉
| | - Sanqiao Yao
- School of Public Health, Xinxiang Medical University, Xinxiang, China,Sanqiao Yao ✉
| |
Collapse
|
33
|
Bouras M, Asehnoune K, Roquilly A. Immune modulation after traumatic brain injury. Front Med (Lausanne) 2022; 9:995044. [PMID: 36530909 PMCID: PMC9751027 DOI: 10.3389/fmed.2022.995044] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 11/14/2022] [Indexed: 07/20/2023] Open
Abstract
Traumatic brain injury (TBI) induces instant activation of innate immunity in brain tissue, followed by a systematization of the inflammatory response. The subsequent response, evolved to limit an overwhelming systemic inflammatory response and to induce healing, involves the autonomic nervous system, hormonal systems, and the regulation of immune cells. This physiological response induces an immunosuppression and tolerance state that promotes to the occurrence of secondary infections. This review describes the immunological consequences of TBI and highlights potential novel therapeutic approaches using immune modulation to restore homeostasis between the nervous system and innate immunity.
Collapse
Affiliation(s)
- Marwan Bouras
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
- CHU Nantes, INSERM, Nantes Université, Anesthesie Reanimation, CIC 1413, Nantes, France
| | - Karim Asehnoune
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
- CHU Nantes, INSERM, Nantes Université, Anesthesie Reanimation, CIC 1413, Nantes, France
| | - Antoine Roquilly
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
- CHU Nantes, INSERM, Nantes Université, Anesthesie Reanimation, CIC 1413, Nantes, France
| |
Collapse
|
34
|
Calycosin Improves Intestinal Mucosal Barrier Function after Gastrectomy in Rats through Alleviating Bacterial Translocation, Inflammation, and Oxidative Stress. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:7412331. [PMID: 35795283 PMCID: PMC9251107 DOI: 10.1155/2022/7412331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/06/2022] [Accepted: 06/10/2022] [Indexed: 11/17/2022]
Abstract
Objective Calycosin is the main bioactive extract of Astragali Radix with anti-inflammation, antioxidant, and anticancer properties. Here, our study evaluated the protective effects and mechanisms of calycosin on intestinal mucosal barrier under gastrectomy. Methods After receiving gastrectomy, the rats were administrated with 20 mg/kg, 40 mg/kg, or 80 mg/kg calycosin. Endotoxin, bacterial translocation, and intestinal bacterial flora were assayed. Intestinal injury was detected via hematoxylin and eosin staining. Tight junction indicators (occludin, claudin, and ZO-1) and apoptotic proteins (Bax, Bcl-2, and cleaved caspase 3) were examined in intestinal tissues. Inflammatory indicators (IL-1β, IL-6, and TNF-α) were examined in serum or intestinal specimens via ELISA. Apoptosis was assessed via TUNEL staining. IgA + B cells in intestinal tissues and sIgA in intestinal lumen were examined through immunohistochemistry and ELISA, respectively. Oxidative stress indicators (TSH, SOD, CAT, GSH-Px, and MDA) were also detected via ELISA. Results Our results showed that calycosin administration decreased endotoxin levels in peripheral blood, intestine, and portal vein blood; lowered the bacterial translocation ratio; and regained the balance among intestinal bacterial flora (comprising bifidobacterium, lactic acid bacillus, enterobacter, enterococcus, aerobic bacteria, and anaerobic bacteria) in the rats with gastrectomy. After calycosin treatment, intestinal mucosal damage of the rats with gastrectomy was ameliorated, with the increase in expression of tight junction proteins. Additionally, calycosin reduced intestinal inflammation, apoptosis, secretion of sIgA, and oxidative stress in the rats with gastrectomy. Conclusion Altogether, our findings demonstrate that calycosin may improve intestinal mucosal barrier function under gastrectomy via reducing bacterial translocation, inflammation, and oxidative stress.
Collapse
|
35
|
Ljunghill Hedberg A, Pauksens K, Enblad P, Larsson A, Sjölin J. Relationship between T-cell-dependent and T-cell-independent vaccines after neurotrauma; is the B-cell response preserved? Hum Vaccin Immunother 2022; 18:2088971. [PMID: 35704795 PMCID: PMC9621013 DOI: 10.1080/21645515.2022.2088971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Background After trauma and central nervous system (CNS) injury, trauma-induced immune deficiency syndrome (TIDS) and CNS injury-induced immune deficiency syndrome (CIDS) may negatively affect responses to T-cell-dependent vaccines, such as pneumococcal conjugate vaccine (PCV) recommended after basilar fracture. This study (NCT02806284) aimed to investigate whether there after neurotrauma is a correlation between T-cell-dependent and independent vaccine responses and, thus, if B-cell activity is similarly depressed and whether the T-cell-dependent response is possible to predict. Methods Adult patients with basilar fracture (n = 33) and those undergoing pituitary gland surgery (n = 23) were within 10 days vaccinated with a T-cell-dependent vaccine against Haemophilus influenzae type b (Hib) and a T-cell-independent pneumococcal polysaccharide vaccine (PPSV). Samples reflecting the systemic inflammatory response and pre- and post-vaccination antibody levels after 3–6 weeks against Hib and PPSV were collected and determined by enzyme immunoassays. Results High and significant correlations were detected in the responses to different pneumococcal serotypes, but none between the Hib and PPSV responses. No differences in trauma scores, C-reactive protein, IL-6, IL-10, pentraxin 3, fractalkine or calprotectin plasma concentrations or in ex vivo TNF-α, IL-6 or IL-10 responses to endotoxin were found between Hib vaccination responders and non-responders. Conclusions There was no correlation between the pneumococcal responses and that to Hib, indicating that B-cell function is not similarly depressed as T-cell function. Grading of the trauma or parameters reflecting the innate immune response could not predict the T-cell-dependent vaccine response. There is a need of further studies evaluating the vaccine response after neurotrauma.
Collapse
Affiliation(s)
- Anna Ljunghill Hedberg
- Department of Medical Sciences, Section of Infectious Diseases, Uppsala University, Uppsala, Sweden
| | - Karlis Pauksens
- Department of Medical Sciences, Section of Infectious Diseases, Uppsala University, Uppsala, Sweden
| | - Per Enblad
- Department of Medical Sciences, Section of Neurosurgery, Uppsala University, Uppsala, Sweden
| | - Anders Larsson
- Department of Medical Sciences, Section of Clinical Chemistry, Uppsala University, Uppsala, Sweden
| | - Jan Sjölin
- Department of Medical Sciences, Section of Infectious Diseases, Uppsala University, Uppsala, Sweden
| |
Collapse
|
36
|
Zhan B, Shen J. Mitochondria and their potential role in acute lung injury (Review). Exp Ther Med 2022; 24:479. [PMID: 35761815 PMCID: PMC9214601 DOI: 10.3892/etm.2022.11406] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 05/16/2022] [Indexed: 11/06/2022] Open
Affiliation(s)
- Biao Zhan
- Center of Emergency and Critical Medicine, Jinshan Hospital of Fudan University, Shanghai 201508, P.R. China
| | - Jie Shen
- Center of Emergency and Critical Medicine, Jinshan Hospital of Fudan University, Shanghai 201508, P.R. China
| |
Collapse
|
37
|
Vourc’h M, Roquilly A, Foucher A, Retiere C, Feuillet F, Devi S, McWilliam HE, Braudeau C, Bourreille G, Hachani A, O’Kane D, Mueller SN, Ischia J, Roussel JC, Rigal JC, Josien R, Rozec B, Villadangos JA, Asehnoune K. Transfusion-Related Renal Dysfunction After Cardiac Surgery. JACC Basic Transl Sci 2022; 7:627-638. [PMID: 35958696 PMCID: PMC9357562 DOI: 10.1016/j.jacbts.2022.02.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/22/2022] [Accepted: 02/23/2022] [Indexed: 11/25/2022]
Abstract
Following cardiac surgery, 20% of patients will present with AKI, which is associated with increased mortality, and transfusion increases the risk of AKI. The main objective was to determine whether the composition of transfusion was associated with AKI. In this study, AKI patients received higher amount of MRP_14 through transfusion vs non-AKI. MRP_14 has been reported to activate and enhance neutrophil transmigration into damaged tissues. In a murine model of ischemia-reperfusion, MRP_14 increased renal damage and enhanced neutrophil influx into the kidney. MRP_14 also increased neutrophilic-trogocytosis toward tubular cells. The sex of the donor and the method of preparation of the blood determined the concentration of MRP_14 in packed red blood cells.
Transfusion is a specific cause of acute kidney injury (AKI) after cardiac surgery. Whether there is an association between the composition of blood products and the onset of AKI is unknown. The present study suggests that the transfusion of packed red blood cells containing a high amount of myeloid-related protein 14 (MRP_14) could increase the incidence of AKI after cardiac surgery. In a mouse model, MRP_14 increased the influx of neutrophils in the kidney after ischemia-reperfusion and their ability to damage tubular cells. Higher concentrations of MRP_14 were found in packed red blood cells from female donors or prepared by whole blood filtration.
Collapse
|
38
|
Ribonuclease-1 treatment after traumatic brain injury preserves blood-brain barrier integrity and delays secondary brain damage in mice. Sci Rep 2022; 12:5731. [PMID: 35388024 PMCID: PMC8986812 DOI: 10.1038/s41598-022-09326-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 03/22/2022] [Indexed: 11/08/2022] Open
Abstract
Traumatic brain injury (TBI) involves primary mechanical damage and delayed secondary damage caused by vascular dysfunction and neuroinflammation. Intracellular components released into the parenchyma and systemic circulation, termed danger-associated molecular patterns (DAMPs), are major drivers of vascular dysfunction and neuroinflammation. These DAMPs include cell-free RNAs (cfRNAs), which damage the blood-brain barrier (BBB), thereby promoting edema, procoagulatory processes, and infiltration of inflammatory cells. We tested the hypothesis that intraperitoneal injection of Ribonuclease-1 (RNase1, two doses of 20, 60, or 180 µg/kg) at 30 min and 12 h after controlled-cortical-impact (CCI) can reduce secondary lesion expansion compared to vehicle treatment 24 h and 120 h post-CCI. The lowest total dose (40 µg/kg) was most effective at reducing lesion volume (- 31% RNase 40 µg/kg vs. vehicle), brain water accumulation (- 5.5%), and loss of BBB integrity (- 21.6%) at 24 h post-CCI. RNase1 also reduced perilesional leukocyte recruitment (- 53.3%) and microglial activation (- 18.3%) at 120 h post-CCI, but there was no difference in lesion volume at this time and no functional benefit. Treatment with RNase1 in the early phase following TBI stabilizes the BBB and impedes leukocyte immigration, thereby suppressing neuroinflammation. RNase1-treatment may be a novel approach to delay brain injury to extend the window for treatment opportunities after TBI.
Collapse
|
39
|
The Influence of Gut Dysbiosis in the Pathogenesis and Management of Ischemic Stroke. Cells 2022; 11:cells11071239. [PMID: 35406804 PMCID: PMC8997586 DOI: 10.3390/cells11071239] [Citation(s) in RCA: 64] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/21/2022] [Accepted: 03/25/2022] [Indexed: 12/12/2022] Open
Abstract
Recent research on the gut microbiome has revealed the influence of gut microbiota (GM) on ischemic stroke pathogenesis and treatment outcomes. Alterations in the diversity, abundance, and functions of the gut microbiome, termed gut dysbiosis, results in dysregulated gut–brain signaling, which induces intestinal barrier changes, endotoxemia, systemic inflammation, and infection, affecting post-stroke outcomes. Gut–brain interactions are bidirectional, and the signals from the gut to the brain are mediated by microbially derived metabolites, such as trimethylamine N-oxide (TMAO) and short-chain fatty acids (SCFAs); bacterial components, such as lipopolysaccharide (LPS); immune cells, such as T helper cells; and bacterial translocation via hormonal, immune, and neural pathways. Ischemic stroke affects gut microbial composition via neural and hypothalamic–pituitary–adrenal (HPA) pathways, which can contribute to post-stroke outcomes. Experimental and clinical studies have demonstrated that the restoration of the gut microbiome usually improves stroke treatment outcomes by regulating metabolic, immune, and inflammatory responses via the gut–brain axis (GBA). Therefore, restoring healthy microbial ecology in the gut may be a key therapeutic target for the effective management and treatment of ischemic stroke.
Collapse
|
40
|
Seong SY, Matzinger P, Land WG. Editorial: DAMPs Across the Tree of Life. Front Immunol 2022; 12:844315. [PMID: 35178047 PMCID: PMC8844022 DOI: 10.3389/fimmu.2021.844315] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 12/29/2021] [Indexed: 01/22/2023] Open
Affiliation(s)
- Seung-Yong Seong
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, South Korea.,Wide River Institute of Immunology, Seoul National University, Seoul, South Korea
| | - Polly Matzinger
- Ghost Lab., Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Walter Gottlieb Land
- German Academy for Transplantation Medicine, Munich, Germany.,Molecular ImmunoRheumatology, INSERM UMR_S1109, Laboratory of Excellence Transplantex, University of Strasbourg, Strasbourg, France
| |
Collapse
|
41
|
Fouladseresht H, Ghamar Talepoor A, Eskandari N, Norouzian M, Ghezelbash B, Beyranvand MR, Nejadghaderi SA, Carson-Chahhoud K, Kolahi AA, Safiri S. Potential Immune Indicators for Predicting the Prognosis of COVID-19 and Trauma: Similarities and Disparities. Front Immunol 2022; 12:785946. [PMID: 35126355 PMCID: PMC8815083 DOI: 10.3389/fimmu.2021.785946] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 12/17/2021] [Indexed: 12/15/2022] Open
Abstract
Although cellular and molecular mediators of the immune system have the potential to be prognostic indicators of disease outcomes, temporal interference between diseases might affect the immune mediators, and make them difficult to predict disease complications. Today one of the most important challenges is predicting the prognosis of COVID-19 in the context of other inflammatory diseases such as traumatic injuries. Many diseases with inflammatory properties are usually polyphasic and the kinetics of inflammatory mediators in various inflammatory diseases might be different. To find the most appropriate evaluation time of immune mediators to accurately predict COVID-19 prognosis in the trauma environment, researchers must investigate and compare cellular and molecular alterations based on their kinetics after the start of COVID-19 symptoms and traumatic injuries. The current review aimed to investigate the similarities and differences of common inflammatory mediators (C-reactive protein, procalcitonin, ferritin, and serum amyloid A), cytokine/chemokine levels (IFNs, IL-1, IL-6, TNF-α, IL-10, and IL-4), and immune cell subtypes (neutrophil, monocyte, Th1, Th2, Th17, Treg and CTL) based on the kinetics between patients with COVID-19 and trauma. The mediators may help us to accurately predict the severity of COVID-19 complications and follow up subsequent clinical interventions. These findings could potentially help in a better understanding of COVID-19 and trauma pathogenesis.
Collapse
Affiliation(s)
- Hamed Fouladseresht
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Atefe Ghamar Talepoor
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nahid Eskandari
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Marzieh Norouzian
- Department of Laboratory Sciences, School of Allied Medical Sciences, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Behrooz Ghezelbash
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Reza Beyranvand
- Social Determinants of Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Aria Nejadghaderi
- Research Center for Integrative Medicine in Aging, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Systematic Review and Meta-Analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Kristin Carson-Chahhoud
- Australian Centre for Precision Health, Allied Health and Human Performance, University of South Australia, Adelaide, SA, Australia
- School of Medicine, The University of Adelaide, Adelaide, SA, Australia
| | - Ali-Asghar Kolahi
- Social Determinants of Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeid Safiri
- Social Determinants of Health Research Center, Department of Community Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
42
|
Rumpret M, von Richthofen HJ, Peperzak V, Meyaard L. Inhibitory pattern recognition receptors. J Exp Med 2022; 219:212908. [PMID: 34905019 PMCID: PMC8674843 DOI: 10.1084/jem.20211463] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 10/03/2021] [Accepted: 12/02/2021] [Indexed: 12/18/2022] Open
Abstract
Pathogen- and damage-associated molecular patterns are sensed by the immune system's pattern recognition receptors (PRRs) upon contact with a microbe or damaged tissue. In situations such as contact with commensals or during physiological cell death, the immune system should not respond to these patterns. Hence, immune responses need to be context dependent, but it is not clear how context for molecular pattern recognition is provided. We discuss inhibitory receptors as potential counterparts to activating pattern recognition receptors. We propose a group of inhibitory pattern recognition receptors (iPRRs) that recognize endogenous and microbial patterns associated with danger, homeostasis, or both. We propose that recognition of molecular patterns by iPRRs provides context, helps mediate tolerance to microbes, and helps balance responses to danger signals.
Collapse
Affiliation(s)
- Matevž Rumpret
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.,Oncode Institute, Utrecht, The Netherlands
| | - Helen J von Richthofen
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.,Oncode Institute, Utrecht, The Netherlands
| | - Victor Peperzak
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Linde Meyaard
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.,Oncode Institute, Utrecht, The Netherlands
| |
Collapse
|
43
|
Yu Y, Xu N, Cheng Q, Deng F, Liu M, Zhu A, Min YQ, Zhu D, Huang W, Feng X, Jing X, Chen Y, Yue D, Fan Y, Shu C, Guan Q, Yang Z, Zhao J, Song W, Guo D, Liu H, Zhao J, Lan P, Shi Z, Liu Y, Chen X, Liang H. IFP35 as a promising biomarker and therapeutic target for the syndromes induced by SARS-CoV-2 or influenza virus. Cell Rep 2021; 37:110126. [PMID: 34910942 PMCID: PMC8639452 DOI: 10.1016/j.celrep.2021.110126] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 07/30/2021] [Accepted: 11/22/2021] [Indexed: 01/08/2023] Open
Abstract
Previous studies have shown that the high mortality caused by viruses such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and influenza virus primarily results from complications of a cytokine storm. Therefore, it is critical to identify the key factors participating in the cytokine storm. Here we demonstrate that interferon-induced protein 35 (IFP35) plays an important role in the cytokine storm induced by SARS-CoV-2 and influenza virus infection. We find that the levels of serum IFP35 in individuals with SARS-CoV-2 correlates with severity of the syndrome. Using mouse model and cell assays, we show that IFP35 is released by lung epithelial cells and macrophages after SARS-CoV-2 or influenza virus infection. In addition, we show that administration of neutralizing antibodies against IFP35 considerably reduces lung injury and, thus, the mortality rate of mice exposed to viral infection. Our findings suggest that IFP35 serves as a biomarker and as a therapeutic target in virus-induced syndromes.
Collapse
Affiliation(s)
- Yang Yu
- School of Medicine, Sun Yat-Sen University, Shenzhen, Guangdong 518107, China
| | - Na Xu
- School of Pharmaceutical Science (Shenzhen), Sun Yat-Sen University, Shenzhen, Guangdong 518107, China
| | - Qi Cheng
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei 430030, China
| | - Fei Deng
- State Key Laboratory of Virology and National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Science, Wuhan, Hubei 430071, China
| | - Meiqin Liu
- CAS Key Laboratory of Special Pathogens, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Science, Wuhan 430071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Airu Zhu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, China
| | - Yuan-Qin Min
- State Key Laboratory of Virology and National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Science, Wuhan, Hubei 430071, China
| | - Dan Zhu
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, NHFPC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education; Beijing Key Laboratory of Cardiovascular Receptors Research. Beijing 100191, China
| | - Wenbo Huang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, China
| | - Xu Feng
- School of Pharmaceutical Science (Shenzhen), Sun Yat-Sen University, Shenzhen, Guangdong 518107, China
| | - Xizhong Jing
- School of Medicine, Sun Yat-Sen University, Shenzhen, Guangdong 518107, China
| | - Ying Chen
- CAS Key Laboratory of Special Pathogens, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Science, Wuhan 430071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Daoyuan Yue
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yawei Fan
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei 430030, China
| | - Chang Shu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei 430030, China
| | - Qing Guan
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Zifeng Yang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, China
| | - Jincun Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, China
| | - Wenjun Song
- State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine, Guangzhou Medical University, Guangzhou, Guangdong 510180, China
| | - Deyin Guo
- School of Medicine, Sun Yat-Sen University, Shenzhen, Guangdong 518107, China
| | - Huanliang Liu
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, Guangdong 510655, China; Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Jindong Zhao
- State Key Laboratory of Protein and Plant Genetic Engineering, College of Life Sciences, Peking University, Beijing 100871, China
| | - Ping Lan
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, Guangdong 510655, China.
| | - Zhengli Shi
- CAS Key Laboratory of Special Pathogens, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Science, Wuhan 430071, China.
| | - Yingfang Liu
- School of Medicine, Sun Yat-Sen University, Shenzhen, Guangdong 518107, China; Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, Guangdong 510655, China.
| | - Xiaoping Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei 430030, China; Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei 430030, China.
| | - Huanhuan Liang
- School of Pharmaceutical Science (Shenzhen), Sun Yat-Sen University, Shenzhen, Guangdong 518107, China.
| |
Collapse
|
44
|
Schenz J, Heilig L, Lohse T, Tichy L, Bomans K, Büttner M, Weigand MA, Uhle F. Extracellular Lactate Acts as a Metabolic Checkpoint and Shapes Monocyte Function Time Dependently. Front Immunol 2021; 12:729209. [PMID: 34899690 PMCID: PMC8652120 DOI: 10.3389/fimmu.2021.729209] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 10/25/2021] [Indexed: 12/26/2022] Open
Abstract
Elevated blood lactate levels are frequently found in critically ill patients and thought to result from tissue hypoperfusion and cellular oxygen shortage. Considering the close relationship between immune cell function and intracellular metabolism, lactate is more than a glycolytic waste molecule but able to regulate the immune response. Our aim was to elucidate the temporal and mechanistic effect of extracellular lactate on monocytes. To this end, primary human monocytes and the human monocytic cell line MonoMac6 were stimulated with various toll-like-receptor agonists after priming with Na-L-lactate under constant pH conditions. As readout, cytokine production was measured, real-time assessment of intracellular energy pathways was performed, and intracellular metabolite concentrations were determined. Irrespective of the immunogenic stimulus, short-term Na-lactate-priming strongly reduced cytokine production capacity. Lactate and hexoses accumulated intracellularly and, together with a decreased glycolytic flux, indicate a lactate-triggered impairment of glycolysis. To counteract intracellular hyperglycemia, glucose is shunted into the branching polyol pathway, leading to sorbitol accumulation. In contrast, long-term priming with Na-L-lactate induced cellular adaption and abolished the suppressive effect. This lactate tolerance is characterized by a decreased cellular respiration due to a reduced complex-I activity. Our results indicate that exogenous lactate shapes monocyte function by altering the intracellular energy metabolism and acts as a metabolic checkpoint of monocyte activation.
Collapse
Affiliation(s)
- Judith Schenz
- Department of Anesthesiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Lena Heilig
- Department of Anesthesiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Tim Lohse
- Department of Anesthesiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Lucas Tichy
- Department of Anesthesiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Katharina Bomans
- Department of Anesthesiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Michael Büttner
- Metabolomics Core Technology Platform (MCTP) at the Centre for Organismal Studies (COS), Heidelberg University, Heidelberg, Germany
| | - Markus A Weigand
- Department of Anesthesiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Florian Uhle
- Department of Anesthesiology, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
45
|
Udovicic I, Stanojevic I, Djordjevic D, Zeba S, Rondovic G, Abazovic T, Lazic S, Vojvodic D, To K, Abazovic D, Khan W, Surbatovic M. Immunomonitoring of Monocyte and Neutrophil Function in Critically Ill Patients: From Sepsis and/or Trauma to COVID-19. J Clin Med 2021; 10:jcm10245815. [PMID: 34945111 PMCID: PMC8706110 DOI: 10.3390/jcm10245815] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 11/27/2021] [Accepted: 12/01/2021] [Indexed: 12/15/2022] Open
Abstract
Immune cells and mediators play a crucial role in the critical care setting but are understudied. This review explores the concept of sepsis and/or injury-induced immunosuppression and immuno-inflammatory response in COVID-19 and reiterates the need for more accurate functional immunomonitoring of monocyte and neutrophil function in these critically ill patients. in addition, the feasibility of circulating and cell-surface immune biomarkers as predictors of infection and/or outcome in critically ill patients is explored. It is clear that, for critically ill, one size does not fit all and that immune phenotyping of critically ill patients may allow the development of a more personalized approach with tailored immunotherapy for the specific patient. In addition, at this point in time, caution is advised regarding the quality of evidence of some COVID-19 studies in the literature.
Collapse
Affiliation(s)
- Ivo Udovicic
- Clinic of Anesthesiology and Intensive Therapy, Military Medical Academy, Crnotravska 17, 11000 Belgrade, Serbia; (I.U.); (D.D.); (S.Z.); (G.R.); (T.A.)
- Faculty of Medicine of the Military Medical Academy, University of Defence, Crnotravska 17, 11000 Belgrade, Serbia; (I.S.); (S.L.); (D.V.)
| | - Ivan Stanojevic
- Faculty of Medicine of the Military Medical Academy, University of Defence, Crnotravska 17, 11000 Belgrade, Serbia; (I.S.); (S.L.); (D.V.)
- Institute for Medical Research, Military Medical Academy, Crnotravska 17, 11000 Belgrade, Serbia
| | - Dragan Djordjevic
- Clinic of Anesthesiology and Intensive Therapy, Military Medical Academy, Crnotravska 17, 11000 Belgrade, Serbia; (I.U.); (D.D.); (S.Z.); (G.R.); (T.A.)
- Faculty of Medicine of the Military Medical Academy, University of Defence, Crnotravska 17, 11000 Belgrade, Serbia; (I.S.); (S.L.); (D.V.)
| | - Snjezana Zeba
- Clinic of Anesthesiology and Intensive Therapy, Military Medical Academy, Crnotravska 17, 11000 Belgrade, Serbia; (I.U.); (D.D.); (S.Z.); (G.R.); (T.A.)
- Faculty of Medicine of the Military Medical Academy, University of Defence, Crnotravska 17, 11000 Belgrade, Serbia; (I.S.); (S.L.); (D.V.)
| | - Goran Rondovic
- Clinic of Anesthesiology and Intensive Therapy, Military Medical Academy, Crnotravska 17, 11000 Belgrade, Serbia; (I.U.); (D.D.); (S.Z.); (G.R.); (T.A.)
- Faculty of Medicine of the Military Medical Academy, University of Defence, Crnotravska 17, 11000 Belgrade, Serbia; (I.S.); (S.L.); (D.V.)
| | - Tanja Abazovic
- Clinic of Anesthesiology and Intensive Therapy, Military Medical Academy, Crnotravska 17, 11000 Belgrade, Serbia; (I.U.); (D.D.); (S.Z.); (G.R.); (T.A.)
| | - Srdjan Lazic
- Faculty of Medicine of the Military Medical Academy, University of Defence, Crnotravska 17, 11000 Belgrade, Serbia; (I.S.); (S.L.); (D.V.)
- Institute of Epidemiology, Military Medical Academy, Crnotravska 17, 11000 Belgrade, Serbia
| | - Danilo Vojvodic
- Faculty of Medicine of the Military Medical Academy, University of Defence, Crnotravska 17, 11000 Belgrade, Serbia; (I.S.); (S.L.); (D.V.)
- Institute for Medical Research, Military Medical Academy, Crnotravska 17, 11000 Belgrade, Serbia
| | - Kendrick To
- Division of Trauma & Orthopaedic Surgery, University of Cambridge, Addenbrooke’s Hospital, Cambridge CB2 2QQ, UK; (K.T.); (W.K.)
| | - Dzihan Abazovic
- Emergency Medical Centar of Montenegro, Vaka Djurovica bb, 81000 Podgorica, Montenegro;
| | - Wasim Khan
- Division of Trauma & Orthopaedic Surgery, University of Cambridge, Addenbrooke’s Hospital, Cambridge CB2 2QQ, UK; (K.T.); (W.K.)
| | - Maja Surbatovic
- Clinic of Anesthesiology and Intensive Therapy, Military Medical Academy, Crnotravska 17, 11000 Belgrade, Serbia; (I.U.); (D.D.); (S.Z.); (G.R.); (T.A.)
- Faculty of Medicine of the Military Medical Academy, University of Defence, Crnotravska 17, 11000 Belgrade, Serbia; (I.S.); (S.L.); (D.V.)
- Correspondence: ; Tel.: +381-11-2665-125
| |
Collapse
|
46
|
Li X, Liu J, Xing Z, Tang J, Sun H, Zhang X, Lv S, Chen Z, Shi M, Chen M, Zuo S, Lyu X, He Y. Polymorphonuclear myeloid-derived suppressor cells link inflammation and damage response after trauma. J Leukoc Biol 2021; 110:1143-1161. [PMID: 34636072 DOI: 10.1002/jlb.3ma0821-029r] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 08/16/2021] [Accepted: 09/06/2021] [Indexed: 12/12/2022] Open
Abstract
Elimination of the posttraumatic inflammatory response and recovery of homeostasis are crucial for the positive prognosis of trauma patients. Myeloid-derived suppressor cells (MDSCs) are known to play a regulatory role in the posttraumatic immune response in mice, but their induction source and involved potential mechanism are poorly understood. Here, we report that polymorphonuclear MDSCs (PMN-MDSCs) are activated after trauma and are closely associated with the progression of the posttraumatic inflammatory response. In humans, lectin-type oxidized LDL receptor 1 (LOX1) was used to specifically characterize LOX1+ PMN-MDSCs. Trauma patients showed high intracellular reactive oxygen species (ROS) production, as well as activation of LOX1+ PMN-MDSCs. These MDSCs contribute to the anti-inflammatory immune response by regulating the Treg/Th17 and Th2/Th1 balances after trauma, increasing the levels of anti-inflammatory factors, and decreasing the levels of proinflammatory factors. The number of LOX1+ PMN-MDSCs was positively correlated with the positive clinical prognosis of trauma patients with infection. Activation of LOX1+ PMN-MDSCs is mediated by NF-κB signal, and TGF-β1 may be as an important inducer for LOX1+ PMN-MDSCs in the posttraumatic cytokine environment. In a pseudofracture trauma mouse model, we also observed the activation of PMN-MDSCs, accompanying high levels of intracellular ROS production, NF-κB phosphorylation, and changes in the inflammatory environment, in particularly by regulating the Treg/Th17 and Th2/Th1 balance. And more significantly, posttraumatic inflammation was alleviated in mice after transferring trauma-derived PMN-MDSCs, but aggravated after injecting with Gr1 agonistic antibody. These findings provide evidence for the specific role of PMN-MDSCs in the regulation of posttraumatic inflammation.
Collapse
Affiliation(s)
- Xinyao Li
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jingping Liu
- Department of Clinical Laboratory, the Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, China
| | - Zhe Xing
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jian Tang
- Department of Gastroenterology, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hengbiao Sun
- Department of Clinical Laboratory, the Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, China
| | - Xiaogang Zhang
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Shuaijun Lv
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Ziyang Chen
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Mengyu Shi
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Meiqi Chen
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Shaowen Zuo
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xiaoming Lyu
- Department of Clinical Laboratory, the Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, China
| | - Yumei He
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.,Department of Clinical Laboratory, the Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Proteomics, Southern Medical University, Guangzhou, China
| |
Collapse
|
47
|
Brammer J, Choi M, Baliban SM, Kambouris AR, Fiskum G, Chao W, Lopez K, Miller C, Al-Abed Y, Vogel SN, Simon R, Cross AS. A Nonlethal Murine Flame Burn Model Leads to a Transient Reduction in Host Defenses and Enhanced Susceptibility to Lethal Pseudomonas aeruginosa Infection. Infect Immun 2021; 89:e0009121. [PMID: 34152806 PMCID: PMC8445176 DOI: 10.1128/iai.00091-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 06/01/2021] [Indexed: 12/03/2022] Open
Abstract
Of the 486,000 burn injuries that required medical treatment in the United States in 2016, 40,000 people were hospitalized, with >3,000 fatalities. After burn injury, humans are at increased risk of sepsis and mortality from infections caused by Pseudomonas aeruginosa, an opportunistic pathogen. We hypothesize that systemic events were initiated from the burn that increased the host's susceptibility to P. aeruginosa. A nonlethal 10% total body surface area (TBSA), full-thickness flame burn was performed in CD-1 mice without and with subsequent P. aeruginosa (strain M2) infection. The 50% lethal dose for subcutaneous infection with P. aeruginosa M2 at the burn site immediately after the burn decreased by 6 log, with mortality occurring between 18 and 26 h, compared with P. aeruginosa-infected mice without burn injury. Bacteria in distal organs were detected by 18 h, concurrent with the onset of clinical symptoms. Serum proinflammatory cytokines (interleukin-6 [IL-6], IL-1β, gamma interferon, and tumor necrosis factor alpha) and the anti-inflammatory cytokine IL-10 were first detected at 12 h postburn with infection and continued to increase until death. Directly after burn alone, serum levels of HMGB1, a danger-associated molecular pattern and TLR4 agonist, transiently increased to 50 ng/ml before returning to 20 ng/ml. Burn with P. aeruginosa infection increased serum HMGB1 concentrations >10-fold (250 ng/ml) at the time of death. This HMGB1-rich serum stimulated TLR4-mediated NF-κB activation in a TLR4 reporter cell line. Treatment of infected burned mice with P5779, a peptide inhibitor of HMGB1, increased the mean survival from 23 to 42 h (P < 0.0001). We conclude that the high level of serum HMGB1, which preceded the increase in proinflammatory cytokines, is associated with postburn mortality.
Collapse
Affiliation(s)
- Jerod Brammer
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Myeongjin Choi
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Scott M. Baliban
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Adrienne R. Kambouris
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Gary Fiskum
- Translational Research Program, Department of Anesthesiology & Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Wei Chao
- Translational Research Program, Department of Anesthesiology & Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Kerri Lopez
- Translational Research Program, Department of Anesthesiology & Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Catriona Miller
- Enroute Care Division, Department of Aeromedical Research, USAF School of Aerospace Medicine, Wright Patterson AFB, Dayton, Ohio, USA
| | - Yousef Al-Abed
- Department of Biomedical Science, The Feinstein Institute for Medical Research, Manhasset, New York, USA
| | - Stefanie N. Vogel
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Raphael Simon
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Alan S. Cross
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
48
|
Postinjury Sepsis-Associations With Risk Factors, Impact on Clinical Course, and Mortality: A Retrospective Observational Study. Crit Care Explor 2021; 3:e0495. [PMID: 34368768 PMCID: PMC8330967 DOI: 10.1097/cce.0000000000000495] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
OBJECTIVES Overall outcomes for trauma patients have improved over time. However, mortality for postinjury sepsis has been reported to be unchanged. Estimate incidence of and risk factors for sepsis in ICU patients after major trauma and the association between sepsis, mortality, and clinical course. DESIGN SETTING AND PATIENTS ICU in a large urban trauma center in Sweden with a well-developed trauma system. Retrospective cohort study of trauma patients admitted to the ICU for more than 24 hours were included. MEASUREMENTS AND MAIN RESULTS Primary outcome measure was 30-day mortality. Secondary outcomes were 1-year mortality and impact on clinical course. In total, 722 patients with a median Injury Severity Score of 26 (interquartile range, 18-38) were included. Incidence of sepsis was 22%. Septic patients had a four-fold increase in length of stay and need for organ supportive therapy. The overall 30-day mortality rate was 9.3%. After exclusion of early trauma-related deaths in the first 48 hours, the 30-day mortality rate was 6.7%. There was an association between sepsis and this adjusted 30-day mortality (day 3 odds ratio, 2.1 [95% CI, 1.1-3.9]; day 4 odds ratio, 3.1 [95% CI, 1.5-6.1]; day 5 odds ratio, 3.0 [95% CI, 1.4-6.2]). Septic patients had a 1-year mortality of 17.7% (nonseptic 11.0%). Development of sepsis was independently associated with age, spine and chest injury, shock, red cell transfusion, and positive blood alcohol concentration at admission. The risk of sepsis increased, in a dose-dependent manner, with the number of transfusions. CONCLUSIONS Postinjury sepsis was associated with a complicated clinical course and with mortality after exclusion of early, trauma-related deaths.
Collapse
|
49
|
Chen K, Cagliani J, Aziz M, Tan C, Brenner M, Wang P. Extracellular CIRP activates STING to exacerbate hemorrhagic shock. JCI Insight 2021; 6:e143715. [PMID: 34291735 PMCID: PMC8410031 DOI: 10.1172/jci.insight.143715] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 06/09/2021] [Indexed: 01/07/2023] Open
Abstract
Stimulator of IFN genes (STING) activates TANK-binding kinase 1 (TBK1) and IFN regulatory factor 3 (IRF3) to produce type I IFNs. Extracellular cold-inducible RNA-binding protein (eCIRP) is released from cells during hemorrhagic shock (HS). We hypothesized that eCIRP activates STING to induce inflammation and acute lung injury (ALI) after HS. WT and STING–/– mice underwent controlled hemorrhage by bleeding, followed by fluid resuscitation. Blood and lungs were collected at 4 hours after resuscitation. Serum ALT, AST, LDH, IL-6, and IFN-β were significantly decreased in STING–/– mice compared with WT mice after HS. In STING–/– mice, the levels of pTBK1 and pIRF3, and expression of TNF-α, IL-6, and IL-1β mRNAs and proteins in the lungs, were significantly decreased compared with WT HS mice. The 10-day mortality rate in STING–/– mice was significantly reduced. I.v. injection of recombinant mouse CIRP (rmCIRP) in STING–/– mice showed a significant decrease in pTBK1 and pIRF3 and in IFN-α and IFN-β mRNAs and proteins in the lungs compared with rmCIRP-treated WT mice. Treatment of TLR4–/–, MyD88–/–, and TRIF–/– macrophages with rmCIRP significantly decreased pTBK1 and pIRF3 levels and IFN-α and IFN-β mRNAs and proteins compared with WT macrophages. HS increases eCIRP levels, which activate STING through TLR4/MyD88/TRIF pathways to exacerbate inflammation.
Collapse
Affiliation(s)
- Kehong Chen
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Joaquin Cagliani
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Monowar Aziz
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Chuyi Tan
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Max Brenner
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Ping Wang
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, New York, USA.,Department of Surgery and Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, New York, USA
| |
Collapse
|
50
|
Land WG. Role of DAMPs in respiratory virus-induced acute respiratory distress syndrome-with a preliminary reference to SARS-CoV-2 pneumonia. Genes Immun 2021; 22:141-160. [PMID: 34140652 PMCID: PMC8210526 DOI: 10.1038/s41435-021-00140-w] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 04/28/2021] [Accepted: 05/25/2021] [Indexed: 02/05/2023]
Abstract
When surveying the current literature on COVID-19, the "cytokine storm" is considered to be pathogenetically involved in its severe outcomes such as acute respiratory distress syndrome, systemic inflammatory response syndrome, and eventually multiple organ failure. In this review, the similar role of DAMPs is addressed, that is, of those molecules, which operate upstream of the inflammatory pathway by activating those cells, which ultimately release the cytokines. Given the still limited reports on their role in COVID-19, the emerging topic is extended to respiratory viral infections with focus on influenza. At first, a brief introduction is given on the function of various classes of activating DAMPs and counterbalancing suppressing DAMPs (SAMPs) in initiating controlled inflammation-promoting and inflammation-resolving defense responses upon infectious and sterile insults. It is stressed that the excessive emission of DAMPs upon severe injury uncovers their fateful property in triggering dysregulated life-threatening hyperinflammatory responses. Such a scenario may happen when the viral load is too high, for example, in the respiratory tract, "forcing" many virus-infected host cells to decide to commit "suicidal" regulated cell death (e.g., necroptosis, pyroptosis) associated with release of large amounts of DAMPs: an important topic of this review. Ironically, although the aim of this "suicidal" cell death is to save and restore organismal homeostasis, the intrinsic release of excessive amounts of DAMPs leads to those dysregulated hyperinflammatory responses-as typically involved in the pathogenesis of acute respiratory distress syndrome and systemic inflammatory response syndrome in respiratory viral infections. Consequently, as briefly outlined in this review, these molecules can be considered valuable diagnostic and prognostic biomarkers to monitor and evaluate the course of the viral disorder, in particular, to grasp the eventual transition precociously from a controlled defense response as observed in mild/moderate cases to a dysregulated life-threatening hyperinflammatory response as seen, for example, in severe/fatal COVID-19. Moreover, the pathogenetic involvement of these molecules qualifies them as relevant future therapeutic targets to prevent severe/ fatal outcomes. Finally, a theory is presented proposing that the superimposition of coronavirus-induced DAMPs with non-virus-induced DAMPs from other origins such as air pollution or high age may contribute to severe and fatal courses of coronavirus pneumonia.
Collapse
Affiliation(s)
- Walter Gottlieb Land
- German Academy for Transplantation Medicine, Munich, Germany.
- Molecular ImmunoRheumatology, INSERM UMR_S1109, Laboratory of Excellence Transplantex, University of Strasbourg, Strasbourg, France.
| |
Collapse
|