1
|
Verdonck P, Peters M, Stroobants T, Gillebeert J, Janssens E, Schnaubelt S, Yogeswaran SK, Lemoyne S, Wittock A, Sypré L, Robert D, Jorens PG, Brouwers D, Slootmans S, Monsieurs K. Effects of major trauma care organisation on mortality in a European level 1 trauma centre: A retrospective analysis of 2016-2023. Injury 2024; 55:112022. [PMID: 39549420 DOI: 10.1016/j.injury.2024.112022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 11/07/2024] [Accepted: 11/08/2024] [Indexed: 11/18/2024]
Abstract
INTRODUCTION The centralisation of care for trauma patients in trauma centres, alongside the creation of inclusive trauma networks, has proven to reduce mortality. In Europe, such structured trauma programs and trauma networks are in development. OBJECTIVE To describe the aetiology and evolution of in-hospital mortality in a developing European level 1 trauma centre, to determine the early effect of trauma care reorganisation on mortality and to identify the areas for future investments in trauma care. MATERIALS AND METHODS This retrospective analysis included the calculation of the standardised mortality ratio (SMR), the time to in-hospital death and the cause of in-hospital death of all primary major trauma admissions to the Antwerp University Hospital from 2016 to 2023. RESULTS A total of 1470 patients was included with a crude mortality of 16.4 %, a median Revised Injury Severity Classification II (RISC II) adjusted mortality of 1.47 %, and a SMR of 1.12. A limitation of care directive was registered for 18.1 % of the patients. The causes of in-hospital death were traumatic brain injury (TBI) in 60 %, haemorrhagic shock in 15 %, organ failure in 10 %, miscellaneous in 14 % and unknown in 1 %. Sixty percent died in the first 48 h of hospital admission (mainly due to TBI and haemorrhagic shock) and 27 % died after more than seven days (mainly due to organ failure and TBI). In 24 % of the deceased patients with severe TBI, a non-TBI related cause of death was found. Overall, the SMR showed a nonsignificant decreasing trend, with a significant decrease of the SMR in the highest risk group (RISCII > 75 %) and a nonsignificant increase in the lowest risk group (RISC II <15 %). CONCLUSION The standardised mortality ratio declined over a period of 8 years, even though the SMR increased nonsignificantly in the lowest risk-adjusted mortality group. Future analysis of this subgroup could clarify whether this trend is due to an increase of limitation of care directives and if these deaths could have been prevented with improved trauma care. There might be opportunities to increase the survival of patients with severe TBI who have a non-TBI cause of death.
Collapse
Affiliation(s)
- Philip Verdonck
- Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium; Emergency Department, Antwerp University Hospital, Drie Eikenstraat 655, 2650 Edegem, Belgium; Major Trauma Service, Antwerp University Hospital, Drie Eikenstraat 655, 2650 Edegem, Belgium.
| | - Matthew Peters
- Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium; Department of Anaesthesiology, Antwerp University Hospital, Drie Eikenstraat 655, 2650 Edegem, Belgium.
| | - Tom Stroobants
- Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium; Department of Anaesthesiology, Antwerp University Hospital, Drie Eikenstraat 655, 2650 Edegem, Belgium.
| | - Johan Gillebeert
- Emergency Department, Ziekenhuis aan de Stroom Cadix, Kempenstraat 100, 2030 Antwerp, Belgium.
| | - Eva Janssens
- Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium; Emergency Department, Antwerp University Hospital, Drie Eikenstraat 655, 2650 Edegem, Belgium.
| | - Sebastian Schnaubelt
- Emergency Department, Antwerp University Hospital, Drie Eikenstraat 655, 2650 Edegem, Belgium; Department of Emergency Medicine, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; Emergency Medical Service Vienna, Radetzkystraße 1, 1030 Vienna, Austria.
| | - Suresh Krishan Yogeswaran
- Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium; Major Trauma Service, Antwerp University Hospital, Drie Eikenstraat 655, 2650 Edegem, Belgium; Department of Thoracovascular surgery, Antwerp University Hospital, Drie Eikenstraat 655, 2650 Edegem, Belgium.
| | - Sabine Lemoyne
- Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium; Emergency Department, Antwerp University Hospital, Drie Eikenstraat 655, 2650 Edegem, Belgium.
| | - Anouk Wittock
- Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium; Department of Anaesthesiology, Antwerp University Hospital, Drie Eikenstraat 655, 2650 Edegem, Belgium.
| | - Lore Sypré
- Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium; Emergency Department, Antwerp University Hospital, Drie Eikenstraat 655, 2650 Edegem, Belgium
| | - Dominique Robert
- Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium; Department of Intensive care, Antwerp University Hospital, Drie Eikenstraat 655, 2650 Edegem, Belgium.
| | - Philippe G Jorens
- Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium; Department of Intensive care, Antwerp University Hospital, Drie Eikenstraat 655, 2650 Edegem, Belgium; Laboratory of Experimental Medicine and Pediatrics, Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium.
| | - Dennis Brouwers
- Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium; Major Trauma Service, Antwerp University Hospital, Drie Eikenstraat 655, 2650 Edegem, Belgium; Department of Orthopaedics and traumatology, Antwerp University Hospital, Drie Eikenstraat 655, 2650 Edegem, Belgium.
| | - Stijn Slootmans
- Major Trauma Service, Antwerp University Hospital, Drie Eikenstraat 655, 2650 Edegem, Belgium; Centre for Research and Innovation of Care, Department of Nursing and Midwifery Sciences, Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium.
| | - Koenraad Monsieurs
- Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium; Emergency Department, Antwerp University Hospital, Drie Eikenstraat 655, 2650 Edegem, Belgium.
| |
Collapse
|
2
|
Ting RS, Weaver NA, King KL, Way TL, Sarrami P, Daniel L, Dinh M, Nair P, Hsu J, D'Amours SK, Balogh ZJ. Epidemiology of postinjury multiple organ failure: a prospective multicenter observational study. Eur J Trauma Emerg Surg 2024; 50:3223-3231. [PMID: 39264428 PMCID: PMC11666632 DOI: 10.1007/s00068-024-02630-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 08/06/2024] [Indexed: 09/13/2024]
Abstract
PURPOSE Postinjury multiple organ failure (MOF) is the sequela to the disease of polytrauma. We aimed to describe the contemporary population-based epidemiology of MOF within a mature trauma system, to analyse the time taken for MOF to develop, and to evaluate the temporal patterns and contributions of the individual constituent organ failures. METHODS Prospective observational study conducted across five Level-1 trauma centers in New South Wales, Australia. Trauma patients at-risk of MOF (Denver > 3 from 48 h post-admission), aged > 16 years, ISS > 15, and who stayed in ICU for ≥ 48 h were eligible for inclusion. RESULTS From May 2018-February 2021, 600 at-risk polytrauma patients were prospectively enrolled (mean(SD)age = 49(21)years, males = 453/600(76%),median(IQR)ISS = 26(20,34)). MOF incidence was 136/600(23%) among at-risk patients, 142/6248(2%) among major trauma patients (ISS > 12 per Australian definition), and 0.8/100,000 in the general population. The mortality rate was 55/600(11%) in the overall study population, and 34/136(25%) in MOF patients. 82/136(60%) of MOF patients developed MOF on day-3. No patients developed MOF after day-13. Among MOF patients, 60/136(44%) had cardiac failures (mortality = 37%), 39/136(29%) had respiratory failures (mortality = 23%), 24/136(18%) had renal failures (mortality = 63%), and 12/136(9%) had hepatic failures (mortality = 50%). CONCLUSION Although a rare syndrome in the general population, MOF occurred in 23% of the most severely injured polytrauma patients. When compared to previous risk-matched cohorts, MOF become more common, but not more lethal, despite a decade older cohort. The heart has superseded the lungs as the most common organ to fail. Cardiac and respiratory failures occurred earlier and were associated with lower mortality than renal and hepatic failures.
Collapse
Affiliation(s)
- Ryan S Ting
- St George & Sutherland Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Natasha A Weaver
- University of Newcastle, Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Kate L King
- John Hunter Hospital, University of Newcastle, Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Teagan L Way
- John Hunter Hospital, University of Newcastle, Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Pooria Sarrami
- NSW Institute of Trauma and Injury Management, NSW Agency for Clinical Innovation and University of New South Wales, South West Sydney Clinical School, Sydney, NSW, Australia
| | - Lovana Daniel
- Westmead Hospital, University of New South Wales, South West Sydney Clinical School, Sydney, NSW, Australia
| | - Michael Dinh
- NSW Institute of Trauma and Injury Management, NSW Agency for Clinical Innovation and University of New South Wales, South West Sydney Clinical School, Sydney, NSW, Australia
| | - Priya Nair
- St Vincent's Hospital, Sydney, Australia
| | - Jeremy Hsu
- Westmead Hospital, Sydney, NSW, Australia
| | - Scott K D'Amours
- Liverpool Hospital Trauma and Acute Care Surgery Unit, University of New South Wales, South West Sydney Clinical School, Sydney, NSW, Australia
| | - Zsolt J Balogh
- John Hunter Hospital, University of Newcastle, Hunter Medical Research Institute, Newcastle, NSW, Australia.
- Department of Traumatology, Division of Surgery, John Hunter Hospital, Hunter Region Mail Centre, University of Newcastle, Locked Bag 1, Newcastle, NSW, 2310, Australia.
| |
Collapse
|
3
|
Yang Z, Cancio TS, Willis RP, Young MD, Kneifel DM, Salinas J, Meyer AD. An early HMGB1 rise 12 hours before creatinine predicts acute kidney injury and multiple organ failure in a smoke inhalation and burn swine model. Front Immunol 2024; 15:1447597. [PMID: 39534595 PMCID: PMC11554498 DOI: 10.3389/fimmu.2024.1447597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 09/26/2024] [Indexed: 11/16/2024] Open
Abstract
Background Acute kidney injury (AKI) and multiple organ failure (MOF) are leading causes of mortality in trauma injuries. Early diagnosis of AKI and MOF is vital to improve outcomes, but current diagnostic criteria rely on laboratory markers that are delayed or unreliable. In this study, we investigated whether damage associated molecular patterns such as high-mobility group box 1 (HMGB1), syndecan-1 (SDC-1) and C3a correlate with the development of trauma-induced AKI and MOF. Methods Thirty-nine swine underwent smoke inhalation and severe burns, then received critical care for 72 hours or until death. AKI was defined by the KDIGO (Kidney Disease: Improving Global Outcomes) criteria, which labels AKI when a 1.5-fold increase in blood creatinine levels from baseline or a urine output < 0.5 mL/kg/h for 6 hours or more occurs. MOF was defined by the presence of both AKI and acute respiratory distress syndrome (PaO2/FiO2<300 for 4 hours). Results Eight of 39 pigs developed AKI and seven of those developed MOF. Pathological analysis revealed that polytrauma induces significantly higher kidney injury scores compared to sham controls. The average time from injury to KDIGO AKI was 24 hours (interquartile range: 22.50-32.25). Twelve hours after injury, HMGB1 levels were significantly increased in animals that went on to develop AKI compared to those that did not (73.07 ± 18.66 ng/mL vs. 31.64 ± 4.15 ng/mL, p<0.01), as well as in animals that developed MOF compared to those that did not (81.52±19.68 ng/mL vs. 31.19 ± 3.972 ng/mL, p<0.05). SDC-1 and C3a levels were not significantly different at any time point between groups. ROC analysis revealed that HMGB1 levels at 12 hours post-injury were predictive of both AKI and MOF development (AKI: AUROC=0.81, cut-off value=36.41 ng/mL; MOF: AUROC=0.89, cut-off value=36.41 ng/mL). Spearman's correlation revealed that HMGB1 levels at 12 hours correlated with multiple parameters of AKI, including blood urea nitrogen, blood creatinine, and blood myoglobin. Conclusion Twelve-hour post-injury HMGB1 levels predict AKI and MOF in a smoke inhalation and burn swine model. Further research is needed to validate this result in other polytrauma models and in critical combat causalities.
Collapse
Affiliation(s)
- Zhangsheng Yang
- Organ Support and Automation Technologies, United States Army Institute of Surgical Research, Fort Sam Houston, TX, United States
| | - Tomas S. Cancio
- Organ Support and Automation Technologies, United States Army Institute of Surgical Research, Fort Sam Houston, TX, United States
| | - Robert P. Willis
- Organ Support and Automation Technologies, United States Army Institute of Surgical Research, Fort Sam Houston, TX, United States
| | - Matthew D. Young
- Organ Support and Automation Technologies, United States Army Institute of Surgical Research, Fort Sam Houston, TX, United States
| | - Dustin M. Kneifel
- Organ Support and Automation Technologies, United States Army Institute of Surgical Research, Fort Sam Houston, TX, United States
| | - Jose Salinas
- Organ Support and Automation Technologies, United States Army Institute of Surgical Research, Fort Sam Houston, TX, United States
| | - Andrew D. Meyer
- Organ Support and Automation Technologies, United States Army Institute of Surgical Research, Fort Sam Houston, TX, United States
- Long School of Medicine, University of Texas Health Science Center, San Antonio, TX, United States
| |
Collapse
|
4
|
Duran I, Banerjee A, Flaherty PJ, Que YA, Ryan CM, Rahme LG, Tsurumi A. Development of a biomarker prediction model for post-trauma multiple organ failure/dysfunction syndrome based on the blood transcriptome. Ann Intensive Care 2024; 14:134. [PMID: 39198331 PMCID: PMC11358370 DOI: 10.1186/s13613-024-01364-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 08/09/2024] [Indexed: 09/01/2024] Open
Abstract
BACKGROUND Multiple organ failure/dysfunction syndrome (MOF/MODS) is a major cause of mortality and morbidity among severe trauma patients. Current clinical practices entail monitoring physiological measurements and applying clinical score systems to diagnose its onset. Instead, we aimed to develop an early prediction model for MOF outcome evaluated soon after traumatic injury by performing machine learning analysis of genome-wide transcriptome data from blood samples drawn within 24 h of traumatic injury. We then compared its performance to baseline injury severity scores and detection of infections. METHODS Buffy coat transcriptome and linked clinical datasets from blunt trauma patients from the Inflammation and the Host Response to Injury Study ("Glue Grant") multi-center cohort were used. According to the inclusion/exclusion criteria, 141 adult (age ≥ 16 years old) blunt trauma patients (excluding penetrating) with early buffy coat (≤ 24 h since trauma injury) samples were analyzed, with 58 MOF-cases and 83 non-cases. We applied the Least Absolute Shrinkage and Selection Operator (LASSO) and eXtreme Gradient Boosting (XGBoost) algorithms to select features and develop models for MOF early outcome prediction. RESULTS The LASSO model included 18 transcripts (AUROC [95% CI]: 0.938 [0.890-0.987] (training) and 0.833 [0.699-0.967] (test)), and the XGBoost model included 41 transcripts (0.999 [0.997-1.000] (training) and 0.907 [0.816-0.998] (test)). There were 16 overlapping transcripts comparing the two panels (0.935 [0.884-0.985] (training) and 0.836 [0.703-0.968] (test)). The biomarker models notably outperformed models based on injury severity scores and sex, which we found to be significantly associated with MOF (APACHEII + sex-0.649 [0.537-0.762] (training) and 0.493 [0.301-0.685] (test); ISS + sex-0.630 [0.516-0.744] (training) and 0.482 [0.293-0.670] (test); NISS + sex-0.651 [0.540-0.763] (training) and 0.525 [0.335-0.714] (test)). CONCLUSIONS The accurate assessment of MOF from blood samples immediately after trauma is expected to aid in improving clinical decision-making and may contribute to reduced morbidity, mortality and healthcare costs. Moreover, understanding the molecular mechanisms involving the transcripts identified as important for MOF prediction may eventually aid in developing novel interventions.
Collapse
Affiliation(s)
- Ivan Duran
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, 50 Blossom St., Their 340, Boston, MA, 02114, USA
| | - Ankita Banerjee
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, 50 Blossom St., Their 340, Boston, MA, 02114, USA
| | - Patrick J Flaherty
- Department of Mathematics and Statistics, University of Massachusetts at Amherst, Amherst, MA, 01003, USA
| | - Yok-Ai Que
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Colleen M Ryan
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, 50 Blossom St., Their 340, Boston, MA, 02114, USA
- Shriners Hospitals for Children-Boston®, 51 Blossom St., Boston, MA, 02114, USA
| | - Laurence G Rahme
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, 50 Blossom St., Their 340, Boston, MA, 02114, USA
- Shriners Hospitals for Children-Boston®, 51 Blossom St., Boston, MA, 02114, USA
- Department of Microbiology and Immunology, Harvard Medical School, 77 Ave. Louis Pasteur, Boston, MA, 02115, USA
| | - Amy Tsurumi
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, 50 Blossom St., Their 340, Boston, MA, 02114, USA.
- Shriners Hospitals for Children-Boston®, 51 Blossom St., Boston, MA, 02114, USA.
| |
Collapse
|
5
|
Tyagi D, Ting RS, Balogh ZJ. Postinjury multiple organ failure: Proposal of the rare syndrome approach. Injury 2024; 55:111599. [PMID: 38876542 DOI: 10.1016/j.injury.2024.111599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 04/30/2024] [Indexed: 06/16/2024]
Affiliation(s)
- Daksh Tyagi
- School of Medicine and Public Health, University of Newcastle, Newcastle, New South Wales, Australia
| | - Ryan S Ting
- St George & Sutherland Clinical School, University of New South Wales, Sydney, New South Wales, Australia
| | - Zsolt J Balogh
- John Hunter Hospital and University of Newcastle, Hunter Medical Research Institute, Newcastle, New South Wales, Australia.
| |
Collapse
|
6
|
Ting RS, King KL, Lewis DP, Weaver NA, Balogh ZJ. Modifiability of surgical timing in postinjury multiple organ failure patients. World J Surg 2024; 48:350-360. [PMID: 38686758 DOI: 10.1002/wjs.12076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 12/27/2023] [Indexed: 05/02/2024]
Abstract
BACKGROUND Postinjury multiple organ failure (MOF) is the leading cause of late trauma deaths, with primarily non-modifiable risk factors. Timing of surgery as a potentially modifiable risk factor is frequently proposed, but has not been quantified. We aimed to compare mortality, hospital length of stay (LOS), and ICU LOS between MOF patients who had surgery that preceded MOF with modifiable timings versus those with non-modifiable timings. METHODS Retrospective analysis of an ongoing 17-year prospective cohort study of ICU polytrauma patients at-risk of MOF. Among MOF patients (Denver score>3), we identified patients who had surgery that preceded MOF, determined whether the timing of these operation(s) were modifiable(M) or non-modifiable (non-M), and evaluated the change in physiological parameters as a result of surgery. RESULTS Of 716 polytrauma patients at-risk of MOF, 205/716 (29%) developed MOF, and 161/205 (79%) had surgery during their ICU admission. Of the surgical MOF patients, 147/161 (91%) had one or more operation(s) that preceded MOF, and 65/161 (40%) of them had operation(s) with modifiable timings. There were no differences in age (mean (SD) 52 (19) vs 53 (21)years), injury severity score (median (IQR) 34 (26-41)vs34 (25-44)), admission physiological and resuscitation parameters, between M and non-M-patients. M patients had longer ICU LOS (median (IQR) 18 (12-28)versus 11 (8-16)days, p < 0.0001) than non-M-patients, without difference in mortality (14%vs16%, p = 0.7347), or hospital LOS (median (IQR) 32 (18-52)vs27 (17-47)days, p = 0.3418). M-patients had less fluids and transfusions intraoperatively. Surgery did not compromise patient physiology. CONCLUSION Operations preceding MOF are common in polytrauma and seem to be safe in maintaining physiology. The margin for improvement from optimizing surgical timing is modest, contrary to historical assumptions.
Collapse
Affiliation(s)
- Ryan S Ting
- St George & Sutherland Clinical School, University of New South Wales, Sydney, New South Wales, Australia
| | - Kate L King
- John Hunter Hospital and University of Newcastle, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Daniel P Lewis
- John Hunter Hospital and University of Newcastle, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Natasha A Weaver
- University of Newcastle, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Zsolt J Balogh
- John Hunter Hospital and University of Newcastle, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| |
Collapse
|
7
|
Barea-Mendoza JA, Chico-Fernández M, Serviá-Goixart L, Quintana-Díaz M, García-Sáez I, Ballesteros-Sanz MÁ, Iglesias-Santiago A, Molina-Díaz I, González-Robledo J, Fernández-Cuervo A, Pérez-Bárcena J, Llompart-Pou JA. Associated Risk Factors and Impact in Clinical Outcomes of Multiorgan Failure in Patients with TBI. Neurocrit Care 2023; 39:411-418. [PMID: 36869209 DOI: 10.1007/s12028-023-01698-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 02/10/2023] [Indexed: 03/05/2023]
Abstract
BACKGROUND Individual extracerebral organ dysfunction is common after severe traumatic brain injury (TBI) and impacts outcomes. However, multiorgan failure (MOF) has received less attention in patients with isolated TBI. Our objective was to analyze the risk factors associated with the development of MOF and its impact in clinical outcomes in patients with TBI. METHODS This was an observational, prospective, multicenter study using data from a nationwide registry that currently includes 52 intensive care units (ICUs) in Spain (RETRAUCI). Isolated significant TBI was defined as Abbreviated Injury Scale (AIS) ≥ 3 in the head area with no AIS ≥ 3 in any other anatomical area. Multiorgan failure was defined using the Sequential-related Organ Failure Assessment as the alteration of two or more organs with a score of ≥ 3. We analyzed the contribution of MOF to crude and adjusted mortality (age and AIS head) by using logistic regression analysis. A multiple logistic regression analysis was performed to analyze the risk factors associated with the development of MOF in patients with isolated TBI. RESULTS A total of 9790 patients with trauma were admitted to the participating ICUs. Of them, 2964 (30.2%) had AIS head ≥ 3 and no AIS ≥ 3 in any other anatomical area, and these patients constituted the study cohort. Mean age was 54.7 (19.5) years, 76% of patients were men, and ground-level falls were the main mechanism of injury (49.1%). In-hospital mortality was 22.2%. Up to 185 patients with TBI (6.2%) developed MOF during their ICU stay. Crude and adjusted (age and AIS head) mortality was higher in patients who developed MOF (odds ratio 6.28 [95% confidence interval 4.58-8.60] and odds ratio 5.20 [95% confidence interval 3.53-7.45]), respectively. The logistic regression analysis showed that age, hemodynamic instability, the need of packed red blood cells concentrates in the initial 24 h, the severity of brain injury, and the need for invasive neuromonitoring were significantly associated with MOF development. CONCLUSIONS MOF occurred in 6.2% of patients with TBI admitted to the ICU and was associated with increased mortality. MOF was associated with age, hemodynamic instability, the need of packed red blood cells concentrates in the initial 24 h, the severity of brain injury, and the need for invasive neuromonitoring.
Collapse
Affiliation(s)
| | - Mario Chico-Fernández
- UCI Trauma y Emergencias, Servicio de Medicina Intensiva, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Lluís Serviá-Goixart
- Servei de Medicina Intensiva, Hospital Universitari Arnau de Vilanova, Institut de Recerca Biomèdica de Lleida, Universitat de Lleida, Lleida, Spain
| | | | - Iker García-Sáez
- Servicio de Medicina Intensiva, Hospital Universitario de Donostia, Donostia, Spain
| | | | - Alberto Iglesias-Santiago
- Servicio de Medicina Intensiva, Hospital Universitario Virgen de las Nieves, Instituto de Investigación Biosanitaria, Granada, Spain
| | - Ismael Molina-Díaz
- Servicio de Medicina Intensiva, Hospital Universitario Nuestra Señora de la Candelaria, Santa Cruz de Tenerife, Spain
| | - Javier González-Robledo
- Servicio de Medicina Intensiva, Complejo Asistencial Universitario de Salamanca, Salamanca, Spain
| | - Ana Fernández-Cuervo
- Servicio de Medicina Intensiva, Hospital Universitario Puerta del Mar, Cádiz, Spain
| | - Jon Pérez-Bárcena
- Servei de Medicina Intensiva, Hospital Universitari Son Espases, Institut d'Investigació Sanitària Illes Balears, Carretera Valldemossa, 79, 07120, Palma, Spain
| | - Juan Antonio Llompart-Pou
- Servei de Medicina Intensiva, Hospital Universitari Son Espases, Institut d'Investigació Sanitària Illes Balears, Carretera Valldemossa, 79, 07120, Palma, Spain.
| |
Collapse
|
8
|
Breslin JW. Edema and lymphatic clearance: molecular mechanisms and ongoing challenges. Clin Sci (Lond) 2023; 137:1451-1476. [PMID: 37732545 PMCID: PMC11025659 DOI: 10.1042/cs20220314] [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: 06/03/2023] [Revised: 08/18/2023] [Accepted: 08/31/2023] [Indexed: 09/22/2023]
Abstract
Resolution of edema remains a significant clinical challenge. Conditions such as traumatic shock, sepsis, or diabetes often involve microvascular hyperpermeability, which leads to tissue and organ dysfunction. Lymphatic insufficiency due to genetic causes, surgical removal of lymph nodes, or infections, leads to varying degrees of tissue swelling that impair mobility and immune defenses. Treatment options are limited to management of edema as there are no specific therapeutics that have demonstrated significant success for ameliorating microvascular leakage or impaired lymphatic function. This review examines current knowledge about the physiological, cellular, and molecular mechanisms that control microvascular permeability and lymphatic clearance, the respective processes for interstitial fluid formation and removal. Clinical conditions featuring edema, along with potential future directions are discussed.
Collapse
Affiliation(s)
- Jerome W Breslin
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, FL, U.S.A
| |
Collapse
|
9
|
Patel NM, Collotta D, Aimaretti E, Ferreira Alves G, Kröller S, Coldewey SM, Collino M, Thiemermann C. Inhibition of the JAK/STAT Pathway With Baricitinib Reduces the Multiple Organ Dysfunction Caused by Hemorrhagic Shock in Rats. Ann Surg 2023; 278:e137-e146. [PMID: 35837955 PMCID: PMC10249600 DOI: 10.1097/sla.0000000000005571] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE The aim of this study was to investigate (a) the effects of the Janus kinase (JAK)/signal transducer and activator of transcription (STAT) pathway inhibitor (baricitinib) on the multiple organ dysfunction syndrome (MODS) in a rat model of hemorrhagic shock (HS) and (b) whether treatment with baricitinib attenuates the activation of JAK/STAT, NF-κB, and NLRP3 caused by HS. BACKGROUND Posttraumatic MODS, which is in part due to excessive systemic inflammation, is associated with high morbidity and mortality. The JAK/STAT pathway is a regulator of numerous growth factor and cytokine receptors and, hence, is considered a potential master regulator of many inflammatory signaling processes. However, its role in trauma-hemorrhage is unknown. METHODS An acute HS rat model was performed to determine the effect of baricitinib on MODS. The activation of JAK/STAT, NF-κB, and NLRP3 pathways were analyzed by western blotting in the kidney and liver. RESULTS We demonstrate here for the first time that treatment with baricitinib (during resuscitation following severe hemorrhage) attenuates the organ injury and dysfunction and the activation of JAK/STAT, NF-κB, and NLRP3 pathways caused by HS in the rat. CONCLUSIONS Our results point to a role of the JAK/STAT pathway in the pathophysiology of the organ injury and dysfunction caused by trauma/hemorrhage and indicate that JAK inhibitors, such as baricitinib, may be repurposed for the treatment of the MODS after trauma and/or hemorrhage.
Collapse
Affiliation(s)
- Nikita M. Patel
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Debora Collotta
- Department of Neurosciences “Rita Levi Montalcini,” University of Turin, Turin, Italy
| | - Eleonora Aimaretti
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | | | - Sarah Kröller
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany
- Septomics Research Center, Jena University Hospital, Jena, Germany
| | - Sina M. Coldewey
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany
- Septomics Research Center, Jena University Hospital, Jena, Germany
| | - Massimo Collino
- Department of Neurosciences “Rita Levi Montalcini,” University of Turin, Turin, Italy
| | - Christoph Thiemermann
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
10
|
Dexmedetomidine improves acute lung injury by activating autophagy in a rat hemorrhagic shock and resuscitation model. Sci Rep 2023; 13:4374. [PMID: 36927753 PMCID: PMC10020563 DOI: 10.1038/s41598-023-31483-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 03/13/2023] [Indexed: 03/18/2023] Open
Abstract
Dexmedetomidine (DEX) can reduce lung injury in a hemorrhagic shock (HS) resuscitation (HSR) model in rats by inhibiting inflammation. Here, we aimed to investigate if these effects of DEX are due to autophagy activation. Therefore, we established HSR rat models and divided them into four groups. HS was induced using a blood draw. The rats were then resuscitated by reinjecting the drawn blood and saline. The rats were sacrificed 24 h after resuscitation. Lung tissues were harvested for histopathological examination, determination of wet/dry lung weight ratio, and detection of the levels of autophagy-related marker proteins LC3, P62, Beclin-1, and the ATG12-ATG5 conjugate. The morphological findings of hematoxylin and eosin staining in lung tissues and the pulmonary wet/dry weight ratio showed that lung injury improved in HSR + DEX rats. However, chloroquine (CQ), an autophagy inhibitor, abolished this effect. Detecting the concentration of autophagy-related proteins showed that DEX administration increased LC3, ATG12-ATG5, and Beclin-1 expression and decreased P62 expression. The expression levels of these proteins were similar to those in the HSR group after CQ + DEX administration. In summary, DEX induced autophagic activation in an HSR model. These findings suggest that DEX administration partially ameliorates HSR-induced lung injury via autophagic activation.
Collapse
|
11
|
Inhibition of Bruton's Tyrosine Kinase Activity Attenuates Hemorrhagic Shock-Induced Multiple Organ Dysfunction in Rats. Ann Surg 2023; 277:e624-e633. [PMID: 35129479 PMCID: PMC9891276 DOI: 10.1097/sla.0000000000005357] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE The aim of this study was to investigate (a) the potential of the Bruton's tyrosine kinase (BTK) inhibitors acalabrutinib and fenebrutinib to reduce multiple organ dysfunction syndrome (MODS) in acute (short-term and long-term follow-up) hemorrhagic shock (HS) rat models and (b) whether treatment with either acalabrutinib or fenebrutinib attenuates BTK, NF-κB and NLRP3 activation in HS. BACKGROUND The MODS caused by an excessive systemic inflammatory response following trauma is associated with a high morbidity and mortality. The protein BTK is known to play a role in the activation of the NLRP3 inflammasome, which is a key component of the innate inflammatory response. However, its role in trauma-hemorrhage is unknown. METHODS Acute HS rat models were performed to determine the influence of acalabrutinib or fenebrutinib on MODS. The activation of BTK, NF-κB and NLRP3 pathways were analyzed by western blot in the kidney. RESULTS We demonstrated that (a) HS caused organ injury and/or dysfunction and hypotension (post-resuscitation) in rats, while (b) treatment of HS-rats with either acalabrutinib or fenebrutinib attenuated the organ injury and dysfunction in acute HS models and (c) reduced the activation of BTK, NF- kB and NLRP3 pathways in the kidney. CONCLUSION Our results point to a role of BTK in the pathophysiology of organ injury and dysfunction caused by trauma/hemorrhage and indicate that BTK inhibitors may be repurposed as a potential therapeutic approach for MODS after trauma and/or hemorrhage.
Collapse
|
12
|
Campwala I, Guyette FX, Brown JB, Yazer MH, Daley BJ, Miller RS, Harbrecht BG, Claridge JA, Phelan HA, Eastridge B, Nirula R, Vercruysse GA, O'Keeffe T, Joseph B, Neal MD, Zuckerbraun BS, Sperry JL. Evaluation of critical care burden following traumatic injury from two randomized controlled trials. Sci Rep 2023; 13:1106. [PMID: 36670216 PMCID: PMC9860020 DOI: 10.1038/s41598-023-28422-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 01/18/2023] [Indexed: 01/22/2023] Open
Abstract
Trauma resuscitation practices have continued to improve with new advances targeting prehospital interventions. The critical care burden associated with severely injured patients at risk of hemorrhage has been poorly characterized. We aim to describe the individual and additive effects of multiorgan failure (MOF) and nosocomial infection (NI) on delayed mortality and resource utilization. A secondary analysis of harmonized data from two large prehospital randomized controlled trials (Prehospital Air Medical Plasma (PAMPer) Trial and Study of Tranexamic Acid during Air and Ground Medical Prehospital Transport (STAAMP) Trial) was conducted. Only those patients who survived beyond the first 24 hours post-injury and spent at least one day in the ICU were included. Patients were stratified by development of MOF only, NI only, both, or neither and diagnosis of early (≤ 3 days) versus late MOF (> 3 days). Risk factors of NI and MOF, time course of these ICU complications, associated mortality, and hospital resource utilization were evaluated. Of the 869 patients who were enrolled in PAMPer and STAAMP and who met study criteria, 27.4% developed MOF only (n = 238), 10.9% developed NI only (n = 95), and 15.3% were diagnosed with both MOF and NI (n = 133). Patients developing NI and/or MOF compared to those who had an uncomplicated ICU course had greater injury severity, lower GCS, and greater shock indexes. Early MOF occurred in isolation, while late MOF more often followed NI. MOF was associated with 65% higher independent risk of 30-day mortality when adjusting for cofounders (OR 1.65; 95% CI 1.04-2.6; p = 0.03), however NI did not significantly affect odds of mortality. NI was individually associated with longer mechanical ventilation, ICU stay, hospital stay, and rehabilitation requirements, and the addition of MOF further increased the burden of inpatient and post-discharge care. MOF and NI remain common complications for those who survive traumatic injury. MOF is a robust independent predictor of mortality following injury in this cohort, and NI is associated with higher resource utilization. Timing of these ICU complications may reveal differences in pathophysiology and offer targets for continued advancements in treatment.
Collapse
Affiliation(s)
- Insiyah Campwala
- Division of Trauma and General Surgery, Department of Surgery, University of Pittsburgh, 200 Lothrop St., Pittsburgh, PA, 15213, USA
| | - Francis X Guyette
- Department of Emergency Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Joshua B Brown
- Division of Trauma and General Surgery, Department of Surgery, University of Pittsburgh, 200 Lothrop St., Pittsburgh, PA, 15213, USA
| | - Mark H Yazer
- The Institute for Transfusion Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Brian J Daley
- Department of Surgery, University of Tennessee Health Science Center, Knoxville, TN, USA
| | | | - Brian G Harbrecht
- Department of Surgery, University of Louisville, Louisville, KY, USA
| | - Jeffrey A Claridge
- Department of Surgery, Metro Health Medical Center, Case Western Reserve University, Cleveland, OH, USA
| | - Herbert A Phelan
- Department of Surgery, University of Texas Southwestern, Dallas, TX, USA
| | - Brian Eastridge
- Department of Surgery, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Raminder Nirula
- Department of Surgery, University of Utah, Salt Lake City, UT, USA
| | | | | | - Bellal Joseph
- Department of Surgery, University of Arizona, Tucson, AZ, USA
| | - Matthew D Neal
- Division of Trauma and General Surgery, Department of Surgery, University of Pittsburgh, 200 Lothrop St., Pittsburgh, PA, 15213, USA
| | - Brian S Zuckerbraun
- Division of Trauma and General Surgery, Department of Surgery, University of Pittsburgh, 200 Lothrop St., Pittsburgh, PA, 15213, USA
| | - Jason L Sperry
- Division of Trauma and General Surgery, Department of Surgery, University of Pittsburgh, 200 Lothrop St., Pittsburgh, PA, 15213, USA.
| |
Collapse
|
13
|
Senda A, Kojima M, Watanabe A, Kobayashi T, Morishita K, Aiboshi J, Otomo Y. Profiles of lipid, protein and microRNA expression in exosomes derived from intestinal epithelial cells after ischemia-reperfusion injury in a cellular hypoxia model. PLoS One 2023; 18:e0283702. [PMID: 36989330 PMCID: PMC10058167 DOI: 10.1371/journal.pone.0283702] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 03/14/2023] [Indexed: 03/30/2023] Open
Abstract
Intestinal ischemia-reperfusion injury leads to proinflammatory responses via gut-derived mediators, and accumulating evidence suggests that exosomes secreted by intestinal epithelial cells are involved in the development of systemic inflammation. Studies have reported changes in protein, lipid, and microRNA (miRNA) expression; however, considering the different experimental conditions, information on the relationships among these biomolecules remains insufficient. The aim of this study was to elucidate the multiple changes that simultaneously occur in exosomes after ischemic stimulation. Here, differentiated human intestinal Caco-2 cells were exposed to 95% air (normoxia group) or 5% O2 (hypoxia group) for 6 h. Cells in each group were subsequently incubated for 24 h in an atmosphere of 5% CO2 plus 95% air. The conditioned medium of each group was collected for isolating intestinal epithelial cell-derived exosomes. Together with proteome analyses, lipid analyses, and miRNA quantification, biological functional assays were performed using monocytic NF-κB reporter cells. Lipid metabolism-related protein expression was upregulated, miRNA levels were slightly altered, and unsaturated fatty acid-containing lysophosphatidylcholine concentration increased after hypoxia and reoxygenation injury; this suggested that the changes in exosomal components associated with ischemia-reperfusion injury activates inflammation, including the NF-κB pathway. This study elucidated the multiple changes that co-occur in exosomes after ischemic stimulation and partially clarified the mechanism underlying exosome-mediated inflammation after intestinal ischemic recanalization.
Collapse
Affiliation(s)
- Atsushi Senda
- Department of Acute Critical Care and Disaster Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Mitsuaki Kojima
- Department of Acute Critical Care and Disaster Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
- Emergency and Critical Care Center, Tokyo Women's Medical University Adachi Medical Center, Adachi-ku, Tokyo, Japan
| | - Arisa Watanabe
- Department of Biological Sciences, Graduate School of Humanities and Sciences, Ochanomizu University, Bunkyo-ku, Tokyo, Japan
| | - Tetsuyuki Kobayashi
- Department of Biological Sciences, Graduate School of Humanities and Sciences, Ochanomizu University, Bunkyo-ku, Tokyo, Japan
| | - Koji Morishita
- Department of Acute Critical Care and Disaster Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Junichi Aiboshi
- Department of Emergency and Critical Care Medicine, Tokyo Women's Medical University Yachiyo Medical Center, Yachiyo, Chiba, Japan
| | - Yasuhiro Otomo
- Department of Acute Critical Care and Disaster Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
14
|
Yang Y, Chen J, Yi C, Yang F, Tang M, Li Z, Bai X. Assessment of serum interleukin-28 as a biomarker to predict mortality in traumatic patients with sepsis. Cytokine 2022; 157:155959. [PMID: 35816926 DOI: 10.1016/j.cyto.2022.155959] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 05/11/2022] [Accepted: 07/01/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND Serious trauma due to various factors is a major global public issue, and sepsis is a major cause of trauma-associated mortality. Timely diagnosis and suitable treatment of post-traumatic sepsis are crucial to improve the hospital outcome of traumatic patients. IL-28 is a newly discovered member of IFN-λ family with multiple functions in inflammatory response. To date, its role in the pathogenic mechanisms of post-traumatic sepsis still remains unknown. METHODS In total, 20 healthy controls, 55 traumatic patients without sepsis and 54 traumatic patients with sepsis were enrolled in this study. Serum IL-28A/B levels were investigated by ELISA. RESULTS IL-28A/B levels were significantly increased in traumatic patients compared to healthy volunteers. Moreover, septic trauma patients displayed a significant increase in IL-28A/B levels compared with non-septic patients. In septic patients, IL-28A/B were negatively correlated with IFN-γ, IL-5, IL-13 and IL-17, and positively associated with IL-10. Moreover, IL-28A (AUC: 0.821, 95 %CI: 0.693-0.949) and IL-28B (AUC: 0.811, 95 %CI: 0.691-0.931) were both beneficial to predict increased mortality risk in septic trauma patients, though there was no statistical difference in the predictive value between them. CONCLUSIONS Early serum levels of IL-28A/B were associated with the development of post-trauma sepsis and could be applied to assess the outcome of traumatic patients with sepsis. Thus, IL-28 may be a potential indicator for post-traumatic sepsis.
Collapse
Affiliation(s)
- Yang Yang
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital, Tongji Medicine College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiajun Chen
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital, Tongji Medicine College, Huazhong University of Science and Technology, Wuhan, China
| | - Chengla Yi
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital, Tongji Medicine College, Huazhong University of Science and Technology, Wuhan, China
| | - Fan Yang
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital, Tongji Medicine College, Huazhong University of Science and Technology, Wuhan, China
| | - Manli Tang
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital, Tongji Medicine College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhanfei Li
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital, Tongji Medicine College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangjun Bai
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital, Tongji Medicine College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
15
|
Pape HC, Moore EE, McKinley T, Sauaia A. Pathophysiology in patients with polytrauma. Injury 2022; 53:2400-2412. [PMID: 35577600 DOI: 10.1016/j.injury.2022.04.009] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 04/13/2022] [Indexed: 02/02/2023]
Abstract
The pathophysiology after polytrauma represents a complex network of interactions. While it was thought for a long time that the direct and indirect effects of hypoperfusion are most relevant due to the endothelial permeability changes, it was discovered that the innate immune response to trauma is equally important in modifying the organ response. Recent multi center studies provided a "genetic storm" theory, according to which certain neutrophil changes are activated at the time of injury. However, a second hit phenomenon can be induced by activation of certain molecules by direct organ injury, or pathogens (damage associated molecular patterns, DAMPS - pathogen associated molecular patterns, PAMPS). The interactions between the four pathogenetic cycles (of shock, coagulopathy, temperature loss and soft tissue injuries) and cross-talk between coagulation and inflammation have also been identified as important modifiers of the clinical status. In a similar fashion, overzealous surgeries and their associated soft tissue injury and blood loss can induce secondary worsening of the patient condition. Therefore, staged surgeries in certain indications represent an important alternative, to allow for performing a "safe definitive surgery" strategy for major fractures. The current review summarizes all these situations in a detailed fashion.
Collapse
Affiliation(s)
- H-C Pape
- Department of Trauma, University Hospital Zurich, University of Zurich, Raemistrasse 100, 8091 Zurich, Switzerland.
| | - E E Moore
- Department of Surgery, Ernest E Moore Shock Trauma Center at Denver Health, University of Colorado, Aurora, CO, USA.
| | - T McKinley
- Department of Orthopaedics, Indiana University, 200 Hawkins Dr, Iowa City, IA 52242, USA.
| | - A Sauaia
- Schools of Public Health and Medicine, University of Colorado, Aurora, Colorado, USA.
| |
Collapse
|
16
|
Walsh SA, Davis TA. Key early proinflammatory signaling molecules encapsulated within circulating exosomes following traumatic injury. J Inflamm (Lond) 2022; 19:6. [PMID: 35551611 PMCID: PMC9097360 DOI: 10.1186/s12950-022-00303-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 03/26/2022] [Indexed: 12/28/2022] Open
Abstract
Background Assessment of immune status in critically ill patients is often based on serial tracking of systemic cytokine levels and clinical laboratory values. Exosomes are extracellular vesicles that can be secreted and internalized by cells to transport important cellular cargo in the regulation of numerous physiological and pathological processes. Here, we characterize the early compartmentalization profile of key proinflammatory mediators in serum exosomes in the steady state and following trauma. Adult male Sprague-Dawley rats (91 including naïve) were divided into one of four traumatic injury model groups incorporating whole-body blast, fracture, soft-tissue crush injury, tourniquet-induced ischemia, and limb amputation. Serum was collected at 1, 3, 6, and 24 h, and 3- and 7-day post-injury. Electrochemiluminescence-based immunoassays for 9 key proinflammatory mediators in whole serum, isolated serum exosomes, and exosome depleted serum were analyzed and compared between naïve and injured rats. Serum clinical chemistry analysis was performed to determine pathological changes. Results In naïve animals, substantial amounts of IL-1β, IL-10, and TNF-α were encapsulated, IL-6 was completely encapsulated, and CXCL1 freely circulating. One hour after blast injury alone, levels of exosome encapsulated IFN-γ, IL-10, IL-6, IL-13, IL-4, and TNF-α increased, whereas freely circulating and membrane-associated levels remained undetectable or low. Rats with the most severe polytraumatic injuries with end organ complications had the earliest rise and most pronounced concentration of IL-1β, IL-10, TNF-α, and IL-6 across all serum compartments. Moreover, CXCL1 levels increased in relation to injury severity, but remained almost entirely freely circulating at all timepoints. Conclusion These findings highlight that conventional ELISA-based assessments, which detect only free circulating and exosome membrane-bound mediators, underestimate the full immunoinflammatory response to trauma. Inclusion of exosome encapsulated mediators may be a better, more accurate and clinically useful early strategy to identify, diagnose, and monitor patients at highest risk for post-traumatic inflammation-associated complications.
Collapse
Affiliation(s)
- Sarah A Walsh
- Department of Surgery, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Thomas A Davis
- Department of Surgery, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.
| |
Collapse
|
17
|
Patel NM, Yamada N, Oliveira FRMB, Stiehler L, Zechendorf E, Hinkelmann D, Kraemer S, Stoppe C, Collino M, Collotta D, Alves GF, Ramos HP, Sordi R, Marzi I, Relja B, Marx G, Martin L, Thiemermann C. Inhibition of Macrophage Migration Inhibitory Factor Activity Attenuates Haemorrhagic Shock-Induced Multiple Organ Dysfunction in Rats. Front Immunol 2022; 13:886421. [PMID: 35464452 PMCID: PMC9019168 DOI: 10.3389/fimmu.2022.886421] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 03/14/2022] [Indexed: 12/13/2022] Open
Abstract
Objective The aim of this study was to investigate (a) macrophage migration inhibitory factor (MIF) levels in polytrauma patients and rats after haemorrhagic shock (HS), (b) the potential of the MIF inhibitor ISO-1 to reduce multiple organ dysfunction syndrome (MODS) in acute (short-term and long-term follow-up) HS rat models and (c) whether treatment with ISO-1 attenuates NF-κB and NLRP3 activation in HS. Background The MODS caused by an excessive systemic inflammatory response following trauma is associated with a high morbidity and mortality. MIF is a pleiotropic cytokine which can modulate the inflammatory response, however, its role in trauma is unknown. Methods The MIF levels in plasma of polytrauma patients and serum of rats with HS were measured by ELISA. Acute HS rat models were performed to determine the influence of ISO-1 on MODS. The activation of NF-κB and NLRP3 pathways were analysed by western blot in the kidney and liver. Results We demonstrated that (a) MIF levels are increased in polytrauma patients on arrival to the emergency room and in rats after HS, (b) HS caused organ injury and/or dysfunction and hypotension (post-resuscitation) in rats, while (c) treatment of HS-rats with ISO-1 attenuated the organ injury and dysfunction in acute HS models and (d) reduced the activation of NF-κB and NLRP3 pathways in the kidney and liver. Conclusion Our results point to a role of MIF in the pathophysiology of trauma-induced organ injury and dysfunction and indicate that MIF inhibitors may be used as a potential therapeutic approach for MODS after trauma and/or haemorrhage.
Collapse
Affiliation(s)
- Nikita M Patel
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Noriaki Yamada
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom.,Gifu University Graduate School of Medicine, Department of Emergency and Disaster Medicine Gifu University Hospital Advanced Critical Care Center, Gifu, Japan
| | - Filipe R M B Oliveira
- Department of Pharmacology, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Lara Stiehler
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom.,Department of Intensive Care and Intermediate Care, University Hospital RWTH Aachen, Aachen, Germany
| | - Elisabeth Zechendorf
- Department of Intensive Care and Intermediate Care, University Hospital RWTH Aachen, Aachen, Germany
| | - Daniel Hinkelmann
- Department of Intensive Care and Intermediate Care, University Hospital RWTH Aachen, Aachen, Germany
| | - Sandra Kraemer
- Department of Intensive Care and Intermediate Care, University Hospital RWTH Aachen, Aachen, Germany
| | - Christian Stoppe
- Department of Anesthesiology & Intensive Care Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Massimo Collino
- Department of Neurosciences "Rita Levi Montalcini", University of Turin, Turin, Italy
| | - Debora Collotta
- Department of Neurosciences "Rita Levi Montalcini", University of Turin, Turin, Italy
| | | | - Hanna Pillmann Ramos
- Department of Pharmacology, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Regina Sordi
- Department of Pharmacology, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Ingo Marzi
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| | - Borna Relja
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Frankfurt, Goethe University, Frankfurt, Germany.,Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto-von-Guericke University, Magdeburg, Germany
| | - Gernot Marx
- Department of Intensive Care and Intermediate Care, University Hospital RWTH Aachen, Aachen, Germany
| | - Lukas Martin
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom.,Department of Intensive Care and Intermediate Care, University Hospital RWTH Aachen, Aachen, Germany
| | - Christoph Thiemermann
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
18
|
Ballesteros MÁ, Sánchez‐Arguiano MJ, Chico‐Fernández M, Barea‐Mendoza JA, Serviá‐Goixart L, Sánchez‐Casado M, García Sáez I, Pino‐Sánchez FI, Antonio Llompart‐Pou J, Miñambres E. Chronic critical illness in polytrauma. Results of the Spanish trauma in ICU registry. Acta Anaesthesiol Scand 2022; 66:722-730. [PMID: 35332519 DOI: 10.1111/aas.14065] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 02/11/2022] [Accepted: 03/08/2022] [Indexed: 11/29/2022]
Abstract
PURPOSE Chronic critical illness after trauma injury has not been fully evaluated, and there is little evidence in this regard. We aim to describe the prevalence and risk factors of chronic critical illness (CCI) in trauma patients admitted to the intensive care unit. MATERIAL AND METHODS Retrospective observational multicenter study (Spanish Registry of Trauma in ICU (RETRAUCI)). Period March 2015 to December 2019. Trauma patients admitted to the ICU, who survived the first 48 h, were included. Chronic critical illness (CCI) was considered as the need for mechanical ventilation for a period greater than 14 days and/or placement of a tracheostomy. The main outcomes measures were prevalence and risk factors of CCI after trauma. RESULTS 1290/9213 (14%) patients developed CCI. These patients were older (51.2 ± 19.4 vs 49 ± 18.9); p < .01) and predominantly male (79.9%). They presented a higher proportion of infectious complications (81.3% vs 12.7%; p < .01) and multiple organ dysfunction syndrome (MODS) (27.02% vs 5.19%; p < .01). CCI patients required longer stays in the ICU and had higher ICU and overall in-hospital mortality. Age, injury severity score, head injury, infectious complications, and development of MODS were independent predictors of CCI. CONCLUSION CCI in trauma is a prevalent entity in our series. Early identification could facilitate specific interventions to change the trajectory of this process.
Collapse
Affiliation(s)
| | | | - Mario Chico‐Fernández
- UCI de Trauma y Emergencias Servicio de Medicina Intensiva, Hospital Universitario Madrid Spain
| | | | - Luis Serviá‐Goixart
- Servicio de Medicina Intensiva Hospital Universitario Arnau de Vilanova Lleida Spain
| | | | - Iker García Sáez
- Servicio de Medicina Intensiva Hospital Universitario Donostia Donostia‐San Sebastian Spain
| | | | - Juan Antonio Llompart‐Pou
- Servei de Medicina Intensiva Hospital Universitari Son Espases, Institut d'Investigació Sanitària Illes Balears (IdISBa) Palma Spain
| | - Eduardo Miñambres
- Transplant Coordination Unit & Service of Intensive Care University Hospital Marqués de Valdecilla‐IDIVAL School of Medicine University of Cantabria Santander Spain
| | | |
Collapse
|
19
|
Bayburina GA, Nurgaleeva EA, Samigullina AF, Farshatova ER, Ganeev TI, Agletdinov EFA, Tarasova TV. Antioxidant Activity Of Rat Liver With A Low Resistance To Hypoxia After Systemic Ischemia Reperfusion. RUSSIAN OPEN MEDICAL JOURNAL 2021. [DOI: 10.15275/rusomj.2021.0427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Objective — To assess the antioxidant activity of rat liver after systemic ischemia reperfusion (IRP). Material and Methods — The study was conducted on 70 male rats. For all animals of the treatment group (n=35) under ether anesthesia, we were stopping stopping systemic circulation for five minutes. After that, the animals were given an external cardiac massage and artificial lung ventilation. We did not perform circulatory arrest after ether anesthesia in animals of the control group (n=35). In all animals, we were measuring the levels of serum hormones (corticosterone, aldosterone), the content of glucocorticoid and mineralocorticoid receptors in liver homogenates, and the activity of enzymes of the antioxidant system (superoxide dismutase and catalase). We were making control measurements on days 1, 3, 5, 7, 14, 21, and 35 after the simulated IRP. Results — On day 1 after simulation of IRP development, the levels of cortisol and aldosterone in the serum of treatment group rats were significantly higher, by 14.3% and 33.5%, respectively, compared with the control group. In response to stress (IRP), we observed the highest concentration of cortisol in the blood of treatment group rats on day 3 (p=0.0002), which decreased afterwards. On day 1 after IRP, there was a reduction in the activity of superoxide dismutase and catalase in treatment group rats, by 50.3% and by 29%, respectively (p<0.0001). The lowest antioxidant activity in the rat liver after IRP was observed on days 3-7. Conclusion — Systemic IRP is associated with pronounced changes in the dynamics of corticosteroid receptors in the liver, which leads to a reduction in the activity of key antioxidant enzymes.
Collapse
|
20
|
The posttraumatic response of CD4+ regulatory T cells is modulated by direct cell-cell contact via CD40L- and P-selectin-dependent pathways. Cent Eur J Immunol 2021; 46:283-294. [PMID: 34764800 PMCID: PMC8574106 DOI: 10.5114/ceji.2021.109171] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 05/14/2021] [Indexed: 12/14/2022] Open
Abstract
CD4+ FoxP3+ regulatory T cells (CD4+ Tregs) are important for the posttraumatic anti-inflammatory host response. As described previously, platelets are able to modulate CD4+ Treg activity in a reciprocally activating interaction following injury. The underlying mechanisms of the posttraumatic interaction between platelets and CD4+ Tregs remain unclear. We investigated the potential influence of CD40L and P-selectin, molecules known to be involved in direct cell contact of these cell types. In a murine burn injury model, the potential interaction pathways were addressed using CD40L- and P-selectin-deficient mice. Draining lymph nodes were harvested following trauma (1 h) and following a sham procedure. Early rapid activation of CD4+ Tregs was assessed by phospho-flow cytometry (signaling molecules (p)PKC-δ and (p)ZAP-70). Platelet function was analyzed performing rotational thromboelastometry (ROTEM). We hypothesized that disruption of the direct cell-cell contact via CD40L and P-selectin would affect posttraumatic activation of CD4+ Tregs and influence the hemostatic function of platelets. Indeed, while injury induced early activation of CD4+ Tregs in wild-type mice (ZAP-70: p = 0.13, pZAP-70: p < 0.05, PKC-δ: p < 0.05, pPKC-δ: p < 0.05), disruption of CD40L-dependent interaction (ZAP-70: p = 0.57, pZAP-70: p = 0.68, PKC-δ: p = 0.68, pPKC-δ: p = 0.9) or P-selectin-dependent interaction (ZAP-70: p = 0.78, pZAP-70: p = 0.58, PKC-δ: p = 0.81, pPKC-δ: p = 0.73) resulted in reduced posttraumatic activation. Furthermore, hemostatic function was impaired towards hypocoagulability in either deficiency. Our results suggest that the posttraumatic activation of CD4+ Tregs and hemostatic function of platelets are affected by direct cell-cell-signaling via CD40L and P-selectin.
Collapse
|
21
|
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
|
22
|
Lopez K, Suen A, Yang Y, Wang S, Williams B, Zhu J, Hu J, Fiskum G, Cross A, Kozar R, Miller C, Zou L, Chao W. Hypobaria Exposure Worsens Cardiac Function and Endothelial Injury in AN Animal Model of Polytrauma: Implications for Aeromedical Evacuation. Shock 2021; 56:601-610. [PMID: 33394971 PMCID: PMC8522996 DOI: 10.1097/shk.0000000000001716] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Aeromedical evacuation can expose traumatically injured patients to low pressure (hypobaria) and hypoxia. Here, we sought to assess the impact of hypobaria on inflammation, organ injury, and mortality in a mouse model of polytrauma. METHODS Eight to 12-week-old male C57BL/6J mice were subjected to sham or polytrauma consisting of bowel ischemia by superior mesenteric artery occlusion, hindlimb muscle crush, and tibia fracture. Two hours after injury, animals were randomized to undergo either 6 h of hypobaria or sea-level, room air conditions. At 8 or 24 h after injury, transthoracic echocardiography was performed. Acute kidney injury (AKI) biomarkers were measured by qRT-PCR. Plasma cytokine and endothelial injury markers were determined by enzyme-linked immunosorbent assay. RESULTS Eight hours after traumatic injury, mice exhibited a marked increase in plasma IL-6 (57 pg/mL vs. 1,216 pg/mL), AKI with increased Ngal and Kim-1, and endothelial injury as evidenced by significantly increased plasma hyaluronic acid (96 ng/mL vs.199 ng/mL), thrombomodulin (23.2 ng/mL vs. 58.9 ng/mL), syndecan-1 (0.99 ng/mL vs. 4.34 ng/mL), and E-selectin (38.6 ng/mL vs. 62.7 ng/mL). The trauma mice also developed cardiac dysfunction with decreased cardiac output and stroke volume at 8 h postinjury. Hypobaric exposure after polytrauma led to decreased ejection fraction (81.0% vs. 74.2%, P < 0.01) and increased plasma hyaluronic acid (199 ng/mL vs. 260 ng/mL, P < 0.05), thrombomodulin (58.9 ng/mL vs. 75.4 ng/mL, P < 0.05), and syndecan-1 (4.34 ng/mL vs. 8.33 ng/mL, P < 0.001) at 8 h postinjury. CONCLUSIONS Hypobaria exposure appeared to worsen cardiac dysfunction and endothelial injury following polytrauma and thus may represent a physiological "second hit" following traumatic injury.
Collapse
Affiliation(s)
- Kerri Lopez
- Translational Research Program, Department of Anesthesiology & Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Andrew Suen
- Translational Research Program, Department of Anesthesiology & Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Anesthesia, Pain Management & Perioperative Medicine, Dalhousie University, Halifax, NS, Canada
| | - Yang Yang
- Translational Research Program, Department of Anesthesiology & Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Ultrasound, The 2 Teaching Hospital, Xiangya School of Medicine, Central South University, Changsha, China
| | - Sheng Wang
- Translational Research Program, Department of Anesthesiology & Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Brittney Williams
- Translational Research Program, Department of Anesthesiology & Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jing Zhu
- Translational Research Program, Department of Anesthesiology & Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jiang Hu
- Translational Research Program, Department of Anesthesiology & Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China
| | - Gary Fiskum
- Translational Research Program, Department of Anesthesiology & Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Alan Cross
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Rosemary Kozar
- Program in Trauma & Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Catriona Miller
- Enroute Care Division, Department of Aeromedical Research, USAF School of Aerospace Medicine, Wright Patterson AFB, Dayton OH, USA
| | - Lin Zou
- Translational Research Program, Department of Anesthesiology & Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Wei Chao
- Translational Research Program, Department of Anesthesiology & Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
23
|
Kleinveld DJB, Simons DDG, Dekimpe C, Deconinck SJ, Sloos PH, Maas MAW, Kers J, Muia J, Brohi K, Voorberg J, Vanhoorelbeke K, Hollmann MW, Juffermans NP. Plasma and rhADAMTS13 reduce trauma-induced organ failure by restoring the ADAMTS13-VWF axis. Blood Adv 2021; 5:3478-3491. [PMID: 34505883 PMCID: PMC8525227 DOI: 10.1182/bloodadvances.2021004404] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 05/06/2021] [Indexed: 11/20/2022] Open
Abstract
Trauma-induced organ failure is characterized by endothelial dysfunction. The aim of this study was to investigate the role of von Willebrand factor (VWF) and its cleaving enzyme, ADAMTS13 (a disintegrin and metalloproteinase with thrombospondin type 1 motifs, member 13) in the occurrence of endothelial permeability and organ failure in trauma. In an observational study in a level-1 trauma center, 169 adult trauma patients with clinical signs of shock and/or severe injuries were included. Trauma was associated with low ADAMTS13 and high VWF antigen levels, thus generating an imbalance of ADAMTS13 to VWF. Patients who developed organ failure (23%) had greater ADAMTS13-to-VWF imbalances, persistently lower platelet counts, and elevated levels of high-molecular-weight VWF multimers compared with those without organ failure, suggesting microthrombi formation. To investigate the effect of replenishing low ADAMTS13 levels on endothelial permeability and organ failure using either recombinant human ADAMTS13 (rhADAMTS13) or plasma transfusion, a rat model of trauma-induced shock and transfusion was used. Rats in traumatic hemorrhagic shock were randomized to receive crystalloids, crystalloids supplemented with rhADAMTS13, or plasma transfusion. A 70-kDa fluorescein isothiocyanate-labeled dextran was injected to determine endothelial leakage. Additionally, organs were histologically assessed. Both plasma transfusion and rhADAMTS13 were associated with a reduction in pulmonary endothelial permeability and organ injury when compared with resuscitation with crystalloids, but only rhADAMTS13 resulted in significant improvement of a trauma-induced decline in ADAMTS13 levels. We conclude that rhADAMTS13 and plasma transfusion can reduce organ failure following trauma. These findings implicate the ADAMTS13-VWF axis in the pathogenesis of organ failure.
Collapse
Affiliation(s)
- Derek J B Kleinveld
- Department of Intensive Care Medicine
- Laboratory of Experimental Intensive Care and Anesthesiology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Derek D G Simons
- Department of Intensive Care Medicine
- Laboratory of Experimental Intensive Care and Anesthesiology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Charlotte Dekimpe
- Laboratory for Thrombosis Research, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium
| | - Shannen J Deconinck
- Laboratory for Thrombosis Research, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium
| | - Pieter H Sloos
- Department of Intensive Care Medicine
- Laboratory of Experimental Intensive Care and Anesthesiology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - M Adrie W Maas
- Laboratory of Experimental Intensive Care and Anesthesiology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Jesper Kers
- Department of Pathology, Amsterdam Infection & Immunity Institute, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Joshua Muia
- Department of Biochemistry and Microbiology, Oklahoma State University Center for Health Sciences, Tulsa, OK
| | - Karim Brohi
- Centre for Trauma Sciences, Queen Mary University of London, London, United Kingdom
| | - Jan Voorberg
- Sanquin, Department of Cellular Hemostasis, Amsterdam, The Netherlands
| | - Karen Vanhoorelbeke
- Laboratory for Thrombosis Research, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium
| | - Markus W Hollmann
- Laboratory of Experimental Intensive Care and Anesthesiology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Department of Anesthesiology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands; and
| | - Nicole P Juffermans
- Laboratory of Experimental Intensive Care and Anesthesiology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Department of Intensive Care Medicine, Onze Lieve Vrouwe Gasthuis, Amsterdam, The Netherlands
| |
Collapse
|
24
|
Leditzke K, Wagner MEH, Neunaber C, Clausen JD, Winkelmann M. Neutrophil Gelatinase-associated Lipocalin Predicts Post-traumatic Acute Kidney Injury in Severely Injured Patients. In Vivo 2021; 35:2755-2762. [PMID: 34410965 DOI: 10.21873/invivo.12560] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/02/2021] [Accepted: 07/08/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Early detection of acute kidney injury (AKI) is crucial in the management of multiple-organ dysfunction syndrome in severely injured patients. Standard laboratory parameters usually increase with temporal delay. Therefore, we evaluated neutrophil gelatinase-associated lipocalin (NGAL) as an early marker for acute kidney injury. PATIENTS AND METHODS We retrospectively evaluated patients admitted to a level 1 trauma center. We collected clinicodemographic data and measured kidney-related factors and plasma cytokines. RESULTS A total of 39 patients were included. Patients with AKI had significantly higher levels not only of serum creatinine and urea, but also of NGAL (all p<0.001) than patients without AKI. The optimal NGAL cut-off value was determined to be 177 ng/ml, showing significant correlation with imminent or manifest AKI (p<0.001). Other independent markers correlated with AKI included pre-existing chronic kidney disease, use of catecholamines, and severe injury (p<0.001). CONCLUSION The serum level of NGAL is feasible early predictor of AKI.
Collapse
|
25
|
Kleinveld DJB, van Amstel RBE, Wirtz MR, Geeraedts LMG, Goslings JC, Hollmann MW, Juffermans NP. Platelet-to-red blood cell ratio and mortality in bleeding trauma patients: A systematic review and meta-analysis. Transfusion 2021; 61 Suppl 1:S243-S251. [PMID: 34269443 PMCID: PMC8362120 DOI: 10.1111/trf.16455] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/18/2021] [Accepted: 01/18/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND In traumatic bleeding, transfusion practice has shifted toward higher doses of platelets and plasma transfusion. The aim of this systematic review was to investigate whether a higher platelet-to-red blood cell (RBC) transfusion ratio improves mortality without worsening organ failure when compared with a lower ratio of platelet-to-RBC. METHODS Pubmed, Medline, and Embase were screened for randomized controlled trials (RCTs) in bleeding trauma patients (age ≥16 years) receiving platelet transfusion between 1946 until October 2020. High platelet:RBC ratio was defined as being the highest ratio within an included study. Primary outcome was 24 hour mortality. Secondary outcomes were 30-day mortality, thromboembolic events, organ failure, and correction of coagulopathy. RESULTS In total five RCTs (n = 1757 patients) were included. A high platelet:RBC compared with a low platelet:RBC ratio significantly improved 24 hour mortality (odds ratio [OR] 0.69 [0.53-0.89]) and 30- day mortality (OR 0.78 [0.63-0.98]). There was no difference between platelet:RBC ratio groups in thromboembolic events and organ failure. Correction of coagulopathy was reported in five studies, in which platelet dose had no impact on trauma-induced coagulopathy. CONCLUSIONS In traumatic bleeding, a high platelet:RBC improves mortality as compared to low platelet:RBC ratio. The high platelet:RBC ratio does not influence thromboembolic or organ failure event rates.
Collapse
Affiliation(s)
- Derek J B Kleinveld
- Department of Intensive Care Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Laboratory of Experimental Intensive Care and Anesthesiology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Rombout B E van Amstel
- Department of Intensive Care Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Laboratory of Experimental Intensive Care and Anesthesiology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Mathijs R Wirtz
- Department of Intensive Care Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Laboratory of Experimental Intensive Care and Anesthesiology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Leo M G Geeraedts
- Department of Trauma Surgery, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - J Carel Goslings
- Department of Trauma Surgery, OLVG Hospital, Amsterdam, The Netherlands
| | - Markus W Hollmann
- Laboratory of Experimental Intensive Care and Anesthesiology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Department of Anesthesiology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Nicole P Juffermans
- Department of Intensive Care Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Laboratory of Experimental Intensive Care and Anesthesiology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Department of Intensive Care, OLVG Hospital, Amsterdam, The Netherlands
| |
Collapse
|
26
|
Alternative Complement Pathway Activation Provokes a Hypercoagulable State with Diminished Fibrinolysis. Shock 2021; 53:560-565. [PMID: 31441792 DOI: 10.1097/shk.0000000000001437] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
INTRODUCTION Several disease processes trigger prolonged activation of the alternative complement pathway. Crosslinks between complement activation and physiologic changes in platelets and neutrophils have been identified, but how this interplay alters the hemostatic potential in humans remains undefined. We hypothesize that activation of the alternative pathway triggers a hypercoagulable state. METHODS C3/C5 convertase Cobra Venom Factor (CVF, 10 Units/mL) was employed to activate the alternative complement pathway in whole blood. Complement inhibition was completed with inhibitors for C3/C3b (Compstatin, 25 and 50 μM), C3a receptor (SB290157, 300 nM, C3aR), and C5a receptor (W54011, 6 nM, C5aR). Coagulation was assessed using native thrombelastography which produces the following: reaction time (R time); angle; maximum amplitude (MA); percent fibrinolysis at 30-min post-MA (LY30). RESULTS Inhibition with C3aR and C5aR inhibitors did not alter clot formation (R time, 11.2 vs 11.6 min, P = 0.36), clot strength (MA, 52.0 vs 52.3 mm, P = 0.43), or fibrinolysis (LY30, 1.6 vs 4.0%, P = 0.19). Compstatin did not influence clot formation or clot strength but did induce a dose-dependent increase in fibrinolysis (control LY30 3.0 vs 7.8% and 12.4% for 25 and 50 μM respectively, P = 0.0002). CVF increased MA (58.0 vs 62.8 mm, P < 0.0001), decreased LY30 (2.3 vs 1.4%, P = 0.004), and increased R time (8.4 vs 9.9 min, P = 0.008). Compstatin reversed the effects of CVF, while C5a reversed only the change in LY30. CONCLUSIONS C3 contributes to fibrinolysis, as inhibition with Compstatin enhanced fibrinolysis, and CVF cleavage of C3 decreased fibrinolysis. CVF also induced a hypercoagulable state with increased clot strength.
Collapse
|
27
|
Haupt J, Krysiak N, Unger M, Bogner-Flatz V, Biberthaler P, Hanschen M, van Griensven M, Haug AT. The potential of adipokines in identifying multiple trauma patients at risk of developing multiple organ dysfunction syndrome. Eur J Med Res 2021; 26:38. [PMID: 33931112 PMCID: PMC8086117 DOI: 10.1186/s40001-021-00511-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 04/22/2021] [Indexed: 01/06/2023] Open
Abstract
Background Multiple organ dysfunction syndrome (MODS) and the consecutive multiple organ failure (MOF) are severe and dreaded complications with a high mortality in multiple trauma patients. The aim of this study was to investigate the potential of the adipokines leptin, resistin, interleukin-17A and interleukin-33 as possible biomarkers in the early posttraumatic inflammatory response and for identifying severely traumatized patients at risk of developing MODS. Methods In total, 14 multiple trauma patients with an injury severity score (ISS) ≥ 16 as well as a control group of 14 non-multiple trauma patients were included in this study and blood samples were taken at the time points 0, 6, 24, 48 and 72 h after admission. For the trauma patients, the SIRS and Denver MOF score were determined daily. The quantitative measurement of the plasma concentrations of the adipokines was performed using ELISA. Results In the statistical analysis, the multiple trauma patients showed statistically significant higher plasma concentrations of leptin, resistin, IL-17A and IL-33 compared to the control group. In addition, there was a statistically significant positive correlation between the concentrations of resistin, IL-17A and IL-33 and the corresponding SIRS scores and between the concentrations of resistin, IL-17A and IL-33 and the corresponding Denver MOF scores. Finally, ROC curve analysis revealed that the adipokines leptin and IL-17A are suitable diagnostic markers for the discrimination between multiple trauma patients with and without MOF. Conclusions Leptin and IL-17A could be suitable diagnostic markers to identify severely injured patients with a developing SIRS and MOF earlier, to adjust surgical therapy planning and intensive care.
Collapse
Affiliation(s)
- Julian Haupt
- Experimental Trauma Surgery, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Strasse 22, 81675, Munich, Germany. .,Bundeswehr Institute of Radiobiology affiliated to the University Ulm, Neuherbergstrasse 11, 80937, Munich, Germany.
| | - Niels Krysiak
- Experimental Trauma Surgery, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Strasse 22, 81675, Munich, Germany
| | - Marina Unger
- Experimental Trauma Surgery, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Strasse 22, 81675, Munich, Germany
| | - Viktoria Bogner-Flatz
- Department of Trauma Surgery, University Hospital Munich, Ludwig-Maximilians-University, Nussbaumstrasse 20, 80336, Munich, Germany
| | - Peter Biberthaler
- Department of Trauma Surgery, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Strasse 22, 81675, Munich, Germany
| | - Marc Hanschen
- Experimental Trauma Surgery, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Strasse 22, 81675, Munich, Germany.,Department of Trauma Surgery, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Strasse 22, 81675, Munich, Germany
| | - Martijn van Griensven
- Experimental Trauma Surgery, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Strasse 22, 81675, Munich, Germany.,Department of Cell Biology-Inspired Tissue Engineering (cBITE), MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, The Netherlands
| | - Alexander T Haug
- Experimental Trauma Surgery, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Strasse 22, 81675, Munich, Germany.,Department of Trauma Surgery, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Strasse 22, 81675, Munich, Germany.,Department of Orthopedics and Sports Orthopedics, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Strasse 22, 81675, Munich, Germany
| |
Collapse
|
28
|
Walsh SA, Hoyt BW, Rowe CJ, Dey D, Davis TA. Alarming Cargo: The Role of Exosomes in Trauma-Induced Inflammation. Biomolecules 2021; 11:biom11040522. [PMID: 33807302 PMCID: PMC8065643 DOI: 10.3390/biom11040522] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 03/26/2021] [Accepted: 03/29/2021] [Indexed: 12/11/2022] Open
Abstract
Severe polytraumatic injury initiates a robust immune response. Broad immune dysfunction in patients with such injuries has been well-documented; however, early biomarkers of immune dysfunction post-injury, which are critical for comprehensive intervention and can predict the clinical course of patients, have not been reported. Current circulating markers such as IL-6 and IL-10 are broad, non-specific, and lag behind the clinical course of patients. General blockade of the inflammatory response is detrimental to patients, as a certain degree of regulated inflammation is critical and necessary following trauma. Exosomes, small membrane-bound extracellular vesicles, found in a variety of biofluids, carry within them a complex functional cargo, comprised of coding and non-coding RNAs, proteins, and metabolites. Composition of circulating exosomal cargo is modulated by changes in the intra- and extracellular microenvironment, thereby serving as a homeostasis sensor. With its extensively documented involvement in immune regulation in multiple pathologies, study of exosomal cargo in polytrauma patients can provide critical insights on trauma-specific, temporal immune dysregulation, with tremendous potential to serve as unique biomarkers and therapeutic targets for timely and precise intervention.
Collapse
Affiliation(s)
- Sarah A. Walsh
- USU Walter Reed Surgery, Uniformed Services University, Bethesda, MD 20814, USA; (S.A.W.); (B.W.H.); (C.J.R.); (D.D.)
| | - Benjamin W. Hoyt
- USU Walter Reed Surgery, Uniformed Services University, Bethesda, MD 20814, USA; (S.A.W.); (B.W.H.); (C.J.R.); (D.D.)
| | - Cassie J. Rowe
- USU Walter Reed Surgery, Uniformed Services University, Bethesda, MD 20814, USA; (S.A.W.); (B.W.H.); (C.J.R.); (D.D.)
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
| | - Devaveena Dey
- USU Walter Reed Surgery, Uniformed Services University, Bethesda, MD 20814, USA; (S.A.W.); (B.W.H.); (C.J.R.); (D.D.)
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
| | - Thomas A. Davis
- USU Walter Reed Surgery, Uniformed Services University, Bethesda, MD 20814, USA; (S.A.W.); (B.W.H.); (C.J.R.); (D.D.)
- Correspondence:
| |
Collapse
|
29
|
Abstract
INTRODUCTION Traumatic brain injury (TBI) is associated with secondary injury to the central nervous system (CNS) via inflammatory mechanisms. The combination of polytrauma and TBI further exacerbates the inflammatory response to injury; however, combined injury phenomena have not been thoroughly studied. In this study, we examined the inflammatory differences between patients with TBI versus patients with polytrauma, but no TBI (polytrauma). We hypothesize that patients with TBI have a heightened early inflammatory response compared with polytrauma. METHODS We conducted a single-center retrospective study of a cohort of patients with polytrauma, who were enrolled in the PROPPR study. These patients had blood samples prospectively collected at eight time points in the first 3 days of admission. Using radiological data to determine TBI, our polytrauma cohort was dichotomized into TBI (n = 30) or polytrauma (n = 54). Inflammatory biomarkers were measured using ELISA. Data across time were compared for TBI versus polytrauma groups using Wilcoxon rank-sum test. Network analysis techniques were used to systematically characterize the inflammatory responses at admission. RESULTS Patients with TBI (51.6%) had a higher 30-day mortality compared with polytrauma (16.9%) (P <0.001). Expression levels of IL6, IL8, and CCL2 were elevated from the 2-h through 24-h time points, becoming significant at the 6-h time point (IL6, IL8, and CCL2; P <0.05) (). CSF3 showed a similar pattern, but did not attain significance. TBI and polytrauma networks underwent diverging trends from admission to the 6-h time point. CONCLUSION Patients with TBI demonstrated upregulations in proinflammatory cytokines IL6, IL8, and CCL2. Utilizing informatics methods, we were able to identify temporal differences in network trends, as well as uncharacterized cytokines and chemokines in TBI. These data suggest TBI induces a distinct inflammatory response and pathologically heightened inflammatory response in the presence of polytrauma and may propagate worsened patient outcomes including mortality.
Collapse
|
30
|
Taghavi S, Abdullah S, Duchesne J, Pociask D, Kolls J, Jackson-Weaver O. Interleukin 22 mitigates endothelial glycocalyx shedding after lipopolysaccharide injury. J Trauma Acute Care Surg 2021; 90:337-345. [PMID: 33502147 PMCID: PMC7872437 DOI: 10.1097/ta.0000000000003019] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND The endothelial glycocalyx (EG) on the luminal surface of endothelial cells contributes to the permeability barrier of vessels and prevents activation of the coagulation cascade. Endothelial glycocalyx damage, which occurs in the shock state, results in endotheliopathy. Interleukin (IL)-22 is a cytokine with both proinflammatory and anti-inflammatory properties, and how IL-22 affects the EG has not been studied. We hypothesized that IL-22:Fc, a recombinant fusion protein with human IL-22 and the Fc portion of human immunoglobulin G1 (which extends the protein half-life), would not affect EG shedding in endothelium after injury. METHODS Human umbilical vein endothelial cells (HUVECs) were exposed to 1 μg/mL lipopolysaccharide (LPS). Lipopolysaccharide-injured cells (n = 284) were compared with HUVECs with LPS injury plus 0.375 μg/mL of IL-22:Fc treatment (n = 293) for 12 hours. These two cohorts were compared with control HUVECs (n = 286) and HUVECs exposed to IL-22:Fc alone (n = 269). Cells were fixed and stained with fluorescein isothiocyanate-labeled wheat germ agglutinin to quantify EG. Total RNA was collected, and select messenger RNAs were quantified by real time - quantitative polymerase chain reaction (RT-qPCR) using SYBR green fluorescence. RESULTS Exposure of HUVECs to LPS resulted in degradation of the EG compared with control (5.86 vs. 6.09 arbitrary unit [AU], p = 0.01). Interleukin-22:Fc alone also resulted in degradation of EG (5.08 vs. 6.09 AU, p = 0.01). Treatment with IL-22:Fc after LPS injury resulted in less degradation of EG compared with LPS injury alone (5.86 vs. 5.08 AU, p = 0.002). Expression of the IL-22Ra1 receptor was not different for IL-22:Fc treated compared with LPS injury only (0.69 vs. 0.86 relative expression, p = 0.10). Treatment with IL-22:Fc after LPS injury resulted in less matrix metalloproteinase 2 (0.79 vs. 1.70 relative expression, p = 0.005) and matrix metalloproteinase 14 (0.94 vs. 2.04 relative expression, p = 0.02). CONCLUSIONS Interleukin-22:Fc alone induces EG degradation. However, IL-22:Fc treatment after LPS injury appears to mitigate EG degradation. This protective effect appears to be mediated via reduced expression of metalloproteinases.
Collapse
Affiliation(s)
- Sharven Taghavi
- From the Department of Surgery (S.T., S.A., J.D., O.J.-W.), and Center for Translational Research in Infection and Inflammation (D.P., J.K.), Tulane University School of Medicine, New Orleans, Louisiana
| | | | | | | | | | | |
Collapse
|
31
|
Cohen JT, Danise M, Machan JT, Zhao R, Lefort CT. Murine Myeloid Progenitors Attenuate Immune Dysfunction Induced by Hemorrhagic Shock. Stem Cell Reports 2021; 16:324-336. [PMID: 33482101 PMCID: PMC7878835 DOI: 10.1016/j.stemcr.2020.12.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 12/20/2020] [Accepted: 12/21/2020] [Indexed: 12/11/2022] Open
Abstract
Hemorrhagic shock induces an aberrant immune response characterized by simultaneous induction of a proinflammatory state and impaired host defenses. The objective of this study was to evaluate the impact of conditionally immortalized neutrophil progenitors (NPs) on this aberrant immune response. We employed a mouse model of hemorrhagic shock, followed by the adoptive transfer of NPs and subsequent inoculation of Staphylococcus aureus to induce pneumonia. We observed that transplant of NPs decreases the proportion of host neutrophils that express programmed death ligand 1 and intercellular adhesion molecule 1 in the context of prior hemorrhage. Following hemorrhage, NP transplant decreased proinflammatory cytokines in the lungs, increased neutrophil migration into the airspaces, and enhanced bacterial clearance. Further, hemorrhagic shock improved NP engraftment in the bone marrow. These results suggest that NPs hold the potential for use as a cellular therapy in the treatment and prevention of secondary infection following hemorrhagic shock. Myeloid progenitors restore a competent inflammatory response to pneumonia Progenitor transplantation promotes clearance of secondary S. aureus pneumonia Hemorrhagic shock enhances engraftment of transplanted myeloid progenitors
Collapse
Affiliation(s)
- Joshua T Cohen
- Division of Surgical Research, Department of Surgery, Rhode Island Hospital, 593 Eddy Street, Providence, RI 02903, USA
| | - Michael Danise
- Division of Surgical Research, Department of Surgery, Rhode Island Hospital, 593 Eddy Street, Providence, RI 02903, USA
| | - Jason T Machan
- Lifespan Biostatistics Core, Rhode Island Hospital, Providence, RI 02903, USA
| | - Runping Zhao
- Division of Surgical Research, Department of Surgery, Rhode Island Hospital, 593 Eddy Street, Providence, RI 02903, USA
| | - Craig T Lefort
- Division of Surgical Research, Department of Surgery, Rhode Island Hospital, 593 Eddy Street, Providence, RI 02903, USA.
| |
Collapse
|
32
|
A Tangled Threesome: Circadian Rhythm, Body Temperature Variations, and the Immune System. BIOLOGY 2021; 10:biology10010065. [PMID: 33477463 PMCID: PMC7829919 DOI: 10.3390/biology10010065] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/06/2021] [Accepted: 01/10/2021] [Indexed: 12/14/2022]
Abstract
Simple Summary In mammals, including humans, the body temperature displays a circadian rhythm and is maintained within a narrow range to facilitate the optimal functioning of physiological processes. Body temperature increases during the daytime and decreases during the nighttime thus influencing the expression of the molecular clock and the clock-control genes such as immune genes. An increase in body temperature (daytime, or fever) also prepares the organism to fight aggression by promoting the activation, function, and delivery of immune cells. Many factors may affect body temperature level and rhythm, including environment, age, hormones, or treatment. The disruption of the body temperature is associated with many kinds of diseases and their severity, thus supporting the assumed association between body temperature rhythm and immune functions. Recent studies using complex analysis suggest that circadian rhythm may change in all aspects (level, period, amplitude) and may be predictive of good or poor outcomes. The monitoring of body temperature is an easy tool to predict outcomes and maybe guide future studies in chronotherapy. Abstract The circadian rhythm of the body temperature (CRBT) is a marker of the central biological clock that results from multiple complex biological processes. In mammals, including humans, the body temperature displays a strict circadian rhythm and has to be maintained within a narrow range to allow optimal physiological functions. There is nowadays growing evidence on the role of the temperature circadian rhythm on the expression of the molecular clock. The CRBT likely participates in the phase coordination of circadian timekeepers in peripheral tissues, thus guaranteeing the proper functioning of the immune system. The disruption of the CRBT, such as fever, has been repeatedly described in diseases and likely reflects a physiological process to activate the molecular clock and trigger the immune response. On the other hand, temperature circadian disruption has also been described as associated with disease severity and thus may mirror or contribute to immune dysfunction. The present review aims to characterize the potential implication of the temperature circadian rhythm on the immune response, from molecular pathways to diseases. The origin of CRBT and physiological changes in body temperature will be mentioned. We further review the immune biological effects of temperature rhythmicity in hosts, vectors, and pathogens. Finally, we discuss the relationship between circadian disruption of the body temperature and diseases and highlight the emerging evidence that CRBT monitoring would be an easy tool to predict outcomes and guide future studies in chronotherapy.
Collapse
|
33
|
Bedard AF, Mata LV, Dymond C, Moreira F, Dixon J, Schauer SG, Ginde AA, Bebarta V, Moore EE, Mould-Millman NK. A scoping review of worldwide studies evaluating the effects of prehospital time on trauma outcomes. Int J Emerg Med 2020; 13:64. [PMID: 33297951 PMCID: PMC7724615 DOI: 10.1186/s12245-020-00324-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 11/21/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Annually, over 1 billion people sustain traumatic injuries, resulting in over 900,000 deaths in Africa and 6 million deaths globally. Timely response, intervention, and transportation in the prehospital setting reduce morbidity and mortality of trauma victims. Our objective was to describe the existing literature evaluating trauma morbidity and mortality outcomes as a function of prehospital care time to identify gaps in literature and inform future investigation. MAIN BODY We performed a scoping review of published literature in MEDLINE. Results were limited to English language publications from 2009 to 2020. Included articles reported trauma outcomes and prehospital time. We excluded case reports, reviews, systematic reviews, meta-analyses, comments, editorials, letters, and conference proceedings. In total, 808 articles were identified for title and abstract review. Of those, 96 articles met all inclusion criteria and were fully reviewed. Higher quality studies used data derived from trauma registries. There was a paucity of literature from studies in low- and middle-income countries (LMIC), with only 3 (3%) of articles explicitly including African populations. Mortality was an outcome measure in 93% of articles, predominantly defined as "in-hospital mortality" as opposed to mortality within a specified time frame. Prehospital time was most commonly assessed as crude time from EMS dispatch to arrival at a tertiary trauma center. Few studies evaluated physiologic morbidity outcomes such as multi-organ failure. CONCLUSION The existing literature disproportionately represents high-income settings and most commonly assessed in-hospital mortality as a function of crude prehospital time. Future studies should focus on how specific prehospital intervals impact morbidity outcomes (e.g., organ failure) and mortality at earlier time points (e.g., 3 or 7 days) to better reflect the effect of early prehospital resuscitation and transport. Trauma registries may be a tool to facilitate such research and may promote higher quality investigations in Africa and LMICs.
Collapse
Affiliation(s)
- Alexander F Bedard
- University of Colorado, Anschutz Medical Campus, 13001 E 17th Place, Aurora, CO, 80045, USA.
- United States Air Force Medical Corps, 7700 Arlington Boulevard, Falls Church, VA, 22042, USA.
| | - Lina V Mata
- University of Colorado, Anschutz Medical Campus, 13001 E 17th Place, Aurora, CO, 80045, USA
| | - Chelsea Dymond
- University of Colorado, Anschutz Medical Campus, 13001 E 17th Place, Aurora, CO, 80045, USA
- Denver Health and Hospital Authority, 777 Bannock St, Denver, CO, 80204, USA
| | - Fabio Moreira
- Western Cape Government, Emergency Medical Services, 9 Wale Street, Cape Town, 8001, South Africa
| | - Julia Dixon
- University of Colorado, Anschutz Medical Campus, 13001 E 17th Place, Aurora, CO, 80045, USA
| | - Steven G Schauer
- US Army Institute of Surgical Research, 3698 Chambers Rd., San Antonio, TX, 78234, USA
| | - Adit A Ginde
- University of Colorado, Anschutz Medical Campus, 13001 E 17th Place, Aurora, CO, 80045, USA
| | - Vikhyat Bebarta
- University of Colorado, Anschutz Medical Campus, 13001 E 17th Place, Aurora, CO, 80045, USA
| | - Ernest E Moore
- University of Colorado, Anschutz Medical Campus, 13001 E 17th Place, Aurora, CO, 80045, USA
- Ernest E. Moore Shock Trauma Center at Denver Health, 777 Bannock St, Denver, CO, 80204, USA
| | - Nee-Kofi Mould-Millman
- University of Colorado, Anschutz Medical Campus, 13001 E 17th Place, Aurora, CO, 80045, USA
| |
Collapse
|
34
|
Carbon Monoxide-Releasing Molecule-3 Ameliorates Acute Lung Injury in a Model of Hemorrhagic Shock and Resuscitation: Roles of p38MAPK Signaling Pathway. Shock 2020; 55:816-826. [PMID: 33105439 DOI: 10.1097/shk.0000000000001684] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVE It was reported that carbon monoxide-releasing molecule-3 (CORM-3) administration immediately after hemorrhagic shock and resuscitation (HSR) ameliorates the HSR-induced acute lung injury (ALI); however, the specific mechanism of the protective effects against HSR-induced ALI remains unclear. METHODS To induce hemorrhagic shock, rats were bled to a mean arterial blood pressure of 30 mm Hg for 45 min and then resuscitated with shed blood via the left vein. CORM-3 (4 mg/kg or 8 mg/kg) was respectively administrated after HSR. Twelve hours post-HSR, lung injury was assessed by wet/dry (W/D) ratio, hematoxylin-eosin staining staining, and lung ultrasound; the apoptotic and pyroptotic macrophages were measured by immunofluorescence staining; and the expression of phosphorylated p38 mitogen activated protein kinase (p-p38MAPK) and total p38MAPK was measured by western blotting. SB203580 (5 mg/kg), a special inhibitor of p-p38MAPK, was administrated by abdominal cavity to assess the roles of p38MAPK in HSR-induced ALI. RESULTS Increased B-line score, lung injury score, and W/D ratio indicated the fact of ALI after HSR. Twelve hours post-HSR, CORM-3 administration significantly decreased the B-line score, lung injury score, W/D ratio, apoptotic and pyroptotic macrophages, and the expressions of p-p38MAPK. Further, SB203580 not only reduced HSR-induced ALI, but also enhanced the protective effects of CORM-3 against ALI. CONCLUSION We identified the protective effects of CORM-3 against HSR-induced ALI. The mechanism might be related to the inhibition of p38MAPK signaling pathway in lung macrophages.
Collapse
|
35
|
Dieteren S, Franz N, Köhler K, Nowak A, Ehnert S, Surov A, Krüger M, Marzi I, Wagner N, Relja B. Ethyl Pyruvate Reduces Systemic Leukocyte Activation via Caspase-1 and NF-κB After Blunt Chest Trauma and Haemorrhagic Shock. Front Med (Lausanne) 2020; 7:562904. [PMID: 33117829 PMCID: PMC7562791 DOI: 10.3389/fmed.2020.562904] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 08/28/2020] [Indexed: 12/22/2022] Open
Abstract
Background: Blunt chest (thoracic) trauma (TxT) and haemorrhagic shock with subsequent resuscitation (H/R) induce strong systemic and local inflammatory response, which is closely associated with apoptotic cell loss and subsequently impaired organ function. The underlying mechanisms are not completely understood, therefore, the treatment of patients suffering from TxT+H/R is challenging. In our recent studies, we have demonstrated local anti-inflammatory effects of ethyl pyruvate (EtP) in lung and liver after TxT+H/R. Here, the therapeutic potential of a reperfusion regime with EtP on the early post-traumatic systemic inflammatory response and apoptotic changes after TxT followed by H/R were investigated. Methods: Female Lewis rats underwent TxT followed by haemorrhagic shock (60 min). Resuscitation was performed with own blood transfusion and either lactated Ringers solution (LR) or LR supplemented with EtP (50 mg/kg). Sham group underwent the surgical procedures. After 2 h blood as well as lung and liver tissues were obtained for analyses. Systemic activation of neutrophils (expression of CD11b and CD62L), leukocyte phagocytosis, apoptosis (caspase-3/7 activation), pyroptosis (caspase-1 activation) and NF-κB p65 activity were assessed. p < 0.05 was considered significant. Results: TxT+H/R-induced systemic activation of neutrophils (increased CD11b and reduced CD62L expression) was significantly reduced by EtP. Trauma-induced delayed neutrophil apoptosis was further reduced by EtP reperfusion but remained unaltered in monocytes. Reperfusion with EtP significantly increased the phagocytizing capacity of granulocytes. Trauma-induced inflammasome activation, which was observed in monocytes and not in neutrophils, was significantly reduced by EtP in both cell entities. NF-κB p65 activation, which was increased in neutrophils and monocytes was significantly decreased in monocytes. Conclusion: TxT+H/R-induced systemic activation of both neutrophils and monocytes concomitant with increased systemic inflammation was reduced by a reperfusion with EtP and was associated with a down-regulation of NF-κB p65 activation.
Collapse
Affiliation(s)
- Scott Dieteren
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto von Guericke University, Magdeburg, Germany.,Department of Trauma, Hand and Reconstructive Surgery, University Hospital of the Goethe University Frankfurt, Frankfurt, Germany
| | - Niklas Franz
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital of the Goethe University Frankfurt, Frankfurt, Germany
| | - Kernt Köhler
- Institute of Veterinary Pathology, Justus Liebig University Giessen, Giessen, Germany
| | - Aleksander Nowak
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto von Guericke University, Magdeburg, Germany
| | - Sabrina Ehnert
- Department of Trauma and Reconstructive Surgery, BG Trauma Center Tuebingen, Siegfried Weller Research Institute, Eberhard Karls University Tuebingen, Tübingen, Germany
| | - Alexey Surov
- Department of Radiology and Nuclear Medicine, Otto von Guericke University, Magdeburg, Germany
| | - Marcus Krüger
- Department of Microgravity and Translational Regenerative Medicine, Clinic for Plastic, Aesthetic and Hand Surgery, Otto von Guericke University, Magdeburg, Germany
| | - Ingo Marzi
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital of the Goethe University Frankfurt, Frankfurt, Germany
| | - Nils Wagner
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital of the Goethe University Frankfurt, Frankfurt, Germany
| | - Borna Relja
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto von Guericke University, Magdeburg, Germany
| |
Collapse
|
36
|
Asim M, Amin F, El-Menyar A. Multiple organ dysfunction syndrome: Contemporary insights on the clinicopathological spectrum. Qatar Med J 2020; 2020:22. [PMID: 33628712 PMCID: PMC7884906 DOI: 10.5339/qmj.2020.22] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Accepted: 03/03/2020] [Indexed: 12/27/2022] Open
Abstract
Multiorgan dysfunction syndrome (MODS) remains a major complication and challenge to treat patients with critical illness in different intensive care unit settings. The exact mechanism and pathophysiology of MODS is complex and remains unexplored. We reviewed the literature from January 2011 to August 2019 to analyze the underlying mechanisms, prognostic factors, MODS scoring systems, organ systems dysfunctions, and the management of MODS. We used the search engines PubMed, MEDLINE, Scopus, and Google Scholar with the keywords "multiple organ dysfunction syndrome," "intensive care units," "multiorgan failure," "MODS scoring system," and "MODS management." The initial search yielded 3550 abstracts, of which 91 articles were relevant to the scope of the present article. A better understanding of a disease course will help differentiate the signs of an intense inflammatory response from the early onset of sepsis and minimize the inappropriate use of medications. This, in turn, will promote organtargeted therapy and prevent occurrence and progression of MODS.
Collapse
Affiliation(s)
- Mohammad Asim
- Department of Surgery, Clinical Research, Trauma Surgery Section, Hamad General Hospital, Doha, Qatar
| | - Farhana Amin
- Sri Ramaswamy Memorial Medical College Hospital & Research Center, Tamil Nadu, India
| | | |
Collapse
|
37
|
Alves NG, Trujillo AN, Breslin JW, Yuan SY. Sphingosine-1-Phosphate Reduces Hemorrhagic Shock and Resuscitation-Induced Microvascular Leakage by Protecting Endothelial Mitochondrial Integrity. Shock 2020; 52:423-433. [PMID: 30339634 DOI: 10.1097/shk.0000000000001280] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Excessive microvascular permeability is a serious complication following hemorrhagic shock and resuscitation (HSR). S1P has been shown to ameliorate microvascular leakage in a model of combined alcohol intoxication and HSR. In the current study, we tested the hypothesis that S1P reduces HSR-induced microvascular leakage by preserving endothelial cell junctional structure and the endothelial glycocalyx through the protection of mitochondrial function. We used an established in vivo rat model of conscious HSR and assessed microvascular leakage, endothelial glycocalyx integrity, and mitochondrial function by intravital microscopy. Junctional integrity in the mesenteric microcirculation was assessed by confocal microscopy. Cultured rat intestinal microvascular endothelial cells monolayers were used to test the ability of S1P to protect against glycocalyx shedding and endothelial barrier dysfunction caused by direct disruption of mitochondrial integrity due to inhibition of mitochondrial complex III. The results show that in vivo, S1P protects against HSR-induced hyperpermeability, preserves the expression of adherens junctional proteins, and protects against glycocalyx degradation. S1P treatment during HSR also protects against mitochondrial membrane depolarization. S1P also protects against mitochondrial dysfunction-induced endothelial barrier dysfunction and glycocalyx degradation by acting through mitochondrial complex III. Taken together, our data indicate that S1P protects against HSR-induced mitochondrial dysfunction in endothelial cells, which in turn improves the structure of the endothelial glycocalyx after HSR and allows for better junctional integrity to the prevention of excess microvascular permeability.
Collapse
Affiliation(s)
- Natascha G Alves
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | | | | | | |
Collapse
|
38
|
Abstract
OBJECTIVE Early differential diagnosis of an infection in a trauma patient is likely to have a significant influence on the prognosis. In the present study, we evaluated the early differential value of plasma presepsin, procalcitonin (PCT), C-reactive protein (CRP), and white blood cells (WBCs) on infection in trauma patients. METHODS Trauma patients were divided into noninfected (n = 89) and infected trauma groups (n = 68); healthy adult volunteers (n = 60) and patients having sterile surgery (n = 60) were enrolled as the controls. Plasma presepsin, PCT, CRP, and WBC counts were measured and the injury severity score (ISS) was calculated. RESULTS Plasma presepsin levels within the first 3 d of admission were only significantly increased in the infected trauma group, but not in the noninfected trauma and sterile groups. This indicated that presepsin might have an ability to differentiate the infection in trauma patients; however, plasma PCT, CRP, and WBCs were significantly increased in both the infected and noninfected trauma patients. Binary logistic regression analysis showed that only increased plasma presepsin, PCT, and ISS were significantly associated with an increased likelihood of infection in trauma patients. Both presepsin and PCT were valuable for diagnosing infection; presepsin had a higher area under the curve than PCT. CONCLUSION Presepsin might be a superior biomarker for early differentiation of infection in trauma patients; however, trauma stress elevates PCT, CRP, and WBCs even in the absence of infection; therefore, caution is advised when using these indicators to diagnose infection.
Collapse
|
39
|
Zhao G, Lu S, Li L, Fan X. Local anesthetic articaine ameliorates LPS-induced acute kidney injury via inhibition of NF-ĸB activation and the NLRP3 inflammasome pathway. J Biochem Mol Toxicol 2020; 34:e22554. [PMID: 32687258 DOI: 10.1002/jbt.22554] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 04/30/2020] [Accepted: 05/29/2020] [Indexed: 12/16/2022]
Abstract
The present study was conducted to determine the protective effect of articaine (ART) in an lipopolysaccharide (LPS)-induced acute kidney injury (AKI) animal model. The results suggest ART causes a significant decrease in serum blood urea nitrogen, creatinine, and serum cystatin C level, showing a protective effect against LPS-induced AKI. This has been further supported by histopathological findings of kidney tissues. The level of tumor necrosis factor-α, interleukin (IL)-6, and IL-1β in serum and kidney tissues was remarkably inhibited by ART in a dose-dependent manner. ART causes a significant reduction of malondialdehyde and increases the activities of glutathione and superoxide dismutase with an increase in dose as compared to the LPS-treated group. Moreover, the ART-treated group showed dose-dependent inhibition of LPS-induced nuclear factor-κB activation and TLR4 expression as confirmed by Western blot analysis. The level of Bcl-2 family genes (Bcl-2 and Bax) was restored near to normal by ART. Collectively, all the above results indicated that ART had protective effects against LPS-induced AKI by blocking inflammatory and oxidative responses.
Collapse
Affiliation(s)
- Guanjie Zhao
- Department of Nephrology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Shan Lu
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Linlin Li
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Xiaodi Fan
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
40
|
Risk Factors Associated With Early and Late Posttraumatic Multiorgan Failure: An Analysis From RETRAUCI. Shock 2020; 55:326-331. [PMID: 32694393 DOI: 10.1097/shk.0000000000001628] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To analyze factors associated with the development of early and late multiorgan failure (MOF) in trauma patients admitted to the intensive care unit (ICU). METHODS Spanish Trauma ICU Registry (RETRAUCI). Data collected from 52 trauma ICU between March 2015 and December 2019. We analyzed the incidence, outcomes, and the risk factors associated with early (< 72 h) or late (beyond 72 h) MOF in trauma ICU patients. Multiple logistic regression analysis was performed to analyze associated factors. RESULTS After excluding patients with incomplete data, 9,598 trauma ICU patients constituted the study population. Up to 965 patients (10.1%) presented with MOF, distributed by early MOF in 780 patients (8.1%) and late MOF in 185 patients (1.9%). The multivariate analysis showed that early MOF was associated with: ISS ≥ 16 (OR 2.80), hemodynamic instability (OR from 2.03 to 43.05), trauma-associated coagulopathy (OR 2.32), and acute kidney injury (OR 4.10). Late MOF was associated with: age > 65 years (OR 1.52), hemodynamic instability (OR from 1.92 to 9.94), acute kidney injury (OR 4.22), and nosocomial infection (OR 17.23). MOF was closely related to mortality (crude OR (95% CI) 4.77 (4.22-5.40)). CONCLUSIONS Multiorgan failure was recorded in 10% of trauma ICU patients, with early MOF being the predominant form. Early and late MOF forms were associated with different risk factors, suggesting different pathophysiological pathways. Early MOF was associated with higher severity of injury and severe bleeding-related complications and late MOF with advanced age and nosocomial infection.
Collapse
|
41
|
White AR, Werner CM, Holmes GM. Diminished enteric neuromuscular transmission in the distal colon following experimental spinal cord injury. Exp Neurol 2020; 331:113377. [PMID: 32526238 DOI: 10.1016/j.expneurol.2020.113377] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/14/2020] [Accepted: 06/03/2020] [Indexed: 12/30/2022]
Abstract
Neurogenic bowel following spinal cord injury (SCI) leads to decreased colonic motility, remodeling of the neuromuscular compartment and results in chronic evacuation difficulties. The distal colon of the rat serves a dual role for fluid absorption and storage that is homologous to the descending colon of humans. Dysmotility of the descending colon is one component of neurogenic bowel. We investigated the integrity of the enteric neuromuscular transmission responsible for the generation of excitatory and inhibitory junction potentials (EJPs and IJPs, respectively) in the distal colon of rats. We previously demonstrated a chronic reduction in colonic enteric neurons from rats with acute and chronic high-thoracic (T3) SCI and hypothesized that neurogenic bowel following T3-SCI results from diminished enteric neuromuscular transmission. Immunohistochemical labeling for myenteric neuronal nitric oxide synthase (nNOS) and choline acetyltransferase (ChAT) neurons demonstrated a significant loss of presumptive nitric oxide (NO) and acetylcholine (ACh) immunoreactive neurons in both 3-day and 3-week injured animals. Colonic neuromuscular transmission in response to transmural electrical stimulation of the colon was significantly reduced 3-days and 3-weeks following SCI in male rats. Specifically, cholinergic-mediated excitatory junction potentials (EJPs) and nitrergic-mediated slow inhibitory junction potentials (IJPs) were significantly reduced while ATP-mediated fast IJPs remained unaffected. We conclude that a reduction in excitatory and inhibitory enteric neuromuscular transmission contributes to neurogenic bowel observed following SCI, and that these loss-of-function changes involve enteric-mediated cholinergic and nitrergic pathways.
Collapse
Affiliation(s)
- Amanda R White
- Department of Neural and Behavioral Sciences, Penn State University College of Medicine, Hershey, PA 17033, United States of America
| | - Claire M Werner
- Department of Neural and Behavioral Sciences, Penn State University College of Medicine, Hershey, PA 17033, United States of America
| | - Gregory M Holmes
- Department of Neural and Behavioral Sciences, Penn State University College of Medicine, Hershey, PA 17033, United States of America.
| |
Collapse
|
42
|
Crimi E, Cirri S, Benincasa G, Napoli C. Epigenetics Mechanisms in Multiorgan Dysfunction Syndrome. Anesth Analg 2020; 129:1422-1432. [PMID: 31397699 DOI: 10.1213/ane.0000000000004331] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Epigenetic mechanisms including deoxyribonucleic acid (DNA) methylation, histone modifications (eg, histone acetylation), and microribonucleic acids (miRNAs) have gained much scientific interest in the last decade as regulators of genes expression and cellular function. Epigenetic control is involved in the modulation of inflammation and immunity, and its dysregulation can contribute to cell damage and organ dysfunction. There is growing evidence that epigenetic changes can contribute to the development of multiorgan dysfunction syndrome (MODS), a leading cause of mortality in the intensive care unit (ICU). DNA hypermethylation, histone deacetylation, and miRNA dysregulation can influence cytokine and immune cell expression and promote endothelial dysfunction, apoptosis, and end-organ injury, contributing to the development of MODS after a critical injury. Epigenetics processes, particularly miRNAs, are emerging as potential biomarkers of severity of disease, organ damage, and prognostic factors in critical illness. Targeting epigenetics modifications can represent a novel therapeutic approach in critical care. Inhibitors of histone deacetylases (HDCAIs) with anti-inflammatory and antiapoptotic activities represent the first class of drugs that reverse epigenetics modifications with human application. Further studies are required to acquire a complete knowledge of epigenetics processes, full understanding of their individual variability, to expand their use as accurate and reliable biomarkers and as safe target to prevent or attenuate MODS in critical disease.
Collapse
Affiliation(s)
- Ettore Crimi
- From the University of Central Florida, College of Medicine, Orlando, Florida.,Department of Anesthesiology and Critical Care Medicine, Ocala Health, Ocala, Florida
| | - Silvia Cirri
- Division of Anesthesiology and Intensive Care, Cardiothoracic Department, Istituto Clinico Sant'Ambrogio, Gruppo Ospedaliero San Donato, Milan, Italy
| | - Giuditta Benincasa
- Clinical Department of Internal Medicine and Specialistics, Department of Advanced Clinical and Surgical Sciences, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Claudio Napoli
- Clinical Department of Internal Medicine and Specialistics, Department of Advanced Clinical and Surgical Sciences, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy.,Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Foundation SDN, Naples, Italy
| |
Collapse
|
43
|
Smith S, Behrens B, McCully B, Murphy J, Bommiasamy A, Goodman A, Dewey E, Pati S, Schreiber M. Aggressive treatment of acute kidney injury and hyperkalemia improves survival in a combat relevant trauma model in swine. Am J Surg 2020; 219:860-864. [PMID: 32245610 DOI: 10.1016/j.amjsurg.2020.02.058] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 02/28/2020] [Accepted: 02/29/2020] [Indexed: 10/24/2022]
Abstract
INTRODUCTION Our swine model of pulmonary contusion (PC) and hemorrhagic shock (HS) was initially complicated by renal failure, hyperkalemia, and premature death. To study the effects of novel therapies on organ failure, improved survival was necessary requiring the design of an aggressive treatment regimen. METHODS Anesthetized swine sustained either PC or PC with grade V liver injury to induce HS (PC + HS). After injury, animals were resuscitated followed by either standard care (SC) with maintenance intravenous fluids (IVF) and treatment at potassium level of 6.0 mmol/L (n = 7; 3 PC, 4 PC + HS) or aggressive care (AC) with increased initial IVF, early and frequent potassium monitoring, and treatment at potassium level of 5.0 mmol/L (n = 15, 8 PC, 7 PC + HS). Hyperkalemia was treated with calcium, insulin, and glucose in both groups. RESULTS Survival to 48 h was achieved in 13/15 (87%) in the AC group and 2/7 (29%) in the SC group (p = 0.014). Compared to SC, AC improved median survival (48 vs. 18 h, p = 0.008) and lowered potassium (5.0 vs. 7.5 mmol/L), creatinine (2.4 vs. 4.0 mg/dL), BUN (27.5 vs. 39.0 mg/dL), and lactate (0.97 vs. 3.57 mmol/L) at the last observed time-point prior to death. For PC + HS animals, survival to 48 h was achieved in 6/7 in the AC group and 0/4 in the SC group with an improved median survival in the AC group (48 vs. 18 h, p = 0.011) DISCUSSION: Aggressive and early hyperkalemia treatment prolongs survival while reducing kidney injury and potassium levels in a combat relevant injury model in swine.
Collapse
Affiliation(s)
- Sawyer Smith
- Oregon Health & Science University, Portland, OR, USA.
| | | | | | - James Murphy
- Oregon Health & Science University, Portland, OR, USA
| | | | | | | | - Shibani Pati
- University of California San Francisco, San Francisco, CA, USA
| | | |
Collapse
|
44
|
Caspase-1-Dependent Pyroptosis of Peripheral Blood Mononuclear Cells Is Associated with the Severity and Mortality of Septic Patients. BIOMED RESEARCH INTERNATIONAL 2020; 2020:9152140. [PMID: 32258157 PMCID: PMC7066402 DOI: 10.1155/2020/9152140] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 01/17/2020] [Indexed: 02/07/2023]
Abstract
Purpose Pyroptosis has been known to play a vital role in the inflammation process which was induced by infection, injury, or inflammatory disease. The present study was aimed at evaluating the percentage of peripheral blood mononuclear cell (PBMC) pyroptosis in septic patients and assessing the correlation of PBMC pyroptosis with the severity and the mortality of septic patients. Methods 128 trauma-induced patients with sepsis were enrolled in this prospective cohort study. Blood samples were collected, and PBMC pyroptosis was measured by flow cytometry within 24 hours after sepsis was diagnosed. Results Percentage of PBMC pyroptosis was positively correlated with the acute physiology and chronic health evaluation (APACHE) II score and sequential organ failure assessment (SOFA) score (all P < 0.01). The area under the curve (AUC) for the percentage of PBMC pyroptosis on a receiver operating characteristic curve was 0.79 (95% confidence interval (CI), 0.68–0.90). A Cox proportional hazard model identified an association between an increased percentage of PBMC pyroptosis (>14.17%) and increased risk of the 28-day mortality (hazard ratio = 1.234, 95% CI, 1.014–1.502). Conclusion The percentage of PBMC pyroptosis increases in septic patients, and the increased percentage of PBMC pyroptosis is associated with the severity of sepsis and the 28-day mortality of patients with sepsis.
Collapse
|
45
|
Peters H, Macke C, Mommsen P, Zeckey C, Clausen JD, Krettek C, Neunaber C, Winkelmann M. Predictive Value of Osteoprotegerin and Neutrophil Gelatinase-associated Lipocalin on Multiple Organ Failure in Multiple Trauma. In Vivo 2020; 33:1573-1580. [PMID: 31471407 DOI: 10.21873/invivo.11639] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 07/01/2019] [Accepted: 07/02/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND/AIM Multiple organ dysfunction syndrome (MODS) is the leading cause of late posttraumatic mortality. This study analyzed the prognostic values of osteoprotegerin (OPG) and neutrophil gelatinase-associated lipocalin (NGAL/lipocalin 2) compared to interleukin-6 (IL-6) in multiply injured patients. PATIENTS AND METHODS A retrospective observational cohort study on multiply injured patients with an injury severity score (ISS) of ≥16 was performed. OPG, NGAL and IL-6 blood concentrations were measured. Statistical analysis comprised receiver-operating-characteristic (ROC) analysis with the corresponding area under the curve (AUC). RESULTS Thirty-nine patients with a mean ISS of 34±11 were included. Fourteen patients (36%) developed MODS and 8 patients (21%) died. Plasma levels of NGAL, OPG, and IL-6 were significantly elevated in the MODS+ group. Each biomarker positively correlated with MODS score and diagnosis of MODS. CONCLUSION NGAL and OPG might be indicative of MODS and could have the potential to be biomarkers in the early detection of patients at risk of posttraumatic MODS.
Collapse
Affiliation(s)
- Henning Peters
- Trauma Department, Hannover Medical School, Hannover, Germany
| | - Christian Macke
- Trauma Department, Hannover Medical School, Hannover, Germany
| | - Philipp Mommsen
- Trauma Department, Hannover Medical School, Hannover, Germany
| | - Christian Zeckey
- Trauma Department, Hannover Medical School, Hannover, Germany.,Department of General, Trauma and Reconstructive Surgery, Ludwig-Maximilians-Universität München, Munich, Germany
| | | | | | | | | |
Collapse
|
46
|
Culver A, Coiffard B, Antonini F, Duclos G, Hammad E, Vigne C, Mege JL, Baumstarck K, Boucekine M, Zieleskiewicz L, Leone M. Circadian disruption of core body temperature in trauma patients: a single-center retrospective observational study. J Intensive Care 2020; 8:4. [PMID: 31921428 PMCID: PMC6945723 DOI: 10.1186/s40560-019-0425-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 12/29/2019] [Indexed: 01/14/2023] Open
Abstract
Background Circadian clock alterations were poorly reported in trauma patients, although they have a critical role in human physiology. Core body temperature is a clinical variable regulated by the circadian clock. Our objective was to identify the circadian temperature disruption in trauma patients and to determine whether these disruptions were associated with the 28-day mortality rate. Methods A retrospective and observational single-center cohort study was conducted. All adult severe trauma patients admitted to the intensive care unit of Aix Marseille University, North Hospital, from November 2013 to February 2018, were evaluated. The variations of core body temperature for each patient were analyzed between days 2 and 3 after intensive care unit admission. Core body temperature variations were defined by three parameters: mesor, amplitude, and period. A logistic regression model was used to determine the variables influencing these three parameters. A survival analysis was performed assessing the association between core body temperature rhythm disruption and 28-day mortality rate. A post hoc subgroup analysis focused on the patients with head trauma. Results Among the 1584 screened patients, 248 were included in this study. The period differed from 24 h in 177 (71%) patients. The mesor value (°C) was associated with body mass index and ketamine use. Amplitude (°C) was associated with ketamine use only. The 28-day mortality rate was 18%. For all trauma patients, age, body mass index, intracranial hypertension, and amplitude were independent risk factors. The patients with a mesor value < 36.9 °C (p < 0.001) and an amplitude > 0.6 °C (p < 0.001) had a higher 28-day mortality rate. Among the patients with head trauma, mesor and amplitude were identified as independent risk factors (HR = 0.40, 95% CI [0.23–0.70], p = 0.001 and HR = 4.73, 95% CI [1.38–16.22], p = 0.01). Conclusions Our results highlight an association between core body temperature circadian alteration and 28-day mortality rate. This association was more pronounced in the head trauma patients than in the non-head trauma patients. Further studies are needed to show a causal link and consider possible interventions.
Collapse
Affiliation(s)
- Aurélien Culver
- Service d'Anesthésie et de Réanimation, APHM, Hôpital Nord, Aix Marseille Université, Chemin des Bourrely, 13915 Marseille, France
| | - Benjamin Coiffard
- Médecine Intensive - Réanimation, APHM, Hôpital Nord, Aix Marseille Université, Marseille, France.,3CNRS, IRD, MEPHI, IHU Méditerranée Infection, Aix Marseille Université, Marseille, France
| | - François Antonini
- Service d'Anesthésie et de Réanimation, APHM, Hôpital Nord, Aix Marseille Université, Chemin des Bourrely, 13915 Marseille, France
| | - Gary Duclos
- Service d'Anesthésie et de Réanimation, APHM, Hôpital Nord, Aix Marseille Université, Chemin des Bourrely, 13915 Marseille, France
| | - Emmanuelle Hammad
- Service d'Anesthésie et de Réanimation, APHM, Hôpital Nord, Aix Marseille Université, Chemin des Bourrely, 13915 Marseille, France
| | - Coralie Vigne
- Service d'Anesthésie et de Réanimation, APHM, Hôpital Nord, Aix Marseille Université, Chemin des Bourrely, 13915 Marseille, France
| | - Jean-Louis Mege
- 3CNRS, IRD, MEPHI, IHU Méditerranée Infection, Aix Marseille Université, Marseille, France
| | - Karine Baumstarck
- 4APHM, EA 3279 CEReSS, School of Medicine - La Timone Medical Campus, Health Service Research and Quality of Life Center, Aix Marseille Université, Marseille, France
| | - Mohamed Boucekine
- 4APHM, EA 3279 CEReSS, School of Medicine - La Timone Medical Campus, Health Service Research and Quality of Life Center, Aix Marseille Université, Marseille, France
| | - Laurent Zieleskiewicz
- Service d'Anesthésie et de Réanimation, APHM, Hôpital Nord, Aix Marseille Université, Chemin des Bourrely, 13915 Marseille, France
| | - Marc Leone
- Service d'Anesthésie et de Réanimation, APHM, Hôpital Nord, Aix Marseille Université, Chemin des Bourrely, 13915 Marseille, France.,3CNRS, IRD, MEPHI, IHU Méditerranée Infection, Aix Marseille Université, Marseille, France
| |
Collapse
|
47
|
Prasad GK, Khanna S, Sharma V. Peripheral nerve blocks in trauma patients: Recent updates and improving patient outcomes: A narrative review. INDIAN JOURNAL OF PAIN 2020. [DOI: 10.4103/ijpn.ijpn_70_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
48
|
Goodus MT, McTigue DM. Hepatic dysfunction after spinal cord injury: A vicious cycle of central and peripheral pathology? Exp Neurol 2019; 325:113160. [PMID: 31863731 DOI: 10.1016/j.expneurol.2019.113160] [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: 07/07/2019] [Revised: 11/17/2019] [Accepted: 12/18/2019] [Indexed: 02/06/2023]
Abstract
The liver is essential for numerous physiological processes, including filtering blood from the intestines, metabolizing fats, proteins, carbohydrates and drugs, and regulating iron storage and release. The liver is also an important immune organ and plays a critical role in response to infection and injury throughout the body. Liver functions are regulated by autonomic parasympathetic innervation from the brainstem and sympathetic innervation from the thoracic spinal cord. Thus, spinal cord injury (SCI) at or above thoracic levels disrupts major regulatory mechanisms for hepatic functions. Work in rodents and humans shows that SCI induces liver pathology, including hepatic inflammation and fat accumulation characteristic of a serious form of non-alcoholic fatty liver disease (NAFLD) called non-alcoholic steatohepatitis (NASH). This hepatic pathology is associated with and likely contributes to indices of metabolic dysfunction often noted in SCI individuals, such as insulin resistance and hyperlipidemia. These occur at greater rates in the SCI population and can negatively impact health and quality of life. In this review, we will: 1) Discuss acute and chronic changes in human and rodent liver pathology and function after SCI; 2) Describe how these hepatic changes affect systemic inflammation, iron regulation and metabolic dysfunction after SCI; 3) Describe how disruption of the hepatic autonomic nervous system may be a key culprit in post-injury chronic liver pathology; and 4) Preview ongoing and future research that aims to elucidate mechanisms driving liver and metabolic dysfunction after SCI.
Collapse
Affiliation(s)
- Matthew T Goodus
- The Belford Center for Spinal Cord Injury, The Ohio State University, Columbus, OH, USA; Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH, USA.
| | - Dana M McTigue
- The Belford Center for Spinal Cord Injury, The Ohio State University, Columbus, OH, USA; Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
49
|
Ogura A, Tsurumi A, Que YA, Almpani M, Zheng H, Tompkins RG, Ryan CM, Rahme LG. Associations between clinical characteristics and the development of multiple organ failure after severe burns in adult patients. Burns 2019; 45:1775-1782. [PMID: 31690472 DOI: 10.1016/j.burns.2019.02.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 01/07/2019] [Accepted: 02/14/2019] [Indexed: 11/24/2022]
Abstract
To determine the association between potential risk factors and multiple organ failure (MOF) in severe burn adult patients, we performed a secondary analysis of data from the "Inflammation and the Host Response to Injury" database, which included patients from six burn centers in the United States between 2003 and 2009. Three hundred twenty-two adult patients (aged ≥16 years) with severe burns (≥20.0% total body surface area [TBSA]) were included. MOF was defined according to the Denver score. Potential risk factors were analyzed for their association with MOF. Models were built using multivariable logistic regression analysis. Eighty-eight patients (27.3%) developed MOF during the study period. We found that TBSA, age, and inhalation injury were significant risk factors for MOF. This predictive model showed good performance, with the total area under the receiver operating characteristic curve being 0.823. Moreover, among patients who developed MOF, inhalation injury was significantly associated with the development of MOF in the acute phase (within three days of injury) (adjusted odds ratio 3.1; 95% confidence interval 1.1-8.3). TBSA, age, lactate, and Denver score within 24h were associated with the late phase development of MOF. Thus, we have identified key risk factors for the onset of MOF after severe burn injury. Our findings contribute to the understanding of individualized treatment and will potentially allow for efficient allocation of resources and a lower threshold for admission to an intensive care unit, which can prevent the development of MOF and eventually reduce mortality.
Collapse
Affiliation(s)
- Asako Ogura
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, 340 Thier Research Building, 50 Blossom Street, Boston MA 02114, USA
| | - Amy Tsurumi
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, 340 Thier Research Building, 50 Blossom Street, Boston MA 02114, USA; Shriners Hospitals for Children, 51 Blossom St., Boston, MA 02114, USA
| | - Yok-Ai Que
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Freiburgstrasse 18, 3010 Bern, Switzerland
| | - Marianna Almpani
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, 340 Thier Research Building, 50 Blossom Street, Boston MA 02114, USA; Shriners Hospitals for Children, 51 Blossom St., Boston, MA 02114, USA
| | - Hui Zheng
- Biostatistics Center, Massachusetts General Hospital, and Harvard Medical School, 50 Staniford St., Boston, MA 02114, USA
| | - Ronald G Tompkins
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, 340 Thier Research Building, 50 Blossom Street, Boston MA 02114, USA
| | - Colleen M Ryan
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, 340 Thier Research Building, 50 Blossom Street, Boston MA 02114, USA; Shriners Hospitals for Children, 51 Blossom St., Boston, MA 02114, USA
| | - Laurence G Rahme
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, 340 Thier Research Building, 50 Blossom Street, Boston MA 02114, USA; Shriners Hospitals for Children, 51 Blossom St., Boston, MA 02114, USA; Department of Microbiology and Immunobiology, Harvard Medical School, 77 Ave. Louis Pasteur, Boston, MA 02114, USA.
| |
Collapse
|
50
|
Torres Filho IP, Barraza D, Hildreth K, Williams C, Dubick MA. Cremaster muscle perfusion, oxygenation, and heterogeneity revealed by a new automated acquisition system in a rodent model of prolonged hemorrhagic shock. J Appl Physiol (1985) 2019; 127:1548-1561. [PMID: 31670599 DOI: 10.1152/japplphysiol.00570.2019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Local blood flow/oxygen partial pressure (Po2) distributions and flow-Po2 relationships are physiologically relevant. They affect the pathophysiology and treatment of conditions like hemorrhagic shock (HS), but direct noninvasive measures of flow, Po2, and their heterogeneity during prolonged HS are infrequently presented. To fill this void, we report the first quantitative evaluation of flow-Po2 relationships and heterogeneities in normovolemia and during several hours of HS using noninvasive, unbiased, automated acquisition. Anesthetized rats were subjected to tracheostomy, arterial/venous catheterizations, cremaster muscle exteriorization, hemorrhage (40% total blood volume), and laparotomy. Control animals equally instrumented were not subjected to hemorrhage/laparotomy. Every 0.5 h for 4.5 h, noninvasive laser speckle contrast imaging and phosphorescence quenching were employed for nearly 7,000 flow/Po2 measurements in muscles from eight animals, using an automated system. Precise alignment of 16 muscle areas allowed overlapping between flow and oxygenation measurements to evaluate spatial heterogeneity, and repeated measurements were used to estimate temporal heterogeneity. Systemic physiological parameters and blood chemistry were simultaneously assessed by blood samplings replaced with crystalloids. Hemodilution was associated with local hypoxia, but increased flow prevented major oxygen delivery decline. Adding laparotomy and prolonged HS resulted in hypoxia, ischemia, decreased tissue oxygen delivery, and logarithmic flow/Po2 relationships in most regions. Flow and Po2 spatial heterogeneities were higher than their respective temporal heterogeneities, although this did not change significantly over the studied period. This quantitative framework establishes a basis for evaluating therapies aimed at restoring muscle homeostasis, positively impacting outcomes of civilian and military trauma/HS victims.NEW & NOTEWORTHY This is the first study on flow-Po2 relationships during normovolemia, hemodilution, and prolonged hemorrhagic shock using noninvasive methods in multiple skeletal muscle areas of monitored animals. Automated flow/Po2 measurements revealed temporal/spatial heterogeneities, hypoxia, ischemia, and decreased tissue oxygen delivery after trauma/severe hemorrhage. Hemodilution was associated with local hypoxia, but hyperemia prevented a major decline in oxygen delivery. This framework provides a quantitative basis for testing therapeutics that positively impacts muscle homeostasis and outcomes of trauma/hemorrhagic shock victims.
Collapse
Affiliation(s)
- Ivo P Torres Filho
- Damage Control Resuscitation, United States Army Institute of Surgical Research, JBSA Fort Sam Houston, Texas
| | - David Barraza
- Damage Control Resuscitation, United States Army Institute of Surgical Research, JBSA Fort Sam Houston, Texas
| | - Kim Hildreth
- Damage Control Resuscitation, United States Army Institute of Surgical Research, JBSA Fort Sam Houston, Texas
| | - Charnae Williams
- Damage Control Resuscitation, United States Army Institute of Surgical Research, JBSA Fort Sam Houston, Texas
| | - Michael A Dubick
- Damage Control Resuscitation, United States Army Institute of Surgical Research, JBSA Fort Sam Houston, Texas
| |
Collapse
|