1
|
Da Silva K, Kumar P, Choonara YE. The paradigm of stem cell secretome in tissue repair and regeneration: Present and future perspectives. Wound Repair Regen 2025; 33:e13251. [PMID: 39780313 PMCID: PMC11711308 DOI: 10.1111/wrr.13251] [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: 08/01/2024] [Revised: 12/04/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025]
Abstract
As the number of patients requiring organ transplants continues to rise exponentially, there is a dire need for therapeutics, with repair and regenerative properties, to assist in alleviating this medical crisis. Over the past decade, there has been a shift from conventional stem cell treatments towards the use of the secretome, the protein and factor secretions from cells. These components may possess novel druggable targets and hold the key to profoundly altering the field of regenerative medicine. Despite the progress in this field, clinical translation of secretome-containing products is limited by several challenges including but not limited to ensuring batch-to-batch consistency, the prevention of further heterogeneity, production of sufficient secretome quantities, product registration, good manufacturing practice protocols and the pharmacokinetic/pharmacodynamic profiles of all the components. Despite this, the secretome may hold the key to unlocking the regenerative blockage scientists have encountered for years. This review critically analyses the secretome derived from different cell sources and used in several tissues for tissue regeneration. Furthermore, it provides an overview of the current delivery strategies and the future perspectives for the secretome as a potential therapeutic. The success and possible shortcomings of the secretome are evaluated.
Collapse
Affiliation(s)
- Kate Da Silva
- Wits Advanced Drug Delivery Platform (WADDP) Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health SciencesUniversity of the WitwatersrandJohannesburgSouth Africa
| | - Pradeep Kumar
- Wits Advanced Drug Delivery Platform (WADDP) Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health SciencesUniversity of the WitwatersrandJohannesburgSouth Africa
| | - Yahya E. Choonara
- Wits Advanced Drug Delivery Platform (WADDP) Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health SciencesUniversity of the WitwatersrandJohannesburgSouth Africa
| |
Collapse
|
2
|
Konkayev A, Bekniyazova A, Khamidullina Z, Konkayeva M. Case series report: use of vibroacoustic pulmonary therapy in patients with thoracic trauma complicated by acute respiratory failure. Front Med (Lausanne) 2024; 11:1399397. [PMID: 39296888 PMCID: PMC11408177 DOI: 10.3389/fmed.2024.1399397] [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: 03/11/2024] [Accepted: 08/19/2024] [Indexed: 09/21/2024] Open
Abstract
Chest injury is often accompanied by polytrauma and is complicated by respiratory failure. This article presents a series of cases with verified acute respiratory failure in patients with chest injury, where vibroacoustic pulmonary therapy was used in complex treatment. Dynamic X-rays and respiratory drive indicators reflected the effect of the use of vibroacoustic lung therapy. Early diagnosis of respiratory disorders and complex therapy using vibroacoustic pulmonary therapy can improve results. As a result, the time spent in the hospital and in the intensive care unit is reduced, and the frequency of adverse outcomes is reduced.
Collapse
Affiliation(s)
- Aidos Konkayev
- Astana Medical University, Astana, Kazakhstan
- The National Scientific Center of Traumatology and Orthopedics named after Academician Batpenov N.D., Astana, Kazakhstan
| | - Assema Bekniyazova
- Astana Medical University, Astana, Kazakhstan
- The National Scientific Center of Traumatology and Orthopedics named after Academician Batpenov N.D., Astana, Kazakhstan
- National Research Oncology Centre, Astana, Kazakhstan
| | | | - Maiya Konkayeva
- Astana Medical University, Astana, Kazakhstan
- The National Scientific Center of Traumatology and Orthopedics named after Academician Batpenov N.D., Astana, Kazakhstan
| |
Collapse
|
3
|
Yang Z, Nicholson SE, Cancio TS, Cancio LC, Li Y. Complement as a vital nexus of the pathobiological connectome for acute respiratory distress syndrome: An emerging therapeutic target. Front Immunol 2023; 14:1100461. [PMID: 37006238 PMCID: PMC10064147 DOI: 10.3389/fimmu.2023.1100461] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 02/27/2023] [Indexed: 03/19/2023] Open
Abstract
The hallmark of acute respiratory distress syndrome (ARDS) pathobiology is unchecked inflammation-driven diffuse alveolar damage and alveolar-capillary barrier dysfunction. Currently, therapeutic interventions for ARDS remain largely limited to pulmonary-supportive strategies, and there is an unmet demand for pharmacologic therapies targeting the underlying pathology of ARDS in patients suffering from the illness. The complement cascade (ComC) plays an integral role in the regulation of both innate and adaptive immune responses. ComC activation can prime an overzealous cytokine storm and tissue/organ damage. The ARDS and acute lung injury (ALI) have an established relationship with early maladaptive ComC activation. In this review, we have collected evidence from the current studies linking ALI/ARDS with ComC dysregulation, focusing on elucidating the new emerging roles of the extracellular (canonical) and intracellular (non-canonical or complosome), ComC (complementome) in ALI/ARDS pathobiology, and highlighting complementome as a vital nexus of the pathobiological connectome for ALI/ARDS via its crosstalking with other systems of the immunome, DAMPome, PAMPome, coagulome, metabolome, and microbiome. We have also discussed the diagnostic/therapeutic potential and future direction of ALI/ARDS care with the ultimate goal of better defining mechanistic subtypes (endotypes and theratypes) through new methodologies in order to facilitate a more precise and effective complement-targeted therapy for treating these comorbidities. This information leads to support for a therapeutic anti-inflammatory strategy by targeting the ComC, where the arsenal of clinical-stage complement-specific drugs is available, especially for patients with ALI/ARDS due to COVID-19.
Collapse
Affiliation(s)
- Zhangsheng Yang
- Combat Casualty Care Research Team (CRT) 3, United States (US) Army Institute of Surgical Research, Joint Base San Antonio (JBSA)-Fort Sam Houston, TX, United States
| | - Susannah E. Nicholson
- Division of Trauma Research, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Tomas S. Cancio
- Combat Casualty Care Research Team (CRT) 3, United States (US) Army Institute of Surgical Research, Joint Base San Antonio (JBSA)-Fort Sam Houston, TX, United States
| | - Leopoldo C. Cancio
- United States (US) Army Burn Center, United States (US) Army Institute of Surgical Research, Joint Base San Antonio (JBSA)-Fort Sam Houston, TX, United States
| | - Yansong Li
- Division of Trauma Research, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
- The Geneva Foundation, Immunological Damage Control Resuscitation Program, Tacoma, WA, United States
- *Correspondence: Yansong Li,
| |
Collapse
|
4
|
Schönfelder J, Seibold T, Morawe M, Sroka R, Schneider N, Cai J, Golomejic J, Schütte L, Armacki M, Huber-Lang M, Kalbitz M, Seufferlein T, Eiseler T. Endothelial Protein kinase D1 is a major regulator of post-traumatic hyperinflammation. Front Immunol 2023; 14:1093022. [PMID: 36936923 PMCID: PMC10017463 DOI: 10.3389/fimmu.2023.1093022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 02/15/2023] [Indexed: 03/06/2023] Open
Abstract
Trauma is a major cause of death worldwide. The post-traumatic immune response culminates in the release of pro-inflammatory mediators, translating in the infiltration of neutrophils (PMNs) at injury sites. The extent of this inflammation is determined by multiple factors, such as PMN adhesion to the endothelium, transendothelial migration, endothelial barrier integrity as well as PMN swarming, mass infiltration and activation. This process is initiated by secondary lipid mediators, such as leukotriene B4 (LTB4). We here provide evidence that Protein kinase D1 (PRKD1) in endothelial cells is implicated in all these processes. Endothelial PRKD1 is activated by pro-inflammatory stimuli and amplifies PMN-mediated inflammation by upregulation of cytokine and chemokines as well as adhesion molecules, such as ICAM-1, VCAM-1 and E-selectin. This induces enhanced PMN adhesion and trans-migration. PRKD1 activation also destabilizes endothelial VE-cadherin adhesion complexes and thus the endothelial barrier, fostering PMN infiltration. We even describe a yet unrecognized PRKD1-dependant mechanism to induce biosynthesis of the PMN-swarming mediator LTB4 directed via intercellular communication through small extracellular vesicles (sEVs) and enhanced CXCL8 secretion from activated endothelial cells. These endothelial sEVs transfer the LTB4 biosynthesis enzyme LTA4 hydrolase (LTA4H) to prime PMNs, while initiating biosynthesis also requires additional signals, like CXCL8. We further demonstrate the respective LTA4H-positive sEVs in the serum of polytrauma patients, peaking 12 h post injury. Therefore, PRKD1 is a key regulator in the coordinated communication of the endothelium with PMNs and a vital signaling node during post-traumatic inflammation.
Collapse
Affiliation(s)
| | - Tanja Seibold
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Mareen Morawe
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Robert Sroka
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Nora Schneider
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Jierui Cai
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Josip Golomejic
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Lena Schütte
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Milena Armacki
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma-Immunology, University Hospital Ulm, Ulm, Germany
| | - Miriam Kalbitz
- Department of Traumatology, Hand-, Plastic, and Reconstructive Surgery, University Hospital Ulm, Ulm, Germany
| | - Thomas Seufferlein
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
- *Correspondence: Tim Eiseler, ; Thomas Seufferlein,
| | - Tim Eiseler
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
- *Correspondence: Tim Eiseler, ; Thomas Seufferlein,
| |
Collapse
|
5
|
Bonenfant J, Li J, Nasouf L, Miller J, Lowe T, Jaroszewski L, Qiu X, Thapamagar S, Mittal A, Godzik A, Klein W, Nair MG. Resistin Concentration in Early Sepsis and All-Cause Mortality at a Safety-Net Hospital in Riverside County. J Inflamm Res 2022; 15:3925-3940. [PMID: 35860230 PMCID: PMC9289958 DOI: 10.2147/jir.s370788] [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: 04/14/2022] [Accepted: 06/29/2022] [Indexed: 11/23/2022] Open
Abstract
Background Sepsis mortality has remained unchanged for greater than a decade, and early recognition continues to be the most important factor in mortality outcome. Plasma resistin concentration is increased in sepsis, but its mechanism and clinical relevance is unclear. As one function, resistin interacts with toll-like receptor 4 in competition with lipopolysaccharide, a main component of the gram-negative bacterial cell wall. It is not known if the type of infection leading to sepsis influences resistin production. The objective of this study was to investigate whether 1) early plasma resistin concentration can predict mortality, 2) elevated plasma resistin concentration is associated with clinical disease severity scores, such as SOFA, mSOFA and APACHE II, and 3) plasma resistin concentrations differ between gram negative versus other etiologies of sepsis. Methods This was an exploratory study in the framework of a prospective observational design. Peripheral venous blood samples were obtained from subjects admitted to the intensive care unit at clinical recognition of sepsis (0 hour) and at 6 and 24 hours. Vasopressor utilization was not a requirement for inclusion. Plasma was analyzed for resistin concentration by ELISA. Cytokine concentrations including IL-6, IL-8, and IL-10 were determined by cytokine bead array. Cytokine data were evaluated against publicly available sepsis RNA expression datasets to compare protein versus RNA expression levels in predicting clinical disease state. Clinical data were collected from electronic health records for clinical severity index calculations and context for interpretation of resistin and cytokine concentrations. Subjects were followed up to 60 days, or until death, whichever came first. Statistical analysis was completed with R package and SPSS software. Results Resistin levels were elevated in subjects admitted to the intensive care unit with sepsis. Four-hundred subjects were screened with 45 subjects included in the final analysis. Thirteen of 45 patients were non-survivors. Mortality within 60 days correlated with significantly higher resistin concentrations than in survivors. A resistin concentration of >126 ng/mL at clinical recognition of sepsis and >197 ng/mL within the first 24 hours were associated with mortality within 60 days with an area under the curve of 0.82 and 0.88, respectively. Most subjects with resistin concentration greater than these threshold values were deceased prior to 30 days. Resistin concentrations correlated with SOFA, mSOFA, and APACHE II scores in addition to having association with increases in inflammatory and sepsis biomarkers. These associations were validated with analysis of RNA expression datasets. Conclusion Plasma resistin concentrations of >126 ng/mL at clinical recognition of sepsis and >197 ng/mL within the first 24 hours of clinical sepsis recognition are associated with all-cause mortality. Resistin concentration within this timeframe also has comparable mortality association to well-validated clinical severity indices of SOFA, mSOFA, and APACHE II scores.
Collapse
Affiliation(s)
- Jeffrey Bonenfant
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Riverside University Health System Medical Center, Moreno Valley, CA, USA.,Division of Pulmonary, Critical Care, Hyperbaric, Allergy and Sleep Medicine, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Jiang Li
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, USA
| | - Luqman Nasouf
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Riverside University Health System Medical Center, Moreno Valley, CA, USA
| | - Joseph Miller
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, USA
| | - Tammy Lowe
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Riverside University Health System Medical Center, Moreno Valley, CA, USA
| | - Lukasz Jaroszewski
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, USA
| | - Xinru Qiu
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, USA
| | - Suman Thapamagar
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Riverside University Health System Medical Center, Moreno Valley, CA, USA.,Division of Pulmonary, Critical Care, Hyperbaric, Allergy and Sleep Medicine, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Aarti Mittal
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Riverside University Health System Medical Center, Moreno Valley, CA, USA.,Division of Pulmonary, Critical Care, Hyperbaric, Allergy and Sleep Medicine, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Adam Godzik
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, USA
| | - Walter Klein
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Riverside University Health System Medical Center, Moreno Valley, CA, USA.,Division of Pulmonary, Critical Care, Hyperbaric, Allergy and Sleep Medicine, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Meera G Nair
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, USA
| |
Collapse
|
6
|
Ma X, Dong Z, Wang Y, Gu P, Fang J, Gao S. Risk Factors Analysis of Thoracic Trauma Complicated With Acute Respiratory Distress Syndrome and Observation of Curative Effect of Lung-Protective Ventilation. Front Surg 2022; 8:826682. [PMID: 35141272 PMCID: PMC8818796 DOI: 10.3389/fsurg.2021.826682] [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: 12/01/2021] [Accepted: 12/14/2021] [Indexed: 11/27/2022] Open
Abstract
Purpose To explore the risk factors of acute respiratory distress syndrome (ARDS) secondary to thoracic trauma and the therapeutic effect of protective lung ventilation in patients with acute respiratory distress syndrome complicated with thoracic trauma. Methods We collected 206 patients with thoracic trauma admitted to our hospital from September 2017 to March 2021, counted the incidence of ARDS and analyzed the risk factors of ARDS. To observe the clinical efficacy of the application of lung-protective ventilation therapy in patients with thoracic trauma combined with ARDS. Results Among 206 patients with thoracic trauma, there were 82 cases of combined ARDS, and its incidence was 39.81%. The 82 patients with ARDS were randomly divided into the control group and the observation group with 42 cases each, and different ventilation methods were used for treatment. The results showed that the mechanical ventilation time (MVT) was shorter in the observation group than in the control group, and the incidence of ventilator-associated lung injury (VALI) and case fatality rate (CFR) were lower than those in the control group (P < 0.05). Arterial partial pressure of oxygen (Pa02), arterial blood carbon dioxide partial pressure (PaCO2), and Oxygenation index (arterial partial pressure of oxygen/Fraction of inspiration O2, PaO2/FiO2) were significantly improved better in both groups after treatment; compared with the control group, patients in the observation group had higher Pa02 levels and lower PaCO2 levels at 8 h and 24 h after ventilation (P < 0.05). Multivariate analysis revealed that blunt trauma, massive blood transfusion, procalcitonin (PCT) level, tumor necrosis factor-α (TNF-α) level, and acute physiology and chronic health score (APACHE II) were all risk factors for Thoracic trauma with ARDS. Conclusion Risk factors for the development of ARDS after thoracic trauma are blunt injuries, massive blood transfusion, high PCT and TNF-α levels, and high APACHE II scores, which can be given active interventions in the early stage of clinical practice to improve patient prognosis. The use of protective lung ventilation therapy can improve the clinical outcome of patients with thoracic trauma combined with ARDS, which is important for improving the ventilation effect and respiratory function of patients.
Collapse
Affiliation(s)
- Xiaoyu Ma
- Department of Thoracic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zefang Dong
- Department of Thoracic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yusuo Wang
- Ningjin County Maternity and Childcare Hospital of Hebei Province Emergency Surgery, Xingtai, China
| | - Peidong Gu
- Ningjin County Maternity and Childcare Hospital of Hebei Province Emergency Surgery, Xingtai, China
| | - Jinghua Fang
- Ningjin County Maternity and Childcare Hospital of Hebei Province Emergency Surgery, Xingtai, China
| | - Shaolin Gao
- Department of Thoracic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- *Correspondence: Shaolin Gao
| |
Collapse
|
7
|
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: 2.7] [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
|
8
|
Seibold T, Schönfelder J, Weeber F, Lechel A, Armacki M, Waldenmaier M, Wille C, Palmer A, Halbgebauer R, Karasu E, Huber‐Lang M, Kalbitz M, Radermacher P, Paschke S, Seufferlein T, Eiseler T. Small Extracellular Vesicles Propagate the Inflammatory Response After Trauma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2102381. [PMID: 34713625 PMCID: PMC8693079 DOI: 10.1002/advs.202102381] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 08/20/2021] [Indexed: 05/03/2023]
Abstract
Trauma is the leading cause of death in individuals under 44 years of age. Thorax trauma (TxT) is strongly associated with trauma-related death, an unbalanced innate immune response, sepsis, acute respiratory distress syndrome, and multiple organ dysfunction. It is shown that different in vivo traumata, such as TxT or an in vitro polytrauma cytokine cocktail trigger secretion of small extracellular nanovesicles (sEVs) from endothelial cells with pro-inflammatory cargo. These sEVs transfer transcripts for ICAM-1, VCAM-1, E-selectin, and cytokines to systemically activate the endothelium, facilitate neutrophil-endothelium interactions, and destabilize barrier integrity. Inhibition of sEV-release after TxT in mice ameliorates local as well as systemic inflammation, neutrophil infiltration, and distant organ damage in kidneys (acute kidney injury, AKI). Vice versa, injection of TxT-plasma-sEVs into healthy animals is sufficient to trigger pulmonary and systemic inflammation as well as AKI. Accordingly, increased sEV concentrations and transfer of similar cargos are observed in polytrauma patients, suggesting a fundamental pathophysiological mechanism.
Collapse
Affiliation(s)
- Tanja Seibold
- Department of Internal Medicine IUniversity Hospital UlmAlbert‐Einstein‐Allee 23Ulm89081Germany
| | - Jonathan Schönfelder
- Department of Internal Medicine IUniversity Hospital UlmAlbert‐Einstein‐Allee 23Ulm89081Germany
| | - Florian Weeber
- Department of Internal Medicine IUniversity Hospital UlmAlbert‐Einstein‐Allee 23Ulm89081Germany
| | - André Lechel
- Department of Internal Medicine IUniversity Hospital UlmAlbert‐Einstein‐Allee 23Ulm89081Germany
| | - Milena Armacki
- Department of Internal Medicine IUniversity Hospital UlmAlbert‐Einstein‐Allee 23Ulm89081Germany
| | - Mareike Waldenmaier
- Department of Internal Medicine IUniversity Hospital UlmAlbert‐Einstein‐Allee 23Ulm89081Germany
| | - Christoph Wille
- Department of Internal Medicine IUniversity Hospital UlmAlbert‐Einstein‐Allee 23Ulm89081Germany
| | - Annette Palmer
- Institute of Clinical and Experimental Trauma‐ImmunologyUniversity Hospital UlmAlbert‐Einstein‐Allee 23Ulm89081Germany
| | - Rebecca Halbgebauer
- Institute of Clinical and Experimental Trauma‐ImmunologyUniversity Hospital UlmAlbert‐Einstein‐Allee 23Ulm89081Germany
| | - Ebru Karasu
- Institute of Clinical and Experimental Trauma‐ImmunologyUniversity Hospital UlmAlbert‐Einstein‐Allee 23Ulm89081Germany
| | - Markus Huber‐Lang
- Institute of Clinical and Experimental Trauma‐ImmunologyUniversity Hospital UlmAlbert‐Einstein‐Allee 23Ulm89081Germany
| | - Miriam Kalbitz
- Department of TraumatologyHandPlastic and Reconstructive SurgeryUniversity Hospital UlmAlbert‐Einstein‐Allee 23Ulm89081Germany
| | - Peter Radermacher
- Institute of Anesthesiological Pathophysiology and Process EngineeringUniversity Hospital UlmAlbert‐Einstein‐Allee 23Ulm89081Germany
| | - Stephan Paschke
- Department of General and Visceral SurgeryUniversity HospitalAlbert‐Einstein‐Allee 23Ulm89081Germany
| | - Thomas Seufferlein
- Department of Internal Medicine IUniversity Hospital UlmAlbert‐Einstein‐Allee 23Ulm89081Germany
| | - Tim Eiseler
- Department of Internal Medicine IUniversity Hospital UlmAlbert‐Einstein‐Allee 23Ulm89081Germany
| |
Collapse
|
9
|
Valade G, Libert N, Martinaud C, Vicaut E, Banzet S, Peltzer J. Therapeutic Potential of Mesenchymal Stromal Cell-Derived Extracellular Vesicles in the Prevention of Organ Injuries Induced by Traumatic Hemorrhagic Shock. Front Immunol 2021; 12:749659. [PMID: 34659252 PMCID: PMC8511792 DOI: 10.3389/fimmu.2021.749659] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/06/2021] [Indexed: 12/28/2022] Open
Abstract
Severe trauma is the principal cause of death among young people worldwide. Hemorrhagic shock is the leading cause of death after severe trauma. Traumatic hemorrhagic shock (THS) is a complex phenomenon associating an absolute hypovolemia secondary to a sudden and significant extravascular blood loss, tissue injury, and, eventually, hypoxemia. These phenomena are responsible of secondary injuries such as coagulopathy, endotheliopathy, microcirculation failure, inflammation, and immune activation. Collectively, these dysfunctions lead to secondary organ failures and multi-organ failure (MOF). The development of MOF after severe trauma is one of the leading causes of morbidity and mortality, where immunological dysfunction plays a central role. Damage-associated molecular patterns induce an early and exaggerated activation of innate immunity and a suppression of adaptive immunity. Severe complications are associated with a prolonged and dysregulated immune–inflammatory state. The current challenge in the management of THS patients is preventing organ injury, which currently has no etiological treatment available. Modulating the immune response is a potential therapeutic strategy for preventing the complications of THS. Mesenchymal stromal cells (MSCs) are multipotent cells found in a large number of adult tissues and used in clinical practice as therapeutic agents for immunomodulation and tissue repair. There is growing evidence that their efficiency is mainly attributed to the secretion of a wide range of bioactive molecules and extracellular vesicles (EVs). Indeed, different experimental studies revealed that MSC-derived EVs (MSC-EVs) could modulate local and systemic deleterious immune response. Therefore, these new cell-free therapeutic products, easily stored and available immediately, represent a tremendous opportunity in the emergency context of shock. In this review, the pathophysiological environment of THS and, in particular, the crosstalk between the immune system and organ function are described. The potential therapeutic benefits of MSCs or their EVs in treating THS are discussed based on the current knowledge. Understanding the key mechanisms of immune deregulation leading to organ damage is a crucial element in order to optimize the preparation of EVs and potentiate their therapeutic effect.
Collapse
Affiliation(s)
- Guillaume Valade
- Institut de Recherche Biomédicale des Armées (IRBA), Inserm UMRS-MD-1197, Clamart, France
| | - Nicolas Libert
- Service d'Anesthésie-Réanimation, Hôpital d'instruction des armées Percy, Clamart, France
| | - Christophe Martinaud
- Unité de Médicaments de Thérapie Innovante, Centre de Transfusion Sanguine des Armées, Clamart, France
| | - Eric Vicaut
- Laboratoire d'Etude de la Microcirculation, Université de Paris, UMRS 942 INSERM, Paris, France
| | - Sébastien Banzet
- Institut de Recherche Biomédicale des Armées (IRBA), Inserm UMRS-MD-1197, Clamart, France
| | - Juliette Peltzer
- Institut de Recherche Biomédicale des Armées (IRBA), Inserm UMRS-MD-1197, Clamart, France
| |
Collapse
|
10
|
Zhao B, Lu R, Chen J, Xie M, Zhao X, Kong L. S100A9 blockade prevents lipopolysaccharide-induced lung injury via suppressing the NLRP3 pathway. Respir Res 2021; 22:45. [PMID: 33549095 PMCID: PMC7866705 DOI: 10.1186/s12931-021-01641-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 01/27/2021] [Indexed: 02/08/2023] Open
Abstract
Background S100 calcium binding protein A9 (S100A9) is a pro-inflammatory alarmin associated with several inflammation-related diseases. However, the role of S100A9 in lung injury in sepsis has not been fully investigated. Therefore, the present study aimed to determine the role of S100A9 in a lipopolysaccharide (LPS)-induced lung injury murine model and its underlying molecular mechanisms. Methods LPS was utilized to induce sepsis and lung injury in C57BL/6 or NOD-like receptor family pyrin domain containing 3 (NLRP3)−/− mice. To investigate the effects of S100A9 blockade, mice were treated with a specific inhibitor of S100A9. Subsequently, lung injury and inflammation were evaluated by histology and enzyme‑linked immunosorbent assay (ELISA), respectively. Furthermore, western blot analysis and RT-qPCR were carried out to investigate the molecular mechanisms underlying the effects of S100A9. Results S100A9 was upregulated in the lung tissues of LPS-treated mice. However, inhibition of S100A9 alleviated LPS-induced lung injury. Additionally, S100A9 blockade also attenuated the inflammatory responses and apoptosis in the lungs of LPS-challenged mice. Furthermore, the increased expression of NLRP3 was also suppressed by S100A9 blockade, while S100A9 blockade had no effect on NLRP3−/− mice. In vitro, S100A9 downregulation mitigated LPS-induced inflammation. Interestingly, these effects were blunted by NLRP3 overexpression. Conclusion The results of the current study suggested that inhibition of S100A9 could protect against LPS-induced lung injury via inhibiting the NLRP3 pathway. Therefore, S100A9 blockade could be considered as a novel therapeutic strategy for lung injury in sepsis.
Collapse
Affiliation(s)
- Boying Zhao
- Department of Cardiothoracic Surgery, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University, No. 1 Jiangkang Road, Yuzhong, Chongqing, 400010, China.,Vascular Surgery Department, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Renfu Lu
- Department of Cardiothoracic Surgery, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University, No. 1 Jiangkang Road, Yuzhong, Chongqing, 400010, China
| | - Jianjun Chen
- Department of Cardiothoracic Surgery, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University, No. 1 Jiangkang Road, Yuzhong, Chongqing, 400010, China
| | - Ming Xie
- Department of Cardiothoracic Surgery, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University, No. 1 Jiangkang Road, Yuzhong, Chongqing, 400010, China
| | - Xingji Zhao
- Department of Cardiothoracic Surgery, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University, No. 1 Jiangkang Road, Yuzhong, Chongqing, 400010, China.
| | - Lingwen Kong
- Department of Cardiothoracic Surgery, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University, No. 1 Jiangkang Road, Yuzhong, Chongqing, 400010, China.
| |
Collapse
|
11
|
Club Cell Protein 16 Attenuates CD16 brightCD62 dim Immunosuppressive Neutrophils in Damaged Tissue upon Posttraumatic Sepsis-Induced Lung Injury. J Immunol Res 2021; 2021:6647753. [PMID: 33575362 PMCID: PMC7861919 DOI: 10.1155/2021/6647753] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 12/24/2020] [Accepted: 01/13/2021] [Indexed: 12/11/2022] Open
Abstract
Background Recently, identification of immunosuppressive polymorphonuclear leukocytes (PMNL) that were traditionally described as proinflammatory cells emerged in the field of posttraumatic immunity. To understand their local and remote distribution after trauma, PMNL-subsets and the impact of immunomodulatory Club Cell protein (CC)16 that correlates with pulmonary complications were assessed. Methods C57BL/6N mice were divided into three groups, receiving isolated blunt chest trauma (TxT), undergoing TxT followed by cecal ligation and puncture (CLP, TxT + CLP) after 24 h, or sham undergoing analgosedation (n = 18/group). Further, each group was subdivided into three groups receiving either no treatment (ctrl) or intratracheal neutralization of CC16 by application of anti-CC16-antibody or application of an unspecific IgG control antibody (n = 6/group). Treatment was set at the time point after TxT. Analyses followed 6 h post-CLP. PMNL were characterized via expression of CD11b, CD16, CD45, CD62L, and Ly6G by flow cytometry in bone marrow (BM), blood, spleen, lung, liver, and bronchoalveolar and peritoneal lavage fluid (BALF and PL). Apoptosis was assessed by activated (cleaved) caspase-3. Results from untreated ctrl and IgG-treated mice were statistically comparable between all corresponding sham, TxT, and TxT + CLP groups. Results Immature (CD16dimCD62Lbright) PMNL increased significantly in BM, circulation, and spleen after TxT vs. sham and were significantly attenuated in the lungs, BALF, PL, and liver. Classical-shaped (CD16brightCD62Lbright) PMNL increased after TxT vs. sham in peripheral tissue and were significantly attenuated in circulation, proposing a trauma-induced migration of mature or peripheral differentiation of circulating immature PMNL. Immunosuppressive (CD16brightCD62Ldim) PMNL decreased significantly in the lungs and spleen, while they systemically increased after TxT vs. sham. CLP in the TxT + CLP group reduced immunosuppressive PMNL in PL and increased their circulatory rate vs. isolated TxT, showing local reduction in affected tissue and their increase in nonaffected tissue. CC16 neutralization enhanced the fraction of immunosuppressive PMNL following TxT vs. sham and decreased caspase-3 in the lungs post-CLP in the TxT + CLP group, while apoptotic cells in the liver diminished post-TxT. Posttraumatic CC16 neutralization promotes the subset of immunosuppressive PMNL and antagonizes their posttraumatic distribution. Conclusion Since CC16 affects both the distribution of PMNL subsets and apoptosis in tissues after trauma, it may constitute as a novel target to beneficially shape the posttraumatic tissue microenvironment and homeostasis to improving outcomes.
Collapse
|
12
|
Tachino J, Katayama Y, Kitamura T, Kiyohara K, Nakao S, Umemura Y, Ishida K, Hirose T, Nakagawa Y, Shimazu T. Assessment of the interaction effect between injury regions in multiple injuries: A nationwide cohort study in Japan. J Trauma Acute Care Surg 2021; 90:185-190. [PMID: 33021602 PMCID: PMC7748042 DOI: 10.1097/ta.0000000000002969] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 09/28/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND There have been no clinical studies to sufficiently reveal the interaction effect generated by combinations of injury regions of multiple injuries. We hypothesized that certain combinations of trauma regions might lead to increased risk of traumatic death and aimed to verify this hypothesis using a nationwide trauma registry in Japan. MATERIALS AND METHODS This was a retrospective study of trauma patients registered in the Japan Trauma Data Bank between 2004 and 2017. We included patients who suffered blunt trauma with an Injury Severity Score of 16 or more. The trauma was classified into four regions (head, chest, abdomen, and extremities), and a multivariable logistic regression analysis was performed that included interaction terms derived from the combination of two regions as covariates. RESULTS We included 78,280 trauma patients in this study. Among them, 16,100 (20.6%) patients were discharged to death. Multivariable logistic regression showed the odds ratio (OR) of in-hospital death compared with patients without injury of an Abbreviated Injury Scale score of 3 or more in each injured region as follows: head score, 2.31 (95% confidence interval [CI], 2.13-2.51); chest score, 2.28 (95% CI, 2.17-2.39); abdomen score, 1.68 (95% CI, 1.56-1.82); and extremities score, 1.84 (95% CI, 1.76-1.93), respectively. In addition, the ORs of the statistically significant interaction terms were as follows: head-chest 1.29 (95% CI, 1.13-1.48), chest-abdomen 0.77 (95% CI, 0.67-0.88), chest-extremities 1.95 (95% CI, 1.77-2.14), and abdomen-extremities 0.70 (95% CI, 0.62-0.79), respectively. CONCLUSION In this population, among patients with multiple injuries, a combination of head-chest trauma and chest-extremities trauma was shown to increase the risk of traumatic death. LEVEL OF EVIDENCE Prognostic, Level III.
Collapse
|
13
|
Ozkaya S, Alhaja E, Karaman I, Erdem D. Pulmonary langerhans cell histiocytosis: Can it originate from chest trauma? ARCHIVES OF TRAUMA RESEARCH 2021. [DOI: 10.4103/atr.atr_85_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
14
|
Cursano S, Battaglia CR, Urrutia-Ruiz C, Grabrucker S, Schön M, Bockmann J, Braumüller S, Radermacher P, Roselli F, Huber-Lang M, Boeckers TM. A CRHR1 antagonist prevents synaptic loss and memory deficits in a trauma-induced delirium-like syndrome. Mol Psychiatry 2021; 26:3778-3794. [PMID: 32051550 PMCID: PMC8550963 DOI: 10.1038/s41380-020-0659-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 01/10/2020] [Accepted: 01/20/2020] [Indexed: 12/15/2022]
Abstract
Older patients with severe physical trauma are at high risk of developing neuropsychiatric syndromes with global impairment of cognition, attention, and consciousness. We employed a thoracic trauma (TxT) mouse model and thoroughly analyzed age-dependent spatial and temporal posttraumatic alterations in the central nervous system. Up to 5 days after trauma, we observed a transient 50% decrease in the number of excitatory synapses specifically in hippocampal pyramidal neurons accompanied by alterations in attention and motor activity and disruption of contextual memory consolidation. In parallel, hippocampal corticotropin-releasing hormone (CRH) expression was highly upregulated, and brain-derived neurotrophic factor (BDNF) levels were significantly reduced. In vitro experiments revealed that CRH application induced neuronal autophagy with rapid lysosomal degradation of BDNF via the NF-κB pathway. The subsequent synaptic loss was rescued by BDNF as well as by specific NF-κB and CRH receptor 1 (CRHR1) antagonists. In vivo, the chronic application of a CRHR1 antagonist after TxT resulted in reversal of the observed histological, molecular, and behavioral alterations. The data suggest that neuropsychiatric syndromes (i.e., delirium) after peripheral trauma might be at least in part due to the activation of the hippocampal CRH/NF-κB/BDNF pathway, which results in a dramatic loss of synaptic contacts. The successful rescue by stress hormone receptor antagonists should encourage clinical trials focusing on trauma-induced delirium and/or other posttraumatic syndromes.
Collapse
Affiliation(s)
- Silvia Cursano
- grid.6582.90000 0004 1936 9748Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany ,International Graduate School in Molecular Medicine, IGradU, 89081 Ulm, Germany
| | - Chiara R. Battaglia
- grid.6582.90000 0004 1936 9748Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany ,International Graduate School in Molecular Medicine, IGradU, 89081 Ulm, Germany
| | - Carolina Urrutia-Ruiz
- grid.6582.90000 0004 1936 9748Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Stefanie Grabrucker
- grid.10049.3c0000 0004 1936 9692Department of Biological Sciences, University of Limerick, Limerick, V94 PH61 Ireland
| | - Michael Schön
- grid.6582.90000 0004 1936 9748Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Jürgen Bockmann
- grid.6582.90000 0004 1936 9748Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Sonja Braumüller
- grid.6582.90000 0004 1936 9748Institute for Anesthesiological Pathophysiology, Ulm University, Helmholtzstr. 8/1, 89081 Ulm, Germany
| | - Peter Radermacher
- grid.6582.90000 0004 1936 9748Institute for Anesthesiological Pathophysiology, Ulm University, Helmholtzstr. 8/1, 89081 Ulm, Germany
| | - Francesco Roselli
- grid.6582.90000 0004 1936 9748Clinic for Neurology, Ulm University, 89081 Ulm, Germany
| | - Markus Huber-Lang
- grid.6582.90000 0004 1936 9748Institute of Clinical and Experimental Trauma-Immunology, Ulm University, 89081 Ulm, Germany
| | - Tobias M. Boeckers
- grid.6582.90000 0004 1936 9748Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| |
Collapse
|
15
|
Huber-Lang MS, Ignatius A, Köhl J, Mannes M, Braun CK. Complement in trauma-Traumatised complement? Br J Pharmacol 2020; 178:2863-2879. [PMID: 32880897 DOI: 10.1111/bph.15245] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/23/2020] [Accepted: 08/17/2020] [Indexed: 12/17/2022] Open
Abstract
Physical trauma represents a major global burden. The trauma-induced response, including activation of the innate immune system, strives for regeneration but can also lead to post-traumatic complications. The complement cascade is rapidly activated by damaged tissue, hypoxia, exogenous proteases and others. Activated complement can sense, mark and clear both damaged tissue and pathogens. However, excessive and insufficient activation of complement can result in a dysfunctional immune and organ response. Similar to acute coagulopathy, complementopathy can develop with enhanced anaphylatoxin generation and an impairment of complement effector functions. Various remote organ effects are induced or modulated by complement activation. Frequently, established trauma treatments are double-edged. On one hand, they help stabilising haemodynamics and oxygen supply as well as injured organs and on the other hand, they also drive complement activation. Immunomodulatory approaches aim to reset trauma-induced disbalance of complement activation and thus may change surgical trauma management procedures to improve outcome. LINKED ARTICLES: This article is part of a themed issue on Canonical and non-canonical functions of the complement system in health and disease. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.14/issuetoc.
Collapse
Affiliation(s)
- Markus S Huber-Lang
- Institute of Clinical and Experimental Trauma-Immunology, University Hospital of Ulm, Ulm, Germany
| | - Anita Ignatius
- Institue of Orthopaedic Research and Biomechanics, University Hospital of Ulm, Ulm, Germany
| | - Jörg Köhl
- Institute for Systemic Inflammatory Research, University of Lübeck, Lübeck, Germany.,Division of Immunobiology, Cincinnati Children's Hospital Medical Centre, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Marco Mannes
- Institute of Clinical and Experimental Trauma-Immunology, University Hospital of Ulm, Ulm, Germany
| | - Christian Karl Braun
- Institute of Clinical and Experimental Trauma-Immunology, University Hospital of Ulm, Ulm, Germany.,Department of Paediatrics and Adolescent Medicine, University Hospital of Ulm, Ulm, Germany
| |
Collapse
|
16
|
Lupu L, Palmer A, Huber-Lang M. Inflammation, Thrombosis, and Destruction: The Three-Headed Cerberus of Trauma- and SARS-CoV-2-Induced ARDS. Front Immunol 2020; 11:584514. [PMID: 33101314 PMCID: PMC7546394 DOI: 10.3389/fimmu.2020.584514] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 09/10/2020] [Indexed: 01/05/2023] Open
Abstract
Physical trauma can be considered an unrecognized "pandemic" because it can occur anywhere and affect anyone and represents a global burden. Following severe tissue trauma, patients frequently develop acute lung injury (ALI) and/or acute respiratory distress syndrome (ARDS) despite modern surgical and intensive care concepts. The underlying complex pathophysiology of life-threatening ALI/ARDS has been intensively studied in experimental and clinical settings. However, currently, the coronavirus family has become the focus of ALI/ARDS research because it represents an emerging global public health threat. The clinical presentation of the infection is highly heterogeneous, varying from a lack of symptoms to multiple organ dysfunction and mortality. In a particular subset of patients, the primary infection progresses rapidly to ALI and ARDS. The pathophysiological mechanisms triggering and driving severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-induced ALI/ARDS are still poorly understood. Although it is also generally unknown whether insights from trauma-induced ARDS may be readily translated to SARS-CoV-2-associated ARDS, it was still recommended to treat coronavirus-positive patients with ALI/ARDS with standard protocols for ALI/ARDS. However, this strategy was questioned by clinical scientists, because it was documented that some severely hypoxic SARS-CoV-2-infected patients exhibited a normal respiratory system compliance, a phenomenon rarely observed in ARDS patients with another underlying etiology. Therefore, coronavirus-induced ARDS was defined as a specific ARDS phenotype, which accordingly requires an adjusted therapeutic approach. These suggestions reflect previous attempts of classifying ARDS into different phenotypes that might overall facilitate ARDS diagnosis and treatment. Based on the clinical data from ARDS patients, two major phenotypes have been proposed: hyper- and hypo-inflammatory. Here, we provide a comparative review of the pathophysiological pathway of trauma-/hemorrhagic shock-induced ARDS and coronavirus-induced ARDS, with an emphasis on the crucial key points in the pathogenesis of both these ARDS forms. Therefore, the manifold available data on trauma-/hemorrhagic shock-induced ARDS may help to better understand coronavirus-induced ARDS.
Collapse
Affiliation(s)
| | | | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma-Immunology, University Hospital Ulm, Ulm, Germany
| |
Collapse
|
17
|
Xu P, Gärtner F, Gihring A, Liu C, Burster T, Wabitsch M, Knippschild U, Paschke S. Influence of obesity on remodeling of lung tissue and organization of extracellular matrix after blunt thorax trauma. Respir Res 2020; 21:238. [PMID: 32943048 PMCID: PMC7496205 DOI: 10.1186/s12931-020-01502-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 09/06/2020] [Indexed: 12/15/2022] Open
Abstract
Background Previously, it has been shown that obesity is a risk factor for recovery, regeneration, and tissue repair after blunt trauma and can affect the rate of muscle recovery and collagen deposition after trauma. To date, lung tissue regeneration and extracellular matrix regulation in obese mice after injury has not been investigated in detail yet. Methods This study uses an established blunt thorax trauma model to analyze morphological changes and alterations on gene and protein level in lean or obese (diet-induced obesity for 16 ± 1 week) male C57BL/6 J mice at various time-points after trauma induction (1 h, 6 h, 24 h, 72 h and 192 h). Results Morphological analysis after injury showed lung parenchyma damage at early time-points in both lean and obese mice. At later time-points a better regenerative capacity of lean mice was observed, since obese animals still exhibited alveoli collapse, wall thickness as well as remaining filled alveoli structures. Although lean mice showed significantly increased collagen and fibronectin gene levels, analysis of collagen deposition showed no difference based on colorimetric quantification of collagen and visual assessment of Sirius red staining. When investigating the organization of the ECM on gene level, a decreased response of obese mice after trauma regarding extracellular matrix composition and organization was detectable. Differences in the lung tissue between the diets regarding early responding MMPs (MMP8/9) and late responding MMPs (MMP2) could be observed on gene and protein level. Obese mice show differences in regulation of extracellular matrix components compared to normal weight mice, which results in a decreased total MMP activity in obese animals during the whole regeneration phase. Starting at 6 h post traumatic injury, lean mice show a 50% increase in total MMP activity compared to control animals, while MMP activity in obese mice drops to 50%. Conclusions In conclusion, abnormal regulation of the levels of extracellular matrix genes in the lung may contribute to an aberrant regeneration after trauma induction with a delay of repair and pathological changes of the lung tissue in obese mice.
Collapse
Affiliation(s)
- Pengfei Xu
- Department of General and Visceral Surgery, Surgery Center; Ulm University Medical Center, Albert-Einstein-Allee 23, 89081, Ulm, Germany
| | - Fabian Gärtner
- Department of General and Visceral Surgery, Surgery Center; Ulm University Medical Center, Albert-Einstein-Allee 23, 89081, Ulm, Germany
| | - Adrian Gihring
- Department of General and Visceral Surgery, Surgery Center; Ulm University Medical Center, Albert-Einstein-Allee 23, 89081, Ulm, Germany
| | - Congxing Liu
- Department of General and Visceral Surgery, Surgery Center; Ulm University Medical Center, Albert-Einstein-Allee 23, 89081, Ulm, Germany
| | - Timo Burster
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, Kabanbay Batyr Ave., 53, Nur-Sultan, 010000, Republic of Kazakhstan
| | - Martin Wabitsch
- Division of Pediatric Endocrinology and Diabetes, Ulm University Hospital for Pediatrics and Adolescent Medicine, Eythstraße 24, 89075, Ulm, Germany.
| | - Uwe Knippschild
- Department of General and Visceral Surgery, Surgery Center; Ulm University Medical Center, Albert-Einstein-Allee 23, 89081, Ulm, Germany.
| | - Stephan Paschke
- Department of General and Visceral Surgery, Surgery Center; Ulm University Medical Center, Albert-Einstein-Allee 23, 89081, Ulm, Germany
| |
Collapse
|
18
|
Relja B, Land WG. Damage-associated molecular patterns in trauma. Eur J Trauma Emerg Surg 2020; 46:751-775. [PMID: 31612270 PMCID: PMC7427761 DOI: 10.1007/s00068-019-01235-w] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 09/27/2019] [Indexed: 12/13/2022]
Abstract
In 1994, the "danger model" argued that adaptive immune responses are driven rather by molecules released upon tissue damage than by the recognition of "strange" molecules. Thus, an alternative to the "self versus non-self recognition model" has been provided. The model, which suggests that the immune system discriminates dangerous from safe molecules, has established the basis for the future designation of damage-associated molecular patterns (DAMPs), a term that was coined by Walter G. Land, Seong, and Matzinger. The pathological importance of DAMPs is barely somewhere else evident as in the posttraumatic or post-surgical inflammation and regeneration. Since DAMPs have been identified to trigger specific immune responses and inflammation, which is not necessarily detrimental but also regenerative, it still remains difficult to describe their "friend or foe" role in the posttraumatic immunogenicity and healing process. DAMPs can be used as biomarkers to indicate and/or to monitor a disease or injury severity, but they also may serve as clinically applicable parameters for optimized indication of the timing for, i.e., secondary surgeries. While experimental studies allow the detection of these biomarkers on different levels including cellular, tissue, and circulatory milieu, this is not always easily transferable to the human situation. Thus, in this review, we focus on the recent literature dealing with the pathophysiological importance of DAMPs after traumatic injury. Since dysregulated inflammation in traumatized patients always implies disturbed resolution of inflammation, so-called model of suppressing/inhibiting inducible DAMPs (SAMPs) will be very briefly introduced. Thus, an update on this topic in the field of trauma will be provided.
Collapse
Affiliation(s)
- Borna Relja
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto von Guericke University Magdeburg, Magdeburg, Germany.
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Frankfurt, Goethe University Frankfurt am Main, 60590, Frankfurt, Germany.
| | - Walter Gottlieb Land
- Molecular ImmunoRheumatology, INSERM UMR_S1109, Laboratory of Excellence Transplantex, University of Strasbourg, Strasbourg, France
| |
Collapse
|
19
|
Ming T, Yuan M, Kong Q, Huang Q, Xia Z, Wu X. Dexmedetomidine alleviates blunt chest trauma and hemorrhagic shock‑resuscitation‑induced acute lung injury through inhibiting the NLRP3 inflammasome. Mol Med Rep 2020; 22:2507-2515. [PMID: 32705267 PMCID: PMC7411430 DOI: 10.3892/mmr.2020.11335] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 06/12/2020] [Indexed: 12/11/2022] Open
Abstract
Blunt chest trauma with hemorrhagic shock frequently induces pulmonary inflammation that leads to acute lung injury (ALI). The present study aimed to explore the protective effects of dexmedetomidine (Dex) in blunt chest trauma and hemorrhagic shock-resuscitation (THSR)-induced ALI by mediating nucleotide binding and oligomerization domain-like receptor family pyrin domain-containing protein 3 (NLRP3) inflammasome formation in rats. An ALI model in rats induced by THSR was constructed and Dex was administered intraperitoneally (5 µg/kg/h) immediately after blunt chest trauma. Blood samples were collected for the determination of proinflammatory factor levels, and lung tissue specimens were harvested for wet/dry (W/D) weight ratio, hematoxylin and eosin staining, and transmission electron microscopy analyses. Additionally, malondialdehyde (MDA), superoxide dismutase (SOD), lactate dehydrogenase (LDH) and myeloperoxidase (MPO) activity were evaluated, and the expression of protein in lung tissues was examined via western blot analysis. Compared with the sham group, pathological alterations in the ALI group and the W/D ratios were significantly increased. MDA, LDH and MPO activity, and the levels of interleukin (IL)-1β, IL-18, IL-6 and tumor necrosis factor-α were significantly elevated. NLRP3, apoptosis-associated speck-like protein containing a caspase recruitment domain and caspase-1 expression was significantly increased. Conversely, Dex treatment significantly reversed these changes. The present study demonstrated that by reducing inflammatory responses, Dex exerted protective effects against THSR-ALI in rats, potentially via the inhibition of NLRP3 signaling pathways.
Collapse
Affiliation(s)
- Tingqian Ming
- Department of Anesthesiology, Renmin Hospital, Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Min Yuan
- Department of Anesthesiology, Renmin Hospital, Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Qian Kong
- Department of Anesthesiology, Renmin Hospital, Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Qin Huang
- Department of Anesthesiology, Renmin Hospital, Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Zhongyuan Xia
- Department of Anesthesiology, Renmin Hospital, Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Xiaojing Wu
- Department of Anesthesiology, Renmin Hospital, Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
20
|
Lin Q, Johns RA. Resistin family proteins in pulmonary diseases. Am J Physiol Lung Cell Mol Physiol 2020; 319:L422-L434. [PMID: 32692581 DOI: 10.1152/ajplung.00040.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The family of resistin-like molecules (RELMs) consists of four members in rodents (RELMα/FIZZ1/HIMF, RELMβ/FIZZ2, Resistin/FIZZ3, and RELMγ/FIZZ4) and two members in humans (Resistin and RELMβ), all of which exhibit inflammation-regulating, chemokine, and growth factor properties. The importance of these cytokines in many aspects of physiology and pathophysiology, especially in cardiothoracic diseases, is rapidly evolving in the literature. In this review article, we attempt to summarize the contribution of RELM signaling to the initiation and progression of lung diseases, such as pulmonary hypertension, asthma/allergic airway inflammation, chronic obstructive pulmonary disease, fibrosis, cancers, infection, and other acute lung injuries. The potential of RELMs to be used as biomarkers or risk predictors of these diseases also will be discussed. Better understanding of RELM signaling in the pathogenesis of pulmonary diseases may offer novel targets or approaches for the development of therapeutics to treat or prevent a variety of inflammation, tissue remodeling, and fibrosis-related disorders in respiratory, cardiovascular, and other systems.
Collapse
Affiliation(s)
- Qing Lin
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Roger A Johns
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
21
|
Mi YM, Qi Q, Zhang L, Wang XF, Chen ZM, Hua CZ. Assessment of serum sialic acid correlated with C3 in children with Mycoplasma pneumoniae pneumonia. J Clin Lab Anal 2020; 34:e23078. [PMID: 31907994 PMCID: PMC7083476 DOI: 10.1002/jcla.23078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 06/23/2019] [Accepted: 07/14/2019] [Indexed: 11/25/2022] Open
Abstract
Background Different from the diagnosis of bacterial infections, Mycoplasma pneumoniae pneumonia (MPP) is still lacking of convenient non‐specific laboratory parameters. Method A total of 125 children with MPP were included in the MPP group and 89 children with Mycoplasma‐negative pneumonia were included in the control group, and the sera were collected from the children at both the acute and recovery stages in the two groups. Results The sialic acid and C3 in the MPP group were significantly higher than those in the control group both at the acute and at the recovery stage. On the other hand, the sialic acid and C3 at the acute stage were significantly higher than those at the recovery stage in the MPP group. However, in the control group, the sialic acid and C3 demonstrated IgG exhibited no significant change between the acute stage and the recovery stage. Lastly, positive correlations between sialic acid level and C3 level were identified in the MPP group at both acute and recovery stages. Conclusion Our study demonstrated that the serum sialic acid correlated with C3 specifically increased in children with MPP, indicating that it might be the important non‐specific parameters in the diagnosis of MPP.
Collapse
Affiliation(s)
- Yu-Mei Mi
- Division of Infectious Disease, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Qi Qi
- Department of Infectious Disease, Hangzhou Red Cross Hospital, Hangzhou, Zhejiang, China
| | - Li Zhang
- Division of Infectious Disease, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Xiao-Fang Wang
- Department of Pediatrics, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Zhi-Min Chen
- Division of Respiration, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Chun-Zhen Hua
- Division of Infectious Disease, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| |
Collapse
|
22
|
Langgartner D, Palmer A, Rittlinger A, Reber SO, Huber-Lang M. Effects of Prior Psychosocial Trauma on Subsequent Immune Response After Experimental Thorax Trauma. Shock 2019; 49:690-697. [PMID: 28846569 DOI: 10.1097/shk.0000000000000973] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Overshooting inflammation during the early phase after blunt thorax trauma promotes the development of acute respiratory distress syndrome, multiple organ failure, and subsequent mortality. Given that individuals diagnosed with stress-related disorders are characterized by chronic low-grade inflammation, we hypothesize that "psychosocial traumatic preload" poses a risk factor for the abovementioned complications after thorax trauma.Here, we used the chronic subordinate colony housing (CSC) paradigm to induce "psychosocial traumatic preload" and systemic low-grade immune activation in male mice, indicated by elevated plasma concentrations of different inflammatory mediators. Subsequent thorax trauma was induced in anaesthetized mice by a single blast wave centered on the thorax; SHAM animals were exposed to anesthesia only. Mice were killed 2, 6, and 24 h after thorax trauma or SHAM treatment.Independent of thorax trauma, CSC caused an increase in adrenal weight, and a decrease in thymus weight, indicating that the stress paradigm worked reliably. Moreover, although lung histology was not affected by prior stress, CSC exposure aggravated the early immune response after thorax trauma, indicated by elevated myeloperoxidase lung concentrations in thorax trauma-exposed CSC versus respective single-housed control (SHC) mice (2 h). Furthermore, thorax trauma caused an increase in total bronchoalveolar lavage fluid (BAL) protein (24 h), BAL C5a (2 h), BAL cell counts (24 h), and BAL keratinocyte chemoattractant (6 h and 24 h) in CSC but not SHC mice.Our data indicate that repeated psychosocial traumatization during adulthood moderately aggravates the local immune response toward thorax trauma, but overall may be considered as a rather minor risk factor in terms of thorax trauma-associated complications.
Collapse
Affiliation(s)
- Dominik Langgartner
- Laboratory for Molecular Psychosomatics, Clinic for Psychosomatic Medicine and Psychotherapy, University Ulm, Ulm, Germany
| | - Annette Palmer
- Institute of Clinical and Experimental Trauma-Immunology, University Ulm, Ulm, Germany
| | - Anne Rittlinger
- Institute of Clinical and Experimental Trauma-Immunology, University Ulm, Ulm, Germany
| | - Stefan O Reber
- Laboratory for Molecular Psychosomatics, Clinic for Psychosomatic Medicine and Psychotherapy, University Ulm, Ulm, Germany
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma-Immunology, University Ulm, Ulm, Germany
| |
Collapse
|
23
|
Haider T, Simader E, Glück O, Ankersmit HJ, Heinz T, Hajdu S, Negrin LL. Systemic release of heat-shock protein 27 and 70 following severe trauma. Sci Rep 2019; 9:9595. [PMID: 31270381 PMCID: PMC6610099 DOI: 10.1038/s41598-019-46034-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 06/20/2019] [Indexed: 12/23/2022] Open
Abstract
Trauma represents a major cause of morbidity and mortality worldwide. The endogenous inflammatory response to trauma remains not fully elucidated. Pro-inflammation in the early phase is followed by immunosuppression leading to infections, multi-organ failure and mortality. Heat-shock proteins (HSPs) act as intracellular chaperons but exert also extracellular functions. However, their role in acute trauma remains unknown. The aim of this study was to evaluate serum concentrations of HSP 27 and HSP 70 in severely injured patients. We included severely injured patients with an injury severity score of at least 16 and measured serum concentration of both markers at admission and on day two. We found significantly increased serum concentrations of both HSP 27 and HSP 70 in severely injured patients. Concomitant thoracic trauma lead to a further increase of both HSPs. Also, elevated concentrations of HSP 27 and HSP 70 were associated with poor outcome in these patients. Standard laboratory parameters did not correlate with neither HSP 27, nor with HSP 70. Our findings demonstrate involvement of systemic release of HSP 27 and HSP 70 after severe trauma and their potential as biomarker in polytraumatized patients.
Collapse
Affiliation(s)
- Thomas Haider
- Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.
| | - Elisabeth Simader
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.,Department of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.,Austrian Research Promotion Agency FFG Projects 852748 and 862068 "APOSEC", Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Olaf Glück
- Department of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.,Austrian Research Promotion Agency FFG Projects 852748 and 862068 "APOSEC", Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Hendrik J Ankersmit
- Department of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.,Austrian Research Promotion Agency FFG Projects 852748 and 862068 "APOSEC", Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Thomas Heinz
- Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Stefan Hajdu
- Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Lukas L Negrin
- Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| |
Collapse
|
24
|
Haider T, Simader E, Hacker P, Ankersmit HJ, Heinz T, Hajdu S, Negrin LL. Increased serum concentrations of soluble ST2 are associated with pulmonary complications and mortality in polytraumatized patients. Clin Chem Lab Med 2019; 56:810-817. [PMID: 29341938 DOI: 10.1515/cclm-2017-0762] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2017] [Accepted: 11/14/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND We sought to evaluate the role of soluble ST2 (suppression of tumorigenicity) serum concentrations in polytraumatized patients and its potential role as biomarker for pulmonary complications. METHODS We included severely injured patients (injury severity score≥16) admitted to our level I trauma center and analyzed serum samples obtained on the day of admission and on day 2. Furthermore, patients with isolated thoracic injury and healthy probands were included and served as control groups. Serum samples were analyzed for soluble ST2 concentrations with a commercially available ELISA kit. RESULTS A total of 130 patients were included in the present study. Five patients with isolated thoracic injury and eight healthy probands were further included. Serum analyses revealed significantly elevated concentrations of soluble ST2 in polytraumatized patients compared to patients suffering from isolated thoracic trauma and healthy probands. In polytraumatized patients who developed pulmonary complications (acute respiratory distress syndrome and pneumonia) and in patients who died, significantly higher serum concentrations of soluble ST2 were found on day 2 (p<0.001). Serum concentrations of soluble ST2 on day 2 were of prognostic value to predict pulmonary complications in polytraumatized patients (area under the curve=0.720, 95% confidence interval=0.623-0.816). Concomitant thoracic trauma had no further impact on serum concentrations of soluble ST2. CONCLUSIONS Serum concentrations of soluble ST2 are upregulated following polytrauma. Increased concentrations were associated with worse outcome.
Collapse
Affiliation(s)
- Thomas Haider
- Department of Trauma Surgery, Medical University of Vienna, Vienna, Austria
| | - Elisabeth Simader
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Philipp Hacker
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Hendrik J Ankersmit
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Thomas Heinz
- Department of Trauma Surgery, Medical University of Vienna, Vienna, Austria
| | - Stefan Hajdu
- Department of Trauma Surgery, Medical University of Vienna, Vienna, Austria
| | - Lukas L Negrin
- Department of Trauma Surgery, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
25
|
Störmann P, Becker N, Künnemeyer L, Wutzler S, Vollrath JT, Lustenberger T, Hildebrand F, Marzi I, Relja B. Contributing factors in the development of acute lung injury in a murine double hit model. Eur J Trauma Emerg Surg 2019; 46:21-30. [PMID: 30937460 DOI: 10.1007/s00068-019-01121-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 03/27/2019] [Indexed: 02/07/2023]
Abstract
OBJECTIVES Blunt chest (thoracic) trauma (TxT) is known to contribute to the development of secondary pulmonary complications. Of these, acute lung injury (ALI) is common especially in multiply injured patients and might not only be due to the direct trauma itself, but seems to be caused by ongoing and multifactorial inflammatory changes. Nevertheless, the exact mechanisms and contributing factors of the development of ALI following blunt chest trauma are still elusive. METHODS 60 CL57BL/6N mice sustained either blunt chest trauma combined with laparotomy without further interventions or a double hit (DH) including TxT and cecal ligation puncture (CLP) after 24 h to induce ALI. Animals were killed either 6 or 24 h after the second procedure. Pulmonary expression of inflammatory mediators cxcl1, cxcl5, IL-1β and IL-6, neutrophil infiltration and lung tissue damage using the Lung Injury Score (LIS) were determined. RESULTS Next to a moderate increase in other inflammatory mediators, a significant increase in CXCL1, neutrophil infiltration and lung injury was observed early after TxT, which returned to baseline levels after 24 h. DH induced significantly increased gene expression of cxcl1, cxcl5, IL-1β and IL-6 after 6 h, which was followed by the postponed significant increase in the protein expression after 24 h compared to controls. Neutrophil infiltration was significantly enhanced 24 h after DH compared to all other groups, and exerted a slight decline after 24 h. LIS has shown a significant increase after both 6 and 24 h compared to both control groups as well the late TxT group. CONCLUSION Early observed lung injury with moderate inflammatory changes after blunt chest trauma recovered quickly, and therefore, may be caused by mechanical lung injury. In contrast, lung injury in the ALI group did not undergo recovery and is closely associated with significant changes of inflammatory mediators. This model may be used for further examinations of contributing factors and therapeutic strategies to prevent ALI.
Collapse
Affiliation(s)
- Philipp Störmann
- Department of Trauma, Hand and Reconstructive Surgery, Hospital of the Goethe University Frankfurt/Main, Theodor-Stern-Kai 7, 60590, Frankfurt/Main, Germany.
| | - Nils Becker
- Department of Trauma, Hand and Reconstructive Surgery, Hospital of the Goethe University Frankfurt/Main, Theodor-Stern-Kai 7, 60590, Frankfurt/Main, Germany
| | - Leander Künnemeyer
- Department of Trauma, Hand and Reconstructive Surgery, Hospital of the Goethe University Frankfurt/Main, Theodor-Stern-Kai 7, 60590, Frankfurt/Main, Germany
| | - Sebastian Wutzler
- Department of Trauma, Hand and Reconstructive Surgery, Hospital of the Goethe University Frankfurt/Main, Theodor-Stern-Kai 7, 60590, Frankfurt/Main, Germany.,Department of Trauma, Hand and Orthopedic Surgery, Helios Horst Schmidt Clinic, Wiesbaden, Germany
| | - Jan Tilmann Vollrath
- Department of Trauma, Hand and Reconstructive Surgery, Hospital of the Goethe University Frankfurt/Main, Theodor-Stern-Kai 7, 60590, Frankfurt/Main, Germany
| | - Thomas Lustenberger
- Department of Trauma, Hand and Reconstructive Surgery, Hospital of the Goethe University Frankfurt/Main, Theodor-Stern-Kai 7, 60590, Frankfurt/Main, Germany
| | | | - Ingo Marzi
- Department of Trauma, Hand and Reconstructive Surgery, Hospital of the Goethe University Frankfurt/Main, Theodor-Stern-Kai 7, 60590, Frankfurt/Main, Germany
| | - Borna Relja
- Department of Trauma, Hand and Reconstructive Surgery, Hospital of the Goethe University Frankfurt/Main, Theodor-Stern-Kai 7, 60590, Frankfurt/Main, Germany
| |
Collapse
|
26
|
Karasu E, Nilsson B, Köhl J, Lambris JD, Huber-Lang M. Targeting Complement Pathways in Polytrauma- and Sepsis-Induced Multiple-Organ Dysfunction. Front Immunol 2019; 10:543. [PMID: 30949180 PMCID: PMC6437067 DOI: 10.3389/fimmu.2019.00543] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 02/28/2019] [Indexed: 12/16/2022] Open
Abstract
Exposure to traumatic or infectious insults results in a rapid activation of the complement cascade as major fluid defense system of innate immunity. The complement system acts as a master alarm system during the molecular danger response after trauma and significantly contributes to the clearance of DAMPs and PAMPs. However, depending on the origin and extent of the damaged macro- and micro -milieu, the complement system can also be either excessively activated or inhibited. In both cases, this can lead to a maladaptive immune response and subsequent multiple cellular and organ dysfunction. The arsenal of complement-specific drugs offers promising strategies for various critical conditions after trauma, hemorrhagic shock, sepsis, and multiple organ failure. The imbalanced immune response needs to be detected in a rational and real-time manner before the translational therapeutic potential of these drugs can be fully utilized. Overall, the temporal-spatial complement response after tissue trauma and during sepsis remains somewhat enigmatic and demands a clinical triad: reliable tissue damage assessment, complement activation monitoring, and potent complement targeting to highly specific rebalance the fluid phase innate immune response.
Collapse
Affiliation(s)
- Ebru Karasu
- Institute for Clinical and Experimental Trauma-Immunology, University Hospital of Ulm, Ulm, Germany
| | - Bo Nilsson
- Department of Immunology, Genetics and Pathology (IGP), Laboratory C5:3, Uppsala University, Uppsala, Sweden
| | - Jörg Köhl
- Institute for Systemic Inflammation Research (ISEF), University of Lübeck, Lübeck, Germany.,Division of Immunobiology, Cincinnati Children's Hospital, Cincinnati, OH, United States
| | - John D Lambris
- Department of Pathology & Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, United States
| | - Markus Huber-Lang
- Institute for Clinical and Experimental Trauma-Immunology, University Hospital of Ulm, Ulm, Germany
| |
Collapse
|
27
|
Abstract
In most cases blunt chest trauma leads to fractures of the bony thorax, i. e. ribs. In the case of accompanying hemothorax or pneumothorax initial management consists of chest tube drainage by mini-thoracotomy. Subsequently patients with blunt chest trauma have to be transferred to the intensive care unit as these patients are at risk of pulmonary insufficiency or persistent blood loss via the chest tube. Injury to the great vessels or heart requires trauma care in specialized centers. Penetrating trauma is always surgically treated and the foreign body is removed in the operating room (OR). Life-threatening conditions, such as tension pneumothorax have to be treated by thorax drainage prior to hospital admission.
Collapse
|
28
|
Rogobete AF, Sandesc D, Bedreag OH, Papurica M, Popovici SE, Bratu T, Popoiu CM, Nitu R, Dragomir T, AAbed HIM, Ivan MV. MicroRNA Expression is Associated with Sepsis Disorders in Critically Ill Polytrauma Patients. Cells 2018; 7:E271. [PMID: 30551680 PMCID: PMC6316368 DOI: 10.3390/cells7120271] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 12/06/2018] [Accepted: 12/06/2018] [Indexed: 12/16/2022] Open
Abstract
A critically ill polytrauma patient is one of the most complex cases to be admitted to the intensive care unit, due to both the primary traumatic complications and the secondary post-traumatic interactions. From a molecular, genetic, and epigenetic point of view, numerous biochemical interactions are responsible for the deterioration of the clinical status of a patient, and increased mortality rates. From a molecular viewpoint, microRNAs are one of the most complex macromolecular systems due to the numerous modular reactions and interactions that they are involved in. Regarding the expression and activity of microRNAs in sepsis, their usefulness has reached new levels of significance. MicroRNAs can be used both as an early biomarker for sepsis, and as a therapeutic target because of their ability to block the complex reactions involved in the initiation, maintenance, and augmentation of the clinical status.
Collapse
Affiliation(s)
- Alexandru Florin Rogobete
- Faculty of Medicine, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania.
- Clinic of Anesthesia and Intensive Care, Emergency County Hospital "Pius Brinzeu", 300723 Timisoara, Romania.
| | - Dorel Sandesc
- Faculty of Medicine, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania.
- Clinic of Anesthesia and Intensive Care, Emergency County Hospital "Pius Brinzeu", 300723 Timisoara, Romania.
| | - Ovidiu Horea Bedreag
- Faculty of Medicine, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania.
- Clinic of Anesthesia and Intensive Care, Emergency County Hospital "Pius Brinzeu", 300723 Timisoara, Romania.
| | - Marius Papurica
- Faculty of Medicine, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania.
- Clinic of Anesthesia and Intensive Care, Emergency County Hospital "Pius Brinzeu", 300723 Timisoara, Romania.
| | - Sonia Elena Popovici
- Faculty of Medicine, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania.
| | - Tiberiu Bratu
- Faculty of Medicine, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania.
| | - Calin Marius Popoiu
- Faculty of Medicine, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania.
| | - Razvan Nitu
- Faculty of Medicine, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania.
| | - Tiberiu Dragomir
- Faculty of Medicine, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania.
| | - Hazzaa I M AAbed
- Faculty of Medicine, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania.
| | - Mihaela Viviana Ivan
- Faculty of Medicine, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania.
| |
Collapse
|
29
|
Abstract
This review summarizes a short list of currently discussed trauma-induced danger-associated molecular patterns (DAMP). Due to the bivalent character and often pleiotropic effects of a DAMP, it is difficult to describe its "friend or foe" role in post-traumatic inflammation and regeneration, both systemically as well locally in tissues. DAMP can be used as biomarkers to indicate or monitor disease or injury severity, but also may serve as clinically applicable parameters for better indication and timing of surgery. Due to the inflammatory processes at the local tissue level or the systemic level, the precise role of DAMP is not always clear to define. While in vitro and experimental studies allow for the detection of these biomarkers at the different levels of an organism-cellular, tissue, circulation-this is not always easily transferable to the human setting. Increased knowledge exploring the dual role of DAMP after trauma, and concentrating on their nuclear functions, transcriptional targets, release mechanisms, cellular sources, multiple functions, their interactions and potential therapeutic targeting is warranted.
Collapse
Affiliation(s)
- Borna Relja
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Frankfurt, Goethe University, 60590, Frankfurt, Germany.
| | - Katharina Mörs
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Frankfurt, Goethe University, 60590, Frankfurt, Germany
| | - Ingo Marzi
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Frankfurt, Goethe University, 60590, Frankfurt, Germany
| |
Collapse
|
30
|
Amann EM, Rojewski MT, Rodi S, Fürst D, Fiedler J, Palmer A, Braumüller S, Huber-Lang M, Schrezenmeier H, Brenner RE. Systemic recovery and therapeutic effects of transplanted allogenic and xenogenic mesenchymal stromal cells in a rat blunt chest trauma model. Cytotherapy 2017; 20:218-231. [PMID: 29223534 DOI: 10.1016/j.jcyt.2017.11.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 08/22/2017] [Accepted: 11/03/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND Effective therapy of Acute Lung Injury (ALI) is still a major scientific and clinical problem. To define novel therapeutic strategies for sequelae of blunt chest trauma (TxT) like ALI/Acute Respiratory Distress Syndrome, we have investigated the immunomodulatory and regenerative effects of a single dose of ex vivo expanded human or rat mesenchymal stromal cells (hMSCs/rMSCs) with or without priming, immediately after the induction of TxT in Wistar rats. METHODS We analyzed the histological score of lung injury, the cell count of the broncho alveolar lavage fluid (BAL), the change in local and systemic cytokine level and the recovery of the administered cells 24 h and 5 days post trauma. RESULTS The treatment with hMSCs reduced the injury score 24 h after trauma by at least 50% compared with TxT rats without MSCs. In general, TxT rats treated with hMSCs exhibited a lower level of pro-inflammatory cytokines (interleukin [IL]-1B, IL-6) and chemokines (C-X-C motif chemokine ligand 1 [CXCL1], C-C motif chemokine ligand 2 [CCL2]), but a higher tumor necrosis factor alpha induced protein 6 (TNFAIP6) level in the BAL compared with TxT rats after 24 h. Five days after trauma, cytokine levels and the distribution of inflammatory cells were similar to sham rats. In contrast, the treatment with rMSCs did not reveal such therapeutic effects on the injury score and cytokine levels, except for TNFAIP6 level. CONCLUSION TxT represents a suitable model to study effects of MSCs as an acute treatment strategy after trauma. However, the source of MSCs has to be carefully considered in the design of future studies.
Collapse
Affiliation(s)
- Elisa Maria Amann
- Institute of Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen and University Hospital Ulm, University of Ulm, Ulm, Germany; Institute of Transfusion Medicine, University of Ulm, Ulm, Germany
| | - Markus Thomas Rojewski
- Institute of Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen and University Hospital Ulm, University of Ulm, Ulm, Germany; Institute of Transfusion Medicine, University of Ulm, Ulm, Germany
| | - Sinja Rodi
- Institute of Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen and University Hospital Ulm, University of Ulm, Ulm, Germany
| | - Daniel Fürst
- Institute of Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen and University Hospital Ulm, University of Ulm, Ulm, Germany
| | - Jörg Fiedler
- Orthopedic Department, Division for Biochemistry of Joint and Connective Tissue Diseases, University of Ulm, Ulm, Germany
| | - Annette Palmer
- Institute for Clinical and Experimental Trauma-Immunology, University of Ulm, Ulm, Germany
| | - Sonja Braumüller
- Institute for Clinical and Experimental Trauma-Immunology, University of Ulm, Ulm, Germany
| | - Markus Huber-Lang
- Institute for Clinical and Experimental Trauma-Immunology, University of Ulm, Ulm, Germany
| | - Hubert Schrezenmeier
- Institute of Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen and University Hospital Ulm, University of Ulm, Ulm, Germany; Institute of Transfusion Medicine, University of Ulm, Ulm, Germany.
| | - Rolf Erwin Brenner
- Orthopedic Department, Division for Biochemistry of Joint and Connective Tissue Diseases, University of Ulm, Ulm, Germany
| |
Collapse
|
31
|
Russo A, Ranieri M, Di Mise A, Dossena S, Pellegrino T, Furia E, Nofziger C, Debellis L, Paulmichl M, Valenti G, Tamma G. Interleukin-13 increases pendrin abundance to the cell surface in bronchial NCI-H292 cells via Rho/actin signaling. Pflugers Arch 2017; 469:1163-1176. [PMID: 28378089 DOI: 10.1007/s00424-017-1970-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 03/20/2017] [Accepted: 03/22/2017] [Indexed: 11/28/2022]
Abstract
Interleukin-13 (IL13) is a major player in the development of airway hyperresponsiveness in several respiratory disorders. Emerging data suggest that an increased expression of pendrin in airway epithelia is associated with elevated airway hyperreactivity in asthma. Here, we investigate the effect of IL13 on pendrin localization and function using bronchiolar NCI-H292 cells. The data obtained revealed that IL13 increases the cell surface expression of pendrin. This effect was paralleled by a significant increase in the intracellular pH, possibly via indirect stimulation of NHE. IL13 effect on pendrin localization and intracellular pH was reversed by theophylline, a bronchodilator compound used to treat asthma. IL13 upregulated RhoA activity, a crucial protein controlling actin dynamics, via G-alpha-13. Specifically, IL13 stabilized actin cytoskeleton and promoted co-localization and a direct molecular interaction between pendrin and F-actin in the plasma membrane region. These effects were reversed following exposure of cells to theophylline. Selective inhibition of Rho kinase, a downstream effector of Rho, reduced the IL13-dependent cell surface expression of pendrin. Together, these data indicate that IL13 increases pendrin abundance to the cell surface via Rho/actin signaling, an effect reversed by theophylline.
Collapse
Affiliation(s)
- Annamaria Russo
- Department of Biosciences Biotechnologies and Biopharmaceutics, University of Bari, Via Orabona 4, 70125, Bari, Italy
| | - Marianna Ranieri
- Department of Biosciences Biotechnologies and Biopharmaceutics, University of Bari, Via Orabona 4, 70125, Bari, Italy.
| | - Annarita Di Mise
- Department of Biosciences Biotechnologies and Biopharmaceutics, University of Bari, Via Orabona 4, 70125, Bari, Italy
| | - Silvia Dossena
- Institute of Pharmacology and Toxicology, Paracelsus Medical University, Salzburg, Austria
| | - Tommaso Pellegrino
- Department of Biosciences Biotechnologies and Biopharmaceutics, University of Bari, Via Orabona 4, 70125, Bari, Italy
| | - Emilia Furia
- Department of Chemistry and Chemical Technologies, University of Calabria, Rende, Italy
| | - Charity Nofziger
- Institute of Pharmacology and Toxicology, Paracelsus Medical University, Salzburg, Austria
| | - Lucantonio Debellis
- Department of Biosciences Biotechnologies and Biopharmaceutics, University of Bari, Via Orabona 4, 70125, Bari, Italy
| | - Markus Paulmichl
- Institute of Pharmacology and Toxicology, Paracelsus Medical University, Salzburg, Austria
| | - Giovanna Valenti
- Department of Biosciences Biotechnologies and Biopharmaceutics, University of Bari, Via Orabona 4, 70125, Bari, Italy.,Istituto Nazionsale di Biostrutture e Biosistemi (I.N.B.B.), Rome, Italy.,Centre of Excellence Genomic and Proteomics GEBCA, University of Bari, Bari, Italy
| | - Grazia Tamma
- Department of Biosciences Biotechnologies and Biopharmaceutics, University of Bari, Via Orabona 4, 70125, Bari, Italy. .,Istituto Nazionsale di Biostrutture e Biosistemi (I.N.B.B.), Rome, Italy.
| |
Collapse
|
32
|
Horst K, Simon TP, Pfeifer R, Teuben M, Almahmoud K, Zhi Q, Santos SA, Wembers CC, Leonhardt S, Heussen N, Störmann P, Auner B, Relja B, Marzi I, Haug AT, van Griensven M, Kalbitz M, Huber-Lang M, Tolba R, Reiss LK, Uhlig S, Marx G, Pape HC, Hildebrand F. Characterization of blunt chest trauma in a long-term porcine model of severe multiple trauma. Sci Rep 2016; 6:39659. [PMID: 28000769 PMCID: PMC5175194 DOI: 10.1038/srep39659] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 11/24/2016] [Indexed: 12/20/2022] Open
Abstract
Chest trauma has a significant relevance on outcome after severe trauma. Clinically, impaired lung function typically occurs within 72 hours after trauma. However, the underlying pathophysiological mechanisms are still not fully elucidated. Therefore, we aimed to establish an experimental long-term model to investigate physiological, morphologic and inflammatory changes, after severe trauma. Male pigs (sus scrofa) sustained severe trauma (including unilateral chest trauma, femur fracture, liver laceration and hemorrhagic shock). Additionally, non-injured animals served as sham controls. Chest trauma resulted in severe lung damage on both CT and histological analyses. Furthermore, severe inflammation with a systemic increase of IL-6 (p = 0.0305) and a local increase of IL-8 in BAL (p = 0.0009) was observed. The pO2/FiO2 ratio in trauma animals decreased over the observation period (p < 0.0001) but not in the sham group (p = 0.2967). Electrical Impedance Tomography (EIT) revealed differences between the traumatized and healthy lung (p < 0.0001). In conclusion, a clinically relevant, long-term model of blunt chest trauma with concomitant injuries has been developed. This reproducible model allows to examine local and systemic consequences of trauma and is valid for investigation of potential diagnostic or therapeutic options. In this context, EIT might represent a radiation-free method for bedside diagnostics.
Collapse
Affiliation(s)
- K Horst
- Department of Orthopaedic Trauma, RWTH Aachen University, Germany.,Harald Tscherne Research Laboratory, RWTH Aachen University, Germany
| | - T P Simon
- Department of Intensive Care and Intermediate Care, RWTH Aachen University, Germany
| | - R Pfeifer
- Department of Orthopaedic Trauma, RWTH Aachen University, Germany.,Harald Tscherne Research Laboratory, RWTH Aachen University, Germany
| | - M Teuben
- Department of Orthopaedic Trauma, RWTH Aachen University, Germany.,Harald Tscherne Research Laboratory, RWTH Aachen University, Germany
| | - K Almahmoud
- Department of Orthopaedic Trauma, RWTH Aachen University, Germany.,Harald Tscherne Research Laboratory, RWTH Aachen University, Germany
| | - Q Zhi
- Harald Tscherne Research Laboratory, RWTH Aachen University, Germany
| | - S Aguiar Santos
- Chair for Medical Information Technology, Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - C Castelar Wembers
- Chair for Medical Information Technology, Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - S Leonhardt
- Chair for Medical Information Technology, Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - N Heussen
- Department of Medical Statistics, RWTH Aachen University, Germany.,Medical School, Sigmund Freud Private University, Vienna, Austria
| | - P Störmann
- Department of Trauma-, Hand- and Reconstructive Surgery, University of Frankfurt/Main, Germany
| | - B Auner
- Department of Trauma-, Hand- and Reconstructive Surgery, University of Frankfurt/Main, Germany
| | - B Relja
- Department of Trauma-, Hand- and Reconstructive Surgery, University of Frankfurt/Main, Germany
| | - I Marzi
- Department of Trauma-, Hand- and Reconstructive Surgery, University of Frankfurt/Main, Germany
| | - A T Haug
- Experimental Trauma Surgery, Department of Trauma Surgery, Klinikum rechts der Isar, Technical University of Munich, Germany
| | - M van Griensven
- Experimental Trauma Surgery, Department of Trauma Surgery, Klinikum rechts der Isar, Technical University of Munich, Germany
| | - M Kalbitz
- Department of Orthopedic Trauma, Hand-, Plastic-, and Reconstructive Surgery, University of Ulm, Germany
| | - M Huber-Lang
- Department of Orthopedic Trauma, Hand-, Plastic-, and Reconstructive Surgery, University of Ulm, Germany
| | - R Tolba
- Institute for Laboratory Animal Science and Experimental Surgery, RWTH Aachen University, Germany
| | - L K Reiss
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Germany
| | - S Uhlig
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Germany
| | - G Marx
- Department of Intensive Care and Intermediate Care, RWTH Aachen University, Germany
| | - H C Pape
- Department of Orthopaedic Trauma, RWTH Aachen University, Germany
| | - F Hildebrand
- Department of Orthopaedic Trauma, RWTH Aachen University, Germany
| |
Collapse
|
33
|
Qi Y. Clinical study on VATS combined mechanical ventilation treatment of ARDS secondary to severe chest trauma. Exp Ther Med 2016; 12:1034-1038. [PMID: 27446317 PMCID: PMC4950469 DOI: 10.3892/etm.2016.3355] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 05/17/2016] [Indexed: 12/25/2022] Open
Abstract
The aim of the study was to investigate the clinical effects of microinvasive video-assisted thoracoscopic surgery (VATS) combined with mechanical ventilation in the treatment of acute respiratory distress syndrome (ARDS) secondary to severe chest trauma. A total of 62 patients with ARDS secondary to severe chest trauma were divided into the observation and control groups. The patients in the observation groups were treated with VATS combined with early mechanical ventilation while patients in the control group were treated using routine open thoracotomy combined with early mechanical ventilation. Compared to the controls, the survival rate of the observation group was significantly higher. The average operation time of the observation group was significantly shorter than that of the control group, and the incidence of complications in the perioperative period of the observation group was significantly lower than that of the control group (p<0.05). The average application time of the observation group was significantly shorter than that of the control group, and the incidence of ventilator-associated complications was significantly lower than that of the control group (p<0.05). In conclusion, a reasonable understanding of the indications and contraindications of VATS, combined with early mechanical treatment significantly improved the success rate of the treatment of ARDS patients secondary to severe chest trauma and reduced the complications.
Collapse
Affiliation(s)
- Yongjun Qi
- Department of Thoracic Surgery, Beijing Mentougou Hospital, Beijing 102300, P.R. China
| |
Collapse
|