1
|
Khalaf F, Touma D, Pappas A, Hatim L, Wojtowicz-Piotrowski S, Jeschke MG. Decoding burn trauma: biomarkers for early diagnosis of burn-induced pathologies. Biomark Res 2024; 12:160. [PMID: 39716257 DOI: 10.1186/s40364-024-00707-5] [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: 09/22/2024] [Accepted: 12/09/2024] [Indexed: 12/25/2024] Open
Abstract
Burn injuries represent a significant global challenge due to their multifaceted nature, characterized by a complex cascade of metabolic and immune dysfunction that can result in severe complications. If not identified and managed promptly, these complications can escalate, often leading to fatal outcomes. This underscores the critical importance of timely and precise diagnosis. Fortunately, biomarkers for burn-induced pathologies and outcomes have emerged as powerful diagnostic and prognostic tools. These biomarkers enable early diagnosis and intervention, facilitate risk assessment, support patient-specific treatment, monitoring of disease progression, and therapeutic efficacy, ultimately contributing to improved patient outcomes. However, while previous studies have provided valuable biomarkers for the detection of burn-induced pathologies, many of these were constrained by the techniques and sample sizes available at the time, which can limit the generalizability of the findings. This review highlights numerous biomarkers studied in the literature to date, underscoring the need to replicate these findings in more diverse and representative populations. It also emphasizes the importance of advancing research efforts to develop more efficient, accurate, and cost-effective approaches for integrating biomarkers into clinical practice.
Collapse
Affiliation(s)
- Fadi Khalaf
- David Braley Research Institute, Hamilton, ON, Canada
- Hamilton Health Sciences, Hamilton, ON, Canada
- Department of Biochemistry, McMaster University, Hamilton, ON, Canada
| | - Daniella Touma
- David Braley Research Institute, Hamilton, ON, Canada
- Hamilton Health Sciences, Hamilton, ON, Canada
| | - Alexandra Pappas
- David Braley Research Institute, Hamilton, ON, Canada
- Hamilton Health Sciences, Hamilton, ON, Canada
| | - Lareina Hatim
- David Braley Research Institute, Hamilton, ON, Canada
- Hamilton Health Sciences, Hamilton, ON, Canada
| | - Stephanie Wojtowicz-Piotrowski
- David Braley Research Institute, Hamilton, ON, Canada
- Hamilton Health Sciences, Hamilton, ON, Canada
- Department of Surgery, McMaster University, Hamilton, ON, Canada
| | - Marc G Jeschke
- David Braley Research Institute, Hamilton, ON, Canada.
- Hamilton Health Sciences, Hamilton, ON, Canada.
- Department of Biochemistry, McMaster University, Hamilton, ON, Canada.
- Department of Surgery, McMaster University, Hamilton, ON, Canada.
- David Braley Research Institute, C5-104, 20 Copeland Ave, Hamilton, ON, L8L 2X2, Canada.
| |
Collapse
|
2
|
Meza Monge K, Rosa C, Sublette C, Pratap A, Kovacs EJ, Idrovo JP. Navigating Hemorrhagic Shock: Biomarkers, Therapies, and Challenges in Clinical Care. Biomedicines 2024; 12:2864. [PMCID: PMC11673713 DOI: 10.3390/biomedicines12122864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/13/2024] [Accepted: 12/15/2024] [Indexed: 01/04/2025] Open
Abstract
Hemorrhagic shock remains a leading cause of preventable death worldwide, with mortality patterns varying significantly based on injury mechanisms and severity. This comprehensive review examines the complex pathophysiology of hemorrhagic shock, focusing on the temporal evolution of inflammatory responses, biomarker utility, and evidence-based therapeutic interventions. The inflammatory cascade progresses through distinct phases, beginning with tissue injury and endothelial activation, followed by a systemic inflammatory response that can transition to devastating immunosuppression. Recent advances have revealed pattern-specific responses between penetrating and blunt trauma, necessitating tailored therapeutic approaches. While damage control resuscitation principles and balanced blood product administration have improved outcomes, many molecular targeted therapies remain investigational. Current evidence supports early hemorrhage control, appropriate blood product ratios, and time-sensitive interventions like tranexamic acid administration. However, challenges persist in biomarker validation, therapeutic timing, and implementation of personalized treatment strategies. Future directions include developing precision medicine approaches, real-time monitoring systems, and novel therapeutic modalities while addressing practical implementation barriers across different healthcare settings. Success in hemorrhagic shock management increasingly depends on integrating multiple interventions across different time points while maintaining focus on patient-centered outcomes.
Collapse
Affiliation(s)
- Kenneth Meza Monge
- Department of Surgery, Division of G.I, Trauma, and Endocrine Surgery, University of Colorado, Aurora, CO 80045, USA; (K.M.M.); (C.R.); (C.S.); (A.P.); (E.J.K.)
| | - Caleb Rosa
- Department of Surgery, Division of G.I, Trauma, and Endocrine Surgery, University of Colorado, Aurora, CO 80045, USA; (K.M.M.); (C.R.); (C.S.); (A.P.); (E.J.K.)
| | - Christopher Sublette
- Department of Surgery, Division of G.I, Trauma, and Endocrine Surgery, University of Colorado, Aurora, CO 80045, USA; (K.M.M.); (C.R.); (C.S.); (A.P.); (E.J.K.)
| | - Akshay Pratap
- Department of Surgery, Division of G.I, Trauma, and Endocrine Surgery, University of Colorado, Aurora, CO 80045, USA; (K.M.M.); (C.R.); (C.S.); (A.P.); (E.J.K.)
| | - Elizabeth J. Kovacs
- Department of Surgery, Division of G.I, Trauma, and Endocrine Surgery, University of Colorado, Aurora, CO 80045, USA; (K.M.M.); (C.R.); (C.S.); (A.P.); (E.J.K.)
- Department of Immunology and Microbiology, University of Colorado, Aurora, CO 80045, USA
| | - Juan-Pablo Idrovo
- Department of Surgery, Division of G.I, Trauma, and Endocrine Surgery, University of Colorado, Aurora, CO 80045, USA; (K.M.M.); (C.R.); (C.S.); (A.P.); (E.J.K.)
| |
Collapse
|
3
|
Keyloun JW, Kelly EJ, Carney BC, Nisar S, Kolachana S, Moffatt LT, Orfeo T, Shupp JW. Plasmas From Patients With Burn Injury Induce Endotheliopathy Through Different Pathways. J Surg Res 2024; 304:90-100. [PMID: 39531984 DOI: 10.1016/j.jss.2024.10.011] [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: 04/30/2024] [Revised: 09/19/2024] [Accepted: 10/13/2024] [Indexed: 11/16/2024]
Abstract
INTRODUCTION The contribution of endothelial injury to the pathogenesis of burn shock is not well characterized. This work investigates potential mechanisms underlying dysregulation of endothelial barrier function by burn patient plasmas. METHODS Plasma was collected from burn-injured patients (n = 8) within 4 h of admission. Demographic and injury characteristics were collected and markers of injury severity including international normalized ratio, activated partial thromboplastin time, and levels of syndecan-1 and interleukin (IL)-6, IL-1B, IL-10, Il-12p70, and tumor necrosis factor-α measured. Human umbilical vein endothelial cell monolayers (HUVEC-m) exposed to either burn patient plasma or multidonor plasma (HHP) were assessed for permeability (40 kDa fluorescein isothiocyanate (FITC)-Dextran diffusion), intercellular gap area (F-actin staining) and incidence of apoptosis (TUNEL assay). Post plasma exposure, RNA was isolated and used in polymerase chain reaction (PCR) arrays targeting genes relevant to cytoskeletal structure or apoptosis. Differences between HHP and burn plasma-treated HUVEC-m were analyzed. RESULTS Five plasmas promoted significant increases in HUVEC-m permeability. When plasmas were grouped based on their capacity to increase permeability, no differences in age, %total body surface area, gender, hospital mortality, international normalized ratio, activated partial thromboplastin time, or cytokine concentration were observed; however, significantly higher syndecan-1 levels were seen in the plasmas inducing increased permeability. Increases in intercellular gap area and apoptosis and relevant gene expression were observed after exposure to patient plasmas but none of these metrics correlated completely with the pattern or magnitude of the changes in permeability. CONCLUSIONS Burn patient plasmas variably disrupt HUVEC-m homeostasis, differentially inducing changes in permeability, intercellular gap area, and apoptosis. Neither increases in intercellular gap size nor apoptosis appear sufficient to explain the pattern of changes in permeability.
Collapse
Affiliation(s)
- John W Keyloun
- The Burn Center, Department of Surgery, MedStar Washington Hospital Center, Washington, District of Columbia; Firefighters' Burn and Surgical Research Laboratory, MedStar Health Research Institute, Washington, District of Columbia
| | - Edward J Kelly
- The Burn Center, Department of Surgery, MedStar Washington Hospital Center, Washington, District of Columbia; Firefighters' Burn and Surgical Research Laboratory, MedStar Health Research Institute, Washington, District of Columbia
| | - Bonnie C Carney
- Firefighters' Burn and Surgical Research Laboratory, MedStar Health Research Institute, Washington, District of Columbia; Departments of Surgery and Biochemistry, Georgetown University School of Medicine, Washington, District of Columbia
| | - Saira Nisar
- Firefighters' Burn and Surgical Research Laboratory, MedStar Health Research Institute, Washington, District of Columbia
| | - Sindhura Kolachana
- Georgetown University School of Medicine, Washington, District of Columbia
| | - Lauren T Moffatt
- Firefighters' Burn and Surgical Research Laboratory, MedStar Health Research Institute, Washington, District of Columbia; Departments of Surgery and Biochemistry, Georgetown University School of Medicine, Washington, District of Columbia
| | - Thomas Orfeo
- Department of Biochemistry, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Jeffrey W Shupp
- The Burn Center, Department of Surgery, MedStar Washington Hospital Center, Washington, District of Columbia; Firefighters' Burn and Surgical Research Laboratory, MedStar Health Research Institute, Washington, District of Columbia; Departments of Surgery and Biochemistry, Georgetown University School of Medicine, Washington, District of Columbia.
| |
Collapse
|
4
|
Matsumoto H, Annen S, Mukai N, Ohshita M, Ogawa S, Okita M, Tanabe T, Takezawa M, Nakabayashi Y, Kikuchi S, Takeba J, Sato N. Association of endotheliopathy with coagulofibrinolytic reactions and disseminated intravascular coagulation after trauma: a retrospective observational study. Sci Rep 2024; 14:29630. [PMID: 39609563 PMCID: PMC11604942 DOI: 10.1038/s41598-024-81123-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 11/25/2024] [Indexed: 11/30/2024] Open
Abstract
We carried out a retrospective observational investigation to explore the association of endotheliopathy with coagulofibrinolytic reactions and the progression of disseminated intravascular coagulation (DIC) in adult trauma patients. We measured syndecan-1 (SDC-1), an indicator of endotheliopathy, and biomarkers of coagulofibrinolysis in 100 trauma patients immediately transferred to Ehime University Hospital. We evaluated the correlations between the coagulofibrinolytic parameters and SDC-1. We also investigated the association between SDC-1 elevations and the development of DIC, and determined the discriminators of DIC development. The median SDC-1 concentration was 82.7 (43.5-178.1) ng/mL. DIC developed in 16 patients (16.0%), and SDC-1 concentrations were significantly higher in DIC patients than in non-DIC patients (218.8 [134.5-798.2] ng/mL vs. 67.2 [39.6-114.5] ng/mL, p < 0.001). Receiver operating characteristic curve analysis revealed that the circulating SDC-1 level effectively predicted the progression of DIC, with an area under the curve of 0.862 (95% confidence interval [CI], 0.789-0.936). The optimal cut-off value was determined to be 92.5 ng/mL, yielding a sensitivity of 100.0% and a specificity of 67.8% (p < 0.001). A simple logistic regression analysis showed that a circulating SDC-1 concentration of > 92.5 ng/mL was significantly correlated with DIC progression (odds ratio [OR], 31.67; 95%CI: 3.97-252.31, p = 0.001). Many coagulofibrinolytic parameters were significantly correlated with SDC-1. Estimating the discriminators of DIC development by the least absolute shrinkage and selection operator (LASSO) and elastic-net regression analysis identified markers of coagulofibrinolytic activation, such as thrombin-antithrombin complex (TAT) and tissue plasminogen activator (tPA). A multivariate logistic regression model using TAT, tPA, and SDC-1 demonstrated that TAT and tPA, but not SDC-1, were independent factors predicting the development of DIC (TAT per 10 µg/L: OR, 1.14, 95%CI: 1.05-1.24, p = 0.003; tPA per 100pg/mL: OR, 1.03, 95%CI: 1.01-1.05, p = 0.003; SDC-1 per 10ng/mL: OR, 1.00, 95%CI: 0.99-1.01, p = 0.973). Mediation analysis showed that SDC-1 elevation was predominantly associated with the development of DIC indirectly through the increase in TAT (proportion mediated = 96.1%, p < 0.001), while there was no significant indirect effect of SDC-1 elevation on the role of TAT elevation in DIC development was observed (p = 0.340). The primary pathogenesis of DIC in the acute phase of trauma is likely driven by coagulofibrinolytic activation. Endotheliopathy, as reflected by elevated circulating levels of SDC-1, is strongly associated with coagulofibrinolytic responses. Although endotheliopathy may contribute to the early development of DIC through coagulation activation, its role appears to be limited.
Collapse
Affiliation(s)
- Hironori Matsumoto
- Department of Emergency and Critical Care Medicine, Ehime University Graduate School of Medicine, Shitsukawa 454, Toon City, Ehime, 791-0295, Japan.
| | - Suguru Annen
- Department of Emergency and Critical Care Medicine, Ehime University Graduate School of Medicine, Shitsukawa 454, Toon City, Ehime, 791-0295, Japan
| | - Naoki Mukai
- Department of Emergency and Critical Care Medicine, Ehime University Graduate School of Medicine, Shitsukawa 454, Toon City, Ehime, 791-0295, Japan
| | - Muneaki Ohshita
- Department of Emergency and Critical Care Medicine, Ehime University Graduate School of Medicine, Shitsukawa 454, Toon City, Ehime, 791-0295, Japan
| | - Shirou Ogawa
- Department of Emergency and Critical Care Medicine, Ehime University Graduate School of Medicine, Shitsukawa 454, Toon City, Ehime, 791-0295, Japan
| | - Mitsuo Okita
- Department of Emergency and Critical Care Medicine, Ehime University Graduate School of Medicine, Shitsukawa 454, Toon City, Ehime, 791-0295, Japan
| | - Tsunenori Tanabe
- Department of Emergency and Critical Care Medicine, Ehime University Graduate School of Medicine, Shitsukawa 454, Toon City, Ehime, 791-0295, Japan
| | - Mitsuaki Takezawa
- Department of Emergency and Critical Care Medicine, Ehime University Graduate School of Medicine, Shitsukawa 454, Toon City, Ehime, 791-0295, Japan
| | - Yuki Nakabayashi
- Department of Emergency and Critical Care Medicine, Ehime University Graduate School of Medicine, Shitsukawa 454, Toon City, Ehime, 791-0295, Japan
| | - Satoshi Kikuchi
- Department of Emergency and Critical Care Medicine, Ehime University Graduate School of Medicine, Shitsukawa 454, Toon City, Ehime, 791-0295, Japan
| | - Jun Takeba
- Department of Emergency and Critical Care Medicine, Ehime University Graduate School of Medicine, Shitsukawa 454, Toon City, Ehime, 791-0295, Japan
| | - Norio Sato
- Department of Emergency and Critical Care Medicine, Ehime University Graduate School of Medicine, Shitsukawa 454, Toon City, Ehime, 791-0295, Japan
| |
Collapse
|
5
|
Patterson PD, Hilditch CJ, Martin SE, Roach DGL, Weaver MD, Okerman TS, Hostler D, Weiss LS, Reis SE. Comparison of 45-min nap versus no-nap during simulated night shift work on endothelial function: a randomized crossover feasibility trial. Pilot Feasibility Stud 2024; 10:137. [PMID: 39533414 PMCID: PMC11555913 DOI: 10.1186/s40814-024-01569-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Night shift workers face increased risk of cardiovascular disease (CVD) compared to non-shift workers. Evidence supports on-shift napping and regular non-invasive monitoring of endothelial function for risk mitigation, yet neither strategy is widely used. METHODS We evaluated the feasibility of non-invasive assessment of peripheral arterial tone (PAT) to assess the effect of napping during simulated night shift work on endothelial function. We used a single-site, randomized crossover trial of simulated night shift work with a 45-min nap condition versus a control, no-nap condition (ClinicalTrials.gov NCT05436951). RESULTS The primary outcome was the number of participants with ≥ 70% of endothelial function assessments. Secondary outcomes included mean reactive hyperemia index (RHI), BP, and cognitive performance with the brief psychomotor vigilance task (PVT-B). Of the 10 consented, 9 completed both conditions. All participants exceeded feasibility benchmarks. Mean RHI did not differ by nap condition, and the delta from pre- to post measure did not differ (difference in delta = - 0.26, 95% CI - 1.09, 0.58). Hourly PVT-B assessments from 19:00 to 07:00 h did not differ by nap condition. Compared to pre-nap measures, cognitive performance on the PVT-B was poorest at + 0 min post-nap. CONCLUSION Our findings can inform larger studies evaluating the effects of night shift work and napping on endothelial function. TRIAL REGISTRATIONS ClinicalTrials.gov (NCT05436951, registered on June 23, 2022).
Collapse
Affiliation(s)
- P Daniel Patterson
- Department of Emergency Medicine, School of Medicine, University of Pittsburgh, PA, Pittsburgh, 15261, USA.
- Emergency Medicine Program, Department of Community Health Services and Rehabilitation Science, School of Health and Rehabilitation Sciences, University of Pittsburgh, PA, Pittsburgh, 15261, USA.
| | - Cassie J Hilditch
- Fatigue Countermeasures Laboratory, San José State University, CA, San José, 95192, USA
| | - Sarah E Martin
- Department of Emergency Medicine, School of Medicine, University of Pittsburgh, PA, Pittsburgh, 15261, USA
| | - David G L Roach
- Department of Emergency Medicine, School of Medicine, University of Pittsburgh, PA, Pittsburgh, 15261, USA
| | - Matthew D Weaver
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, MA, Boston, 02115, USA
- Division of Sleep Medicine, Harvard Medical School, MA, Boston, 02115, USA
| | - Tiffany S Okerman
- Department of Emergency Medicine, School of Medicine, University of Pittsburgh, PA, Pittsburgh, 15261, USA
| | - David Hostler
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, University at Buffalo, NY, Buffalo, 14214, USA
| | - Leonard S Weiss
- Department of Emergency Medicine, School of Medicine, University of Pittsburgh, PA, Pittsburgh, 15261, USA
| | - Steven E Reis
- Department of Medicine, University of Pittsburgh, PA, Pittsburgh, 15261, USA
| |
Collapse
|
6
|
Braunstein M, Annecke T, Frey K, Kusmenkov T, Wörnle M, Ney L, Böcker W, Bogner-Flatz V. Effect on Syndecan-1 and Hyaluronan Levels Depending on Multiple Organ Failure, Coagulopathy and Survival: An Observational Study in Major Trauma Patients. J Clin Med 2024; 13:6768. [PMID: 39597912 PMCID: PMC11595190 DOI: 10.3390/jcm13226768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 10/25/2024] [Accepted: 11/07/2024] [Indexed: 11/29/2024] Open
Abstract
Background: Major trauma, as well as traumatic hemorrhagic shock go along with early damage to the endothelial glycocalyx (EG). Shed glycocalyx constituents can activate the innate immune system and aggravate secondary injury. Subsequently, we investigated the relationship between glycocalyx shedding and the occurrence of coagulopathy, multiple organ failure (MOF) and outcome in our cohort after severe trauma. Methods: We included multiple trauma patients, as defined by Injury Severity Score (ISS). Polytraumatized patients must have arrived in our level 1 trauma center within 60 min after trauma. Retrospectively, patients were assigned to predefined clinical conditions, based on injury severity (ISS ≥ 16 points), multiple organ failure (MOF score ≥ 6 points), need for massive transfusion (≥10 RBC units/first 24 h), coagulopathy (prothrombin time < 70% at 0 h) and survival (90-day survival). Syndecan-1 (Sdc-1) and hyaluronan (HA) plasma concentrations were evaluated immediately (0 h), 6 h and 12 h after trauma. Results: 49 patients (mean ISS 35.7 ± 12.1 SD, mean age 45.78 ± 15.6 SD) were included in this study. A total of 37 patients (75.5%) survived, while 12 patients died within the observation period of 90 days after trauma (24.5%). A total of 77% of all patients suffered multiple organ failure (MOF score ≥ 6, n = 30). Initial prothrombin time at 0 h was <70% in 31 patients. Plasma concentrations of circulating both glycocalyx constituents showed a significant increase over the first 12 h after trauma (p = 0.001; p = 0.008). Patients with multiple organ failure showed significantly increased hyaluronan concentrations at all three time points (p = 0.007/0.006/<0.001), and the syndecan-1 levels were significantly elevated 12 h after trauma in the MOF group (p = 0.01). Patients with coagulopathy on admission exhibited significantly higher hyaluronan levels at 12 h (p = 0.042). Non-survivors showed significantly increased syndecan-1 levels at 12 h after trauma (p = 0.024). Conclusions: Glycocalyx shedding occurs immediately after major trauma. Coagulopathy is associated with significantly increased plasma hyaluronan. Further, significant changes in plasma concentrations within the first 12 h help to identify subgroups at risk for developing MOF and death.
Collapse
Affiliation(s)
- Mareen Braunstein
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Centre Munich (MUM), University Hospital, LMU Munich, Ziemssenstraße 5, 80336 Munich, Germany; (K.F.); (W.B.); (V.B.-F.)
| | - Thorsten Annecke
- Department of Anaesthesiology and Critical Care Medicine, Merheim Medical Centre, University of Witten/Herdecke, 51109 Cologne, Germany;
| | - Kathrin Frey
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Centre Munich (MUM), University Hospital, LMU Munich, Ziemssenstraße 5, 80336 Munich, Germany; (K.F.); (W.B.); (V.B.-F.)
| | - Thomas Kusmenkov
- Niels-Stensen-Klinken, Marienhospital Osnabrück, Bischofsstr. 1, 49072 Osnabrück, Germany;
| | - Markus Wörnle
- Emergency Department, University Hospital, LMU Munich, Ziemssenstraße 5, 80336 Munich, Germany;
| | - Ludwig Ney
- Department of Anesthesiology, University Hospital, LMU Munich, Ziemssenstraße 5, 80336 Munich, Germany;
| | - Wolfgang Böcker
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Centre Munich (MUM), University Hospital, LMU Munich, Ziemssenstraße 5, 80336 Munich, Germany; (K.F.); (W.B.); (V.B.-F.)
| | - Viktoria Bogner-Flatz
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Centre Munich (MUM), University Hospital, LMU Munich, Ziemssenstraße 5, 80336 Munich, Germany; (K.F.); (W.B.); (V.B.-F.)
| |
Collapse
|
7
|
Wen K, Lin Z, Tan H, Han M. Correlations between coagulation abnormalities and inflammatory markers in trauma-induced coagulopathy. Front Physiol 2024; 15:1474707. [PMID: 39539951 PMCID: PMC11557354 DOI: 10.3389/fphys.2024.1474707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 09/30/2024] [Indexed: 11/16/2024] Open
Abstract
Introduction In multiple trauma patients, the occurrence of trauma-induced coagulopathy (TIC) is closely associated with tissue damage and coagulation function abnormalities in the pathophysiological process. Methods This study established a multiple trauma and shock model in Sprague-Dawley (SD) rats and comprehensively utilized histological staining and radiographic imaging techniques to observe injuries in the intestine, liver, skeletal muscles, and bones. Monitoring activated partial thromboplastin time (APTT), platelet (PLT) count, respiratory rate, blood pressure, and other physiological indicators revealed time-dependent alterations in coagulation function and physiological indicators. Enzyme-linked immunosorbent assay (ELISA) measurements of inflammatory factors Tumor Necrosis Factor-alpha (TNF-α), Interleukin-6 (IL-6) and vascular endothelial injury marker (Syndecan-1) were also conducted. Results Experimental results demonstrated significant changes in tissue structure after multiple traumas, although widespread necrosis or hemorrhagic lesions were not observed. There were time-dependent alterations in coagulation function and physiological indicators. ELISA measurements showed a strong positive correlation between the significant decrease in PLT count and the increase in TNF-α and IL-6 concentrations. Discussion The study provides crucial information for the early diagnosis and treatment of TIC. The findings suggest that structured monitoring of coagulation and inflammatory indicators can help in understanding the pathophysiological changes and aid in the management of TIC in multiple trauma patients.
Collapse
Affiliation(s)
- Ke Wen
- Department of Hand and Microsurgery, Taihe Hospital, Affiliated Hospital of Hubei University of Medicine, Shiyan, Hubei, China
| | - Zhexuan Lin
- Bio-Analytical Laboratory of Shantou University Medical College, Shantou, China
| | - Haizhu Tan
- Department of Preventive Medicine, Shantou University Medical College, Shantou, China
| | - Ming Han
- Emergency Department of Shenzhen University General Hospital, Shenzhen, China
| |
Collapse
|
8
|
Weaver AJ, Venn EC, Ford R, Ewer N, Hildreth KE, Williams CE, Duncan CE, Calhoun CL, Grantham LE, Hoareau GL, Edwards TH. Comparing the effects of various fluid resuscitative strategies on Glycocalyx damage in a canine hemorrhage model. Vet J 2024; 307:106221. [PMID: 39127347 DOI: 10.1016/j.tvjl.2024.106221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 08/05/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
Hemorrhagic shock and subsequent resuscitation can cause significant dysregulation of critical systems, including the vascular endothelium. Following hemorrhage, the endothelial lining (glycocalyx) can shed, causing release of glycocalyx components, endothelial activation, and systemic inflammation. A canine model of hemorrhagic shock was used to evaluate five resuscitation fluids, including Lactated Ringers+Hetastarch, Whole Blood (WB), Fresh Frozen Plasma+packed Red Blood Cells (FFP+pRBC), and two hemoglobin-based oxygen carrier (HBOC) fluids, for their impact on glycocalyx shedding. Under anesthesia, purpose-bred adult canines were instrumented and subjected to a controlled hemorrhage with blood being drawn until a mean arterial pressure of <50 mmHg was reached or 40 % of the estimated blood volume was removed. Canines were left in shock for 45 mins before being resuscitated with one of the resuscitation fluids over 30 mins. Following resuscitation, the dogs were monitored up to 2 weeks. Following an additional 3-4 weeks for washout, the canines repeated the protocol, undergoing each resuscitation fluid individually. Blood samples were collected during each round at various timepoints for serum isolation, which was used for detection of glycocalyx biomarker. Comparison of baseline and post-hemorrhage alone showed a significant reduction in serum protein (p<0.0001), heparan sulfate (p<0.001), and syndecan-1 (p<0.0001) concentrations, and a significant increase in hyaluronan (p<0.0001) concentration. Intercomparisons of resuscitation fluids indicated minimal differences in glycocalyx markers over time. Comparisons within each fluid showed dynamic responses in glycocalyx biomarkers over time. Relative to individual baselines, syndecan-1 was significantly reduced after resuscitation in most cases (p<0.0001), excluding WB and FFP+pRBC. In all cases, VE-cadherin was significantly elevated at 24 hr compared to baseline (p<0.001). Hyaluronan was significantly elevated by 3 hr in all cases (p<0.01), except for HBOC fluids. Total glycosaminoglycans were significantly reduced only at 3 hr (p<0.001) for non-HBOC fluids. Similarly, heparan sulfate was significantly reduced with all fluids between resuscitation and 24 hr (p<0.01), except WB. The temporal changes in canine glycocalyx biomarkers were atypical of hemorrhage response in other species. This suggests that the hemorrhage lacked severity and/or typical glycocalyx biomarkers do not reflect the canine endothelium compared to other species. Further research is needed to characterize the canine endothelium and the response to resuscitation fluids.
Collapse
Affiliation(s)
- Alan J Weaver
- From the US Army Institute of Surgical Research, JBSA Fort Sam Houston, TX, United States.
| | - Emilee C Venn
- From the US Army Institute of Surgical Research, JBSA Fort Sam Houston, TX, United States
| | - Rebekah Ford
- Department of Emergency Medicine, University of Utah Health, Salt Lake City, UT, United States
| | - Nicole Ewer
- Department of Emergency Medicine, University of Utah Health, Salt Lake City, UT, United States
| | - Kim E Hildreth
- From the US Army Institute of Surgical Research, JBSA Fort Sam Houston, TX, United States
| | - Charnae E Williams
- From the US Army Institute of Surgical Research, JBSA Fort Sam Houston, TX, United States
| | - Christina E Duncan
- From the US Army Institute of Surgical Research, JBSA Fort Sam Houston, TX, United States
| | - Cheresa L Calhoun
- From the US Army Institute of Surgical Research, JBSA Fort Sam Houston, TX, United States
| | - Lonnie E Grantham
- From the US Army Institute of Surgical Research, JBSA Fort Sam Houston, TX, United States; Oak Ridge Institute for Science and Education, Oak Ridge, TN, United States
| | - Guillaume L Hoareau
- Department of Emergency Medicine, University of Utah Health, Salt Lake City, UT, United States; Nora Eccles-Harrison Cardiovascular Research Institute, Salt Lake City, UT, United States
| | - Thomas H Edwards
- From the US Army Institute of Surgical Research, JBSA Fort Sam Houston, TX, United States; School of Veterinary Medicine, Texas A&M University, College Station, TX, United States
| |
Collapse
|
9
|
Kemberi M, Minns AF, Santamaria S. Soluble Proteoglycans and Proteoglycan Fragments as Biomarkers of Pathological Extracellular Matrix Remodeling. PROTEOGLYCAN RESEARCH 2024; 2:e70011. [PMID: 39600538 PMCID: PMC11587194 DOI: 10.1002/pgr2.70011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 10/09/2024] [Accepted: 10/23/2024] [Indexed: 11/29/2024]
Abstract
Proteoglycans and their proteolytic fragments diffuse into biological fluids such as plasma, serum, urine, or synovial fluid, where they can be detected by antibodies or mass-spectrometry. Neopeptides generated by the proteolysis of proteoglycans are recognized by specific neoepitope antibodies and can act as a proxy for the activity of certain proteases. Proteoglycan and proteoglycan fragments can be potentially used as prognostic, diagnostic, or theragnostic biomarkers for several diseases characterized by dysregulated extracellular matrix remodeling such as osteoarthritis, rheumatoid arthritis, atherosclerosis, thoracic aortic aneurysms, central nervous system disorders, viral infections, and cancer. Here, we review the main mechanisms accounting for the presence of soluble proteoglycans and their fragments in biological fluids, their potential application as diagnostic, prognostic, or theragnostic biomarkers, and highlight challenges and opportunities ahead of their clinical translation.
Collapse
Affiliation(s)
- Marsioleda Kemberi
- Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonEnglandUK
| | - Alexander F. Minns
- Department of Biochemical SciencesSchool of Biosciences, Faculty of Health and Medical Sciences, University of SurreyGuildfordSurreyUK
| | - Salvatore Santamaria
- Department of Biochemical SciencesSchool of Biosciences, Faculty of Health and Medical Sciences, University of SurreyGuildfordSurreyUK
| |
Collapse
|
10
|
Andersen HG, DellaValle B, Bøgehave H, Mogensen PB, Hahn MK, Goth CK, Sørensen ME, Sigvard AK, Tangmose K, Bojesen KB, Nielsen MØ, Tonetto S, Jørgensen ML, Hempel C, Rungby J, Glenthøj BY, Ambrosen KS, Ebdrup BH. Glycocalyx shedding patterns identifies antipsychotic-naïve patients with first-episode psychosis. Psychiatry Res 2024; 339:116037. [PMID: 38959578 DOI: 10.1016/j.psychres.2024.116037] [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: 12/11/2023] [Accepted: 06/13/2024] [Indexed: 07/05/2024]
Abstract
Psychotic disorders have been linked to immune-system abnormalities, increased inflammatory markers, and subtle neuroinflammation. Studies further suggest a dysfunctional blood brain barrier (BBB). The endothelial Glycocalyx (GLX) functions as a protective layer in the BBB, and GLX shedding leads to BBB dysfunction. This study aimed to investigate whether a panel of 11 GLX molecules derived from peripheral blood could differentiate antipsychotic-naïve first-episode psychosis patients (n47) from healthy controls (HC, n49) and whether GLX shedding correlated with symptom severity. Blood samples were collected at baseline and serum was isolated for GLX marker detection. Machine learning models were applied to test whether patterns in GLX markers could classify patient groups. Associations between GLX markers and symptom severity were explored. Patients showed significantly increased levels of three GLX markers compared to HC. Based on the panel of 11 GLX markers, machine learning models achieved a significant mean classification accuracy of 81%. Post hoc analysis revealed associations between increased GLX markers and symptom severity. This study demonstrates the potential of GLX molecules as immuno-neuropsychiatric biomarkers for early diagnosis of psychosis, as well as indicate a compromised BBB. Further research is warranted to explore the role of GLX in the early detection of psychotic disorders.
Collapse
Affiliation(s)
- Helle G Andersen
- Center for Neuropsychiatric Schizophrenia Research and Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Mental Health Centre Glostrup, Copenhagen University Hospital, Glostrup, Denmark; Copenhagen Research Centre for Mental Health and VIRTU Research Group, Mental Health Centre Copenhagen, Denmark.
| | - Brian DellaValle
- Department of Endocrinology, Copenhagen University Hospital, Bispebjerg and Frederiksberg, Denmark; Copenhagen Center for Translational Research, Copenhagen University Hospital, Bispebjerg and Frederiksberg Hospital, Denmark; GLX Analytix ApS, Copenhagen, Denmark
| | - Hjalte Bøgehave
- Department of Endocrinology, Copenhagen University Hospital, Bispebjerg and Frederiksberg, Denmark; Copenhagen Center for Translational Research, Copenhagen University Hospital, Bispebjerg and Frederiksberg Hospital, Denmark; GLX Analytix ApS, Copenhagen, Denmark
| | - Phillip Bredahl Mogensen
- Copenhagen Center for Translational Research, Copenhagen University Hospital, Bispebjerg and Frederiksberg Hospital, Denmark; GLX Analytix ApS, Copenhagen, Denmark
| | - Margaret K Hahn
- Center for Neuropsychiatric Schizophrenia Research and Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Mental Health Centre Glostrup, Copenhagen University Hospital, Glostrup, Denmark; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Centre for Addiction and Mental Health, Toronto, Canada; Banting and Best Diabetes Centre, University of Toronto, Canada; Department of Pharmacology, University of Toronto, Canada
| | - Christoffer K Goth
- Department of Endocrinology, Copenhagen University Hospital, Bispebjerg and Frederiksberg, Denmark; Copenhagen Center for Translational Research, Copenhagen University Hospital, Bispebjerg and Frederiksberg Hospital, Denmark; GLX Analytix ApS, Copenhagen, Denmark
| | - Mikkel E Sørensen
- Center for Neuropsychiatric Schizophrenia Research and Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Mental Health Centre Glostrup, Copenhagen University Hospital, Glostrup, Denmark
| | - Anne K Sigvard
- Center for Neuropsychiatric Schizophrenia Research and Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Mental Health Centre Glostrup, Copenhagen University Hospital, Glostrup, Denmark
| | - Karen Tangmose
- Center for Neuropsychiatric Schizophrenia Research and Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Mental Health Centre Glostrup, Copenhagen University Hospital, Glostrup, Denmark
| | - Kirsten B Bojesen
- Center for Neuropsychiatric Schizophrenia Research and Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Mental Health Centre Glostrup, Copenhagen University Hospital, Glostrup, Denmark
| | - Mette Ø Nielsen
- Center for Neuropsychiatric Schizophrenia Research and Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Mental Health Centre Glostrup, Copenhagen University Hospital, Glostrup, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Simone Tonetto
- Copenhagen Center for Translational Research, Copenhagen University Hospital, Bispebjerg and Frederiksberg Hospital, Denmark; Laboratory of Neuropsychiatry, Psychiatric Center Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark; Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mathias L Jørgensen
- Department of Endocrinology, Copenhagen University Hospital, Bispebjerg and Frederiksberg, Denmark; Copenhagen Center for Translational Research, Copenhagen University Hospital, Bispebjerg and Frederiksberg Hospital, Denmark; GLX Analytix ApS, Copenhagen, Denmark
| | - Casper Hempel
- GLX Analytix ApS, Copenhagen, Denmark; DTU Health, Department of Health Technology, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Jørgen Rungby
- Department of Endocrinology, Copenhagen University Hospital, Bispebjerg and Frederiksberg, Denmark; Copenhagen Center for Translational Research, Copenhagen University Hospital, Bispebjerg and Frederiksberg Hospital, Denmark
| | - Birte Y Glenthøj
- Center for Neuropsychiatric Schizophrenia Research and Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Mental Health Centre Glostrup, Copenhagen University Hospital, Glostrup, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Karen S Ambrosen
- Center for Neuropsychiatric Schizophrenia Research and Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Mental Health Centre Glostrup, Copenhagen University Hospital, Glostrup, Denmark
| | - Bjørn H Ebdrup
- Center for Neuropsychiatric Schizophrenia Research and Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Mental Health Centre Glostrup, Copenhagen University Hospital, Glostrup, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
11
|
Johansson PI, Vigstedt M, Curry NS, Davenport R, Juffermans NP, Stanworth SJ, Maegele M, Gaarder C, Brohi K, Stensballe J, Henriksen HH. Trauma induced coagulopathy is limited to only one out of four shock induced endotheliopathy (SHINE) phenotypes among moderate-severely injured trauma patients: an exploratory analysis. Scand J Trauma Resusc Emerg Med 2024; 32:71. [PMID: 39160625 PMCID: PMC11331676 DOI: 10.1186/s13049-024-01236-8] [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: 04/22/2024] [Accepted: 07/14/2024] [Indexed: 08/21/2024] Open
Abstract
BACKGROUND Trauma induced coagulopathy remains to be an important cause of high transfusion requirements and mortality and shock induced endotheliopathy (SHINE) has been implicated. METHODS European multicenter observational study of adult trauma patients with injury severity score ≥ 16 arriving within 2 h from injury to the trauma centers. Admission blood samples obtained were used for analysis of the SHINE biomarkers (syndecan-1, soluble thrombomodulin, adrenaline) and extensive analysis of coagulation, -and fibrinolytic factors together with collection of clinical data. Hierarchical clustering of the SHINE biomarkers was used to identify the SHINE phenotypes. RESULTS The 313 patients clustered into four SHINE phenotypes. Phenotype 2, having the highest glycocalyx shedding, encompassing 22% of the whole cohort, had severe coagulopathy with lower levels of prothrombin, FV, IX, X, XI and severe hyperfibrinolysis with higher plasmin - alpha 2-antiplasmin (PAP) - and tPA levels and lower alpha2 - antiplasmin levels. This phenotype had significantly higher transfusion requirements and higher mortality (39% vs. 23%, 15% and 14%) but similar injury severity score (ISS) compared to the others phenotypes. CONCLUSIONS Hierarchical clustering identified four SHINE phenotype in a cohort of trauma patients. Trauma induced coagulopathy was confined to only one of the SHINE phenotypes, encompassing 22% of the total cohort. This phenotype was characterized by severe hypocoagulability and hyperfibrinolysis, which translated to significantly higher transfusion requirements and higher mortality compared to the other SHINE phenotypes with similar injury severity, warranting further investigation.
Collapse
Affiliation(s)
- Pär I Johansson
- CAG Center for Endotheliomics, Copenhagen University Hospital - Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark.
- Department of Clinical Immunology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark.
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark.
| | - Martin Vigstedt
- CAG Center for Endotheliomics, Copenhagen University Hospital - Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
- Department of Clinical Immunology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Nicola S Curry
- Oxford Haemophilia and Thrombosis Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- Radcliffe Department of Medicine, Oxford University, Oxford, UK
| | - Ross Davenport
- Trauma Sciences, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Nicole P Juffermans
- Department of Intensive Care Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Simon J Stanworth
- Radcliffe Department of Medicine, Oxford University, Oxford, UK
- NHS Blood and Transplant, John Radcliffe Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Marc Maegele
- Department of Traumatology and Orthopaedic Surgery, Cologne-Merheim Medical Center, University of Witten/Herdecke, Cologne, Germany
| | | | - Karim Brohi
- Trauma Sciences, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Jakob Stensballe
- CAG Center for Endotheliomics, Copenhagen University Hospital - Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
- Department of Clinical Immunology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Department of Anesthesiology and Trauma Center, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Hanne H Henriksen
- CAG Center for Endotheliomics, Copenhagen University Hospital - Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
- Department of Clinical Immunology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
12
|
Jiang W, Su Y, Su Y, Xu J, Fang Y, Teng J, Ding X, Luo Z, Xu X. Assessing the predictive value of elevated postoperative syndecan-1 levels for progressive acute kidney injury and kidney replacement therapy necessity in adult cardiac surgery patients. BMC Cardiovasc Disord 2024; 24:414. [PMID: 39123133 PMCID: PMC11312249 DOI: 10.1186/s12872-024-04061-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 07/18/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND The development of acute kidney injury (AKI) post-cardiac surgery significantly increases patient morbidity and healthcare costs. Prior researches have established Syndecan-1 (SDC-1) as a potential biomarker for endothelial injury and subsequent acute kidney injury development. This study assessed whether postoperative SDC-1 levels could further predict AKI requiring kidney replacement therapy (AKI-KRT) and AKI progression. METHODS In this prospective study, 122 adult cardiac surgery patients, who underwent valve or coronary artery bypass grafting (CABG) or a combination thereof and developed AKI within 48 h post-operation from May to September 2021, were monitored for the progression to stage 2-3 AKI or the need for KRT. We analyzed the predictive value of postoperative serum SDC-1 levels in relation to multiple endpoints. RESULTS In the study population, 110 patients (90.2%) underwent cardiopulmonary bypass, of which thirty received CABG or combined surgery. Fifteen patients (12.3%) required KRT, and thirty-eight (31.1%) developed progressive AKI, underscoring the severe AKI incidence. Multivariate logistic regression indicated that elevated SDC-1 levels were independent risk factors for progressive AKI (OR = 1.006) and AKI-KRT (OR = 1.011). The AUROC for SDC-1 levels in predicting AKI-KRT and AKI progression was 0.892 and 0.73, respectively, outperforming the inflammatory cytokines. Linear regression revealed a positive correlation between SDC-1 levels and both hospital (β = 0.014, p = 0.022) and ICU stays (β = 0.013, p < 0.001). CONCLUSION Elevated postoperative SDC-1 levels significantly predict AKI progression and AKI-KRT in patients following cardiac surgery. The study's findings support incorporating SDC-1 level monitoring into post-surgical care to improve early detection and intervention for severe AKI.
Collapse
Affiliation(s)
- Wuhua Jiang
- Department of Nephrology, Zhongshan Hospital, Fudan University, No 180 Fenglin Rd, Shanghai, China
| | - Ying Su
- Department of Cardiac Surgery Intensive Care Unit, Zhongshan Hospital, Fudan University, No 180 Fenglin Rd, Shanghai, China
| | - Yiqi Su
- Department of Nephrology, Zhongshan Hospital, Fudan University, No 180 Fenglin Rd, Shanghai, China
- Department of Nephrology, Zhongshan Hospital (Xiamen), Fudan University, Fujian, China
- Xiamen Nephrology Clinical Quality Control Center, Xiamen, China
| | - Jiarui Xu
- Department of Nephrology, Zhongshan Hospital, Fudan University, No 180 Fenglin Rd, Shanghai, China
- Shanghai Institute of Kidney and Dialysis, Shanghai, China
| | - Yi Fang
- Department of Nephrology, Zhongshan Hospital, Fudan University, No 180 Fenglin Rd, Shanghai, China
- Shanghai Institute of Kidney and Dialysis, Shanghai, China
| | - Jie Teng
- Department of Nephrology, Zhongshan Hospital, Fudan University, No 180 Fenglin Rd, Shanghai, China
- Department of Nephrology, Zhongshan Hospital (Xiamen), Fudan University, Fujian, China
- Xiamen Nephrology Clinical Quality Control Center, Xiamen, China
- Shanghai Institute of Kidney and Dialysis, Shanghai, China
| | - Xiaoqiang Ding
- Department of Nephrology, Zhongshan Hospital, Fudan University, No 180 Fenglin Rd, Shanghai, China
- Department of Nephrology, Zhongshan Hospital (Xiamen), Fudan University, Fujian, China
- Xiamen Nephrology Clinical Quality Control Center, Xiamen, China
- Shanghai Institute of Kidney and Dialysis, Shanghai, China
| | - Zhe Luo
- Department of Cardiac Surgery Intensive Care Unit, Zhongshan Hospital, Fudan University, No 180 Fenglin Rd, Shanghai, China.
| | - Xialian Xu
- Department of Nephrology, Zhongshan Hospital, Fudan University, No 180 Fenglin Rd, Shanghai, China.
- Shanghai Institute of Kidney and Dialysis, Shanghai, China.
| |
Collapse
|
13
|
Băetu AE, Mirea L, Cobilinschi C, Grințescu IC, Grințescu IM. Beyond Trauma-Induced Coagulopathy: Detection of Auto-Heparinization as a Marker of Endotheliopathy Using Rotational Thromboelastometry. J Clin Med 2024; 13:4219. [PMID: 39064259 PMCID: PMC11278177 DOI: 10.3390/jcm13144219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 07/13/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Background/Objectives: The complexity of trauma-induced coagulopathy (TIC) is a result of the unique interactions between the patient, trauma, and resuscitation-related causes. The main objective of trauma resuscitation is to create the optimal milieu for both the development of immediate reparatory mechanisms and the prevention of further secondary injuries. Endotheliopathy represents one of the hallmarks of trauma-induced coagulopathy, and comprises endothelial dysfunction, abnormal coagulation, and inflammation, all of which arise after severe trauma and hemorrhagic shock. Methods: We retrospectively and descriptively evaluated 217 patients admitted to the Bucharest Clinical Emergency Hospital who met the Berlin criteria for the diagnosis of multiple trauma. Patients with high suspicion of auto-heparinization were identified according to the dynamic clinical and para-clinical evolution and subsequently tested using rotational thromboelastometry (ROTEM). The ratio between the clot formation time (CT) was used, obtained on the two channels of interest (INTEM/HEPTEM). Results: Among the 217 patients with a mean age of 43.43 ± 15.45 years and a mean injury severity score (ISS) of 36.98 ± 1.875, 42 patients had a reasonable clinical and para-clinical suspicion of auto-heparinization, which was later confirmed by the INTEM/HEPTEM clotting time ratio in 28 cases (12.9% from the entire study population). A multiple linear regression analysis highlighted that serum lactate (estimated 0.02, p = 0.0098) and noradrenaline requirement (estimated 0.03, p = 0.0053) influenced the CT (INTEM/HEPTEM) ratio. Conclusions: There is a subset of multiple trauma patients in which the CT (INTEM/HEPTEM) ratio was influenced only by serum lactate levels and patients' need for vasopressor use, reinforcing the relationship between shock, hypoperfusion, and clotting derangements. This emphasizes the unique response that each patient has to trauma.
Collapse
Affiliation(s)
- Alexandru Emil Băetu
- Department of Anesthesiology and Intensive Care II, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (A.E.B.); (I.M.G.)
- Department of Anesthesiology and Intensive Care, Grigore Alexandrescu Clinical Emergency Hospital for Children, 011743 Bucharest, Romania
| | - Liliana Mirea
- Department of Anesthesiology and Intensive Care II, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (A.E.B.); (I.M.G.)
- Department of Anesthesiology and Intensive Care, Clinical Emergency Hospital Bucharest, 014461 Bucharest, Romania
| | - Cristian Cobilinschi
- Department of Anesthesiology and Intensive Care II, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (A.E.B.); (I.M.G.)
- Department of Anesthesiology and Intensive Care, Clinical Emergency Hospital Bucharest, 014461 Bucharest, Romania
| | | | - Ioana Marina Grințescu
- Department of Anesthesiology and Intensive Care II, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (A.E.B.); (I.M.G.)
- Department of Anesthesiology and Intensive Care, Clinical Emergency Hospital Bucharest, 014461 Bucharest, Romania
| |
Collapse
|
14
|
O'Neil M, Demeulenaere SK, DeChristopher PJ, Holthaus E, Jeske W, Glynn L, Husain A, Muraskas J. Syndecan-1 Level, a Marker of Endothelial Glycocalyx Degradation, Is Associated With Fetal Exposure to Chorioamnionitis and Is a Potential Biomarker for Early-Onset Neonatal Sepsis. Pediatr Dev Pathol 2024; 27:318-326. [PMID: 38616561 DOI: 10.1177/10935266241235504] [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] [Indexed: 04/16/2024]
Abstract
The goal of this investigation was to identify the association between Syndecan-1 (S1) serum levels in preterm newborns exposed to chorioamnionitis (CA) in utero and the potential of S1 as a biomarker of early-onset neonatal sepsis. A cohort of preterm newborns born <33 weeks gestational age was recruited. Within 48 hours of birth, 0.5 mL of blood was drawn to obtain S1 levels, measured via ELISA. Placentas were examined and classified as having (1) no CA, (2) CA without umbilical cord involvement, or (3) CA with inflammation of the umbilical cord (funisitis). S1 levels were compared between preterm newborns without exposure to CA verus newborns with exposure to CA (including with and without funisitis). Preterm newborns exposed to CA were found to have significantly elevated S1 levels compared to those unexposed. Although S1 levels could not differentiate fetal exposure to CA from exposure to CA with funisitis, the combined CA groups had significantly higher S1 levels compared to those not exposed to CA. S1 level has the potential to become a clinically useful biomarker that could assist in the management of mothers and preterm newborns with CA and funisitis. Furthermore, S1 level could aid in the diagnosis and treatment of early-onset neonatal sepsis.
Collapse
Affiliation(s)
- Michaela O'Neil
- Loyola University Chicago, Maywood, IL, USA
- The University of Chicago, Chicago, IL, USA
| | | | | | - Emily Holthaus
- Loyola University Chicago, Maywood, IL, USA
- UT Southwestern Medical Center, Chicago, IL, USA
| | | | | | | | | |
Collapse
|
15
|
Hayashi K, Koyama D, Hamazaki Y, Kamiyama T, Yamada S, Furukawa M, Tanino Y, Shibata Y, Ikezoe T. Syndecan-1 as a prognostic biomarker in COVID-19 patients: a retrospective study of a Japanese cohort. Thromb J 2024; 22:52. [PMID: 38907229 PMCID: PMC11191303 DOI: 10.1186/s12959-024-00619-2] [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: 03/25/2024] [Accepted: 06/09/2024] [Indexed: 06/23/2024] Open
Abstract
BACKGROUND The coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has had a profound global impact, with millions of confirmed cases and deaths worldwide. While most cases are mild, a subset progresses to severe respiratory complications and death, with factors such as thromboembolism, age, and underlying health conditions increasing the risk. Vascular endothelial damage has been implicated in severe outcomes, but specific biomarkers remain elusive. This study investigated syndecan-1 (SDC-1), a marker of endothelial damage, as a potential prognostic factor for COVID-19, focusing on the Japanese population, which is known for its aging demographics and high prevalence of comorbidities. METHODS A multicenter retrospective study of COVID-19 patients in Fukushima Prefecture in Japan who were admitted between February 2020 and August 2021 was conducted. SDC-1 levels were measured along with other clinical and laboratory parameters. Outcomes including thrombosis, 28-day survival, and disease severity were assessed, and disease severity was categorized according to established guidelines. RESULTS SDC-1 levels were correlated with disease severity. Patients who died from COVID-19 had greater SDC-1 levels than survivors, and the area under the receiver operating characteristic curve (AUC) analysis suggested the potential of the SDC-1 level as a predictor of mortality (AUC 0.714). K‒M analysis also revealed a significant difference in survival based on an SDC-1 cutoff of 10.65 ng/mL. DISCUSSION This study suggested that SDC-1 may serve as a valuable biomarker for assessing COVID-19 severity and predicting mortality within 28 days of hospitalization, particularly in the Japanese population. However, further investigations are required to assess longitudinal changes in SDC-1 levels, validate its predictive value for long-term survival, and consider its applicability to new viral variants. CONCLUSIONS SDC-1 is emerging as a potential biomarker for assessing the severity and life expectancy of COVID-19 in the Japanese population, offering promise for improved risk stratification and patient management in the ongoing fight against the virus.
Collapse
Affiliation(s)
- Kiyohito Hayashi
- Department of Hematology, Fukushima Medical University, Fukushima, Fukushima, 960-1295, Japan
| | - Daisuke Koyama
- Department of Hematology, Fukushima Medical University, Fukushima, Fukushima, 960-1295, Japan
| | - Yoichi Hamazaki
- Department of Hematology, Iwaki City Medical Center, Iwaki, Fukushima, Japan
| | - Takamichi Kamiyama
- Department of Pediatric Surgery, Iwaki City Medical Center, Iwaki, Fukushima, Japan
| | - Shingo Yamada
- R&D Center, Shino-Test Corporation, Sagamihara, Kanagawa, Japan
| | - Miki Furukawa
- Division of Hematology, Kita-Fukushima Medical Center, Date, Fukushima, Japan
| | - Yoshinori Tanino
- Department of Pulmonary Medicine, Fukushima Medical University, Fukushima, Fukushima, Japan
| | - Yoko Shibata
- Department of Pulmonary Medicine, Fukushima Medical University, Fukushima, Fukushima, Japan
| | - Takayuki Ikezoe
- Department of Hematology, Fukushima Medical University, Fukushima, Fukushima, 960-1295, Japan.
| |
Collapse
|
16
|
Song Z, Wang Z, Cai J, Zhou Y, Jiang Y, Tan J, Gu L. Down-regulating lncRNA KCNQ1OT1 relieves type II alveolar epithelial cell apoptosis during one-lung ventilation via modulating miR-129-5p/HMGB1 axis induced pulmonary endothelial glycocalyx. ENVIRONMENTAL TOXICOLOGY 2024; 39:3578-3596. [PMID: 38488667 DOI: 10.1002/tox.24201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 02/02/2024] [Accepted: 02/25/2024] [Indexed: 05/16/2024]
Abstract
OBJECTIVE Endothelial glycocalyx (EG) maintains vascular homeostasis and is destroyed after one-lung ventilation (OLV)-induced lung injury. Long noncoding RNAs (lncRNAs) are critically involved in various lung injuries. This study aimed to investigate the role and regulatory mechanism of KCNQ1 overlapping transcript 1 (KCNQ1OT1) in OLV-induced lung injury and LPS-induced type II alveolar epithelial cell (AECII) apoptosis. METHODS The rat OLV model was established, and the effects of KCNQ1OT1 on OLV-induced ALI in vivo were explored. Bax and Caspase-3 expression in rat lung tissues was measured by immunochemistry (IHC). AECIIs were isolated from rat lungs and treated with LPS or normal saline (control) for in vitro analysis. The expression of KCNQ1OT1, miR-129-5p, and HMGB1 was measured by quantitative real-time PCR (qRT-PCR) or Western blot (WB). Cell proliferation and apoptosis were examined by 3-(4,5)-dimethylthiahiazo (-z-y1)-3,5-di- phenytetrazoliumromide (MTT) and flow cytometry. The downstream targets of KCNQ1OT1 were predicted by bioinformatics, and the binding relationship between KCNQ1OT1 and miR-129-3p was verified by dual-luciferase reporter assays. The potential target of miR-129-5p was further explored on the Targetscan website and revealed to target HMGB1. Enzyme-linked immunosorbent assay (ELISA) or WB was adopted to determine the levels of IL-1β, TNF-α, MDA, SOD, heparanase (HPA), matrix metalloproteinase 9 (MMP9), heparan sulfate (HS) and syndecan-1 (SDC-1). RESULTS KCNQ1OT1 and HMGB1 were up-regulated during OLV-induced lung injury, and their expression was positively correlated. KCNQ1OT1 knockdown reduced OLV-induced pulmonary edema and lung epithelial cell apoptosis, increased vascular permeability, reduced IL-1β, TNF-α, MDA, and SOD levels and glycocalyx markers by targeting miR-129-5p or upregulating HMGB1. Overexpressing KCNQ1OT1 promoted cell apoptosis, reduced cell proliferation, aggravated inflammation and oxidative stress, and up-regulated HMGB1, HPA and MMP9 in LPS-treated AECIIs, while the HMGB1 silencing showed the opposite effects. MiR-129-5p mimics partially eliminated the KCNQ1OT1-induced effects, while recombinant HMGB1 restored the effects of miR-129-5p overexpression on AECIIs. Additionally, KCNQ1OT1 was demonstrated to promote the activation of the p38 MAPK/Akt/ERK signaling pathways in AECIIs via HMGB1. CONCLUSION KCNQ1OT1 knockdown alleviated AECII apoptosis and EG damage during OLV by targeting miR-129-5p/HMGB1 to inactivate the p38 MAPK/Akt/ERK signaling. The findings of our study might deepen our understanding of the molecular basis in OLV-induced lung injury and provide clues for the targeted disease management.
Collapse
Affiliation(s)
- Zhenghuan Song
- Department of Anesthesiology, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing City, China
| | - Zhongqiu Wang
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing City, China
| | - Jiaqin Cai
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou City, Jiangsu Province, China
| | - Yihu Zhou
- Department of Anesthesiology, Nanjing Medical University, Nanjing City, Jiangsu Province, China
| | - Yueyi Jiang
- Department of Anesthesiology, Nanjing Medical University, Nanjing City, Jiangsu Province, China
| | - Jing Tan
- Department of Anesthesiology, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing City, China
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou City, Jiangsu Province, China
| | - Lianbin Gu
- Department of Anesthesiology, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing City, China
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou City, Jiangsu Province, China
| |
Collapse
|
17
|
Rabia B, Thanigaimani S, Golledge J. The potential involvement of glycocalyx disruption in abdominal aortic aneurysm pathogenesis. Cardiovasc Pathol 2024; 70:107629. [PMID: 38461960 DOI: 10.1016/j.carpath.2024.107629] [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: 01/07/2024] [Revised: 03/03/2024] [Accepted: 03/04/2024] [Indexed: 03/12/2024] Open
Abstract
BACKGROUND Abdominal aortic aneurysm is a weakening and expansion of the abdominal aorta. Currently, there is no drug treatment to limit abdominal aortic aneurysm growth. The glycocalyx is the outermost layer of the cell surface, mainly composed of glycosaminoglycans and proteoglycans. OBJECTIVE The aim of this review was to identify a potential relationship between glycocalyx disruption and abdominal aortic aneurysm pathogenesis. METHODS A narrative review of relevant published research was conducted. RESULTS Glycocalyx disruption has been reported to enhance vascular permeability, impair immune responses, dysregulate endothelial function, promote extracellular matrix remodeling and modulate mechanotransduction. All these effects are implicated in abdominal aortic aneurysm pathogenesis. Glycocalyx disruption promotes inflammation through exposure of adhesion molecules and release of proinflammatory mediators. Glycocalyx disruption affects how the endothelium responds to shear stress by reducing nitric oxide availabilty and adversely affecting the storage and release of several antioxidants, growth factors, and antithromotic proteins. These changes exacerbate oxidative stress, stimulate vascular smooth muscle cell dysfunction, and promote thrombosis, all effects implicated in abdominal aortic aneurysm pathogenesis. Deficiency of key component of the glycocalyx, such as syndecan-4, were reported to promote aneurysm formation and rupture in the angiotensin-II and calcium chloride induced mouse models of abdominal aortic aneurysm. CONCLUSION This review provides a summary of past research which suggests that glycocalyx disruption may play a role in abdominal aortic aneurysm pathogenesis. Further research is needed to establish a causal link between glycocalyx disruption and abdominal aortic aneurysm development.
Collapse
Affiliation(s)
- Bibi Rabia
- Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, Queensland 4811, Australia; Department of Pharmacy, Hazara University, Mansehra 21300, Pakistan
| | - Shivshankar Thanigaimani
- Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, Queensland 4811, Australia; The Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, Queensland 4811, Australia
| | - Jonathan Golledge
- Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, Queensland 4811, Australia; The Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, Queensland 4811, Australia; The Department of Vascular and Endovascular Surgery, The Townsville University Hospital, Townsville, Queensland 4810, Australia.
| |
Collapse
|
18
|
Abel B, Mares J, Hutzler J, Parajuli B, Kurada L, White JM, Propper BW, Stewart IJ, Burmeister DM. The degree of aortic occlusion in the setting of trauma alters the extent of acute kidney injury associated with mitochondrial preservation. Am J Physiol Renal Physiol 2024; 326:F669-F679. [PMID: 38450433 DOI: 10.1152/ajprenal.00323.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 02/09/2024] [Accepted: 02/24/2024] [Indexed: 03/08/2024] Open
Abstract
Resuscitative endovascular balloon occlusion of the aorta (REBOA) is used to control noncompressible hemorrhage not addressed with traditional tourniquets. However, REBOA is associated with acute kidney injury (AKI) and subsequent mortality in severely injured trauma patients. Here, we investigated how the degree of aortic occlusion altered the extent of AKI in a porcine model. Female Yorkshire-cross swine (n = 16, 68.1 ± 0.7 kg) were anesthetized and had carotid and bilateral femoral arteries accessed for REBOA insertion and distal and proximal blood pressure monitoring. Through a laparotomy, a 6-cm liver laceration was performed and balloon inflation was performed in zone 1 of the aorta for 90 min, during which animals were randomized to target distal mean arterial pressures of 25 or 45 mmHg via balloon volume adjustment. Blood draws were taken at baseline, end of occlusion, and time of death, at which point renal tissues were harvested 6 h after balloon deflation for histological and molecular analyses. Renal blood flow was lower in the 25-mmHg group (48.5 ± 18.3 mL/min) than in the 45-mmHg group (177.9 ± 27.2 mL/min) during the occlusion phase, which recovered and was not different after balloon deflation. AKI was more severe in the 25-mmHg group, as evidenced by circulating creatinine, blood urea nitrogen, and urinary neutrophil gelatinase-associated lipocalin. The 25-mmHg group had increased tubular necrosis, lower renal citrate synthase activity, increased tissue and circulating syndecan-1, and elevated systemic inflammatory cytokines. The extent of renal ischemia-induced AKI is associated with the magnitude of mitochondrial biomass and systemic inflammation, highlighting potential mechanistic targets to combine with partial REBOA strategies to prevent AKI.NEW & NOTEWORTHY Large animal models of ischemia-reperfusion acute kidney injury (IR-AKI) are lacking. This report establishes a titratable IR-AKI model in swine in which a balloon catheter can be used to alter distal pressures experienced by the kidney, thus controlling renal blood flow. Lower blood flow results in greater renal dysfunction and structural damage, as well as lower mitochondrial biomass, elevated systemic inflammation, and vascular dysfunction.
Collapse
Affiliation(s)
- Biebele Abel
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Incorporated, Bethesda, Maryland, United States
- Department of Surgery, Uniformed Services University of the Health Science, Bethesda, Maryland, United States
| | - John Mares
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Incorporated, Bethesda, Maryland, United States
- Department of Surgery, Uniformed Services University of the Health Science, Bethesda, Maryland, United States
| | - Justin Hutzler
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Incorporated, Bethesda, Maryland, United States
- Department of Surgery, Uniformed Services University of the Health Science, Bethesda, Maryland, United States
| | - Babita Parajuli
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States
| | - Lalitha Kurada
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Incorporated, Bethesda, Maryland, United States
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States
| | - Joseph M White
- Division of Vascular Surgery and Endovascular Therapy, Johns Hopkins School of Medicine, Baltimore, Maryland, United States
| | - Brandon W Propper
- Department of Surgery, Uniformed Services University of the Health Science, Bethesda, Maryland, United States
- Department of Surgery, Walter Reed National Military Medical Center, Bethesda, Maryland, United States
| | - Ian J Stewart
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States
| | - David M Burmeister
- Department of Surgery, Uniformed Services University of the Health Science, Bethesda, Maryland, United States
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States
| |
Collapse
|
19
|
Baucom MR, Weissman N, Price AD, England L, Schuster RM, Pritts TA, Goodman MD. Syndecan-1 as the Effect or Effector of the Endothelial Inflammatory Response? J Surg Res 2024; 295:611-618. [PMID: 38096775 DOI: 10.1016/j.jss.2023.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 09/11/2023] [Accepted: 10/27/2023] [Indexed: 02/25/2024]
Abstract
INTRODUCTION Syndecan-1 is a heparan sulfate proteoglycan found in the glycocalyx of vascular endothelial cells. Serum levels of syndecan-1 have repeatedly been demonstrated to increase following traumatic injury and shock, but it is unclear whether syndecan-1 plays an active role in the inflammatory response or is simply a biomarker of a state of hypoperfusion. The aim of this study was to identify the role of syndecan-1 role in the inflammatory process in the absence of trauma. METHODS Male mice were randomized into five groups (n = 3). Four groups received increasing concentrations of syndecan-1 (1, 10, 100, and 1000pg/mL per blood volume) and a fifth group was given normal saline as a control via intravenous injection. These concentrations were selected based on previous syndecan-1 enzyme-linked immunosorbent assay data acquired following induced hemorrhagic shock in mice resulting in serum levels of 10-6000 pg/mL. Mice from each group were sacrificed at 1-, 4-, and 24-h time points for serum biomarker evaluation. A multiplex enzyme-linked immunosorbent assay was performed to analyze proinflammatory cytokines and chemokines including interleukin (IL)-1a, IL-1b, IL-2, IL-3, IL-4, IL-6, IL-10, IL-12, IL-17, monocyte chemoattractant protein-1, TNF-α, macrophage inflammatory protein-1α, granulocyte-macrophage colony-stimulating factor, and normal T cell expressed and presumably secreted levels. Whole blood was analyzed via rotational thromboelastometry in a separate group of mice dosed with syndecan-1 at 1000 pg/mL and compared to sham mice at 1 h. RESULTS Tumor necrosis factor-α was significantly elevated in the 1000 pg/mL group compared to sham animals. There were no significant changes in IL-1a, IL-1b, IL-2, IL-3, IL-4, IL-6, IL-10, IL-12, monocyte chemoattractant protein--1, macrophage inflammatory protein-1α, granulocyte-macrophage colony-stimulating factor, or normal T cell expressed and presumably secretedat 1, 4, and 24 h for any group when compared to mice receiving saline alone. No significant differences were noted in coagulability between the 1000 pg/mL syndecan-1 group and shams at 1 h CONCLUSIONS: Inflammatory cytokine concentrations did not change with increasing dosage of syndecan-1 within mice at any timepoint, except for an acute change in tumor necrosis factor-α which was transient. Based on our results, syndecan-1 appears to be a biomarker for inflammation rather than an active participant in eliciting an inflammatory response. Further research will focus on the role of syndecan-1 following hemorrhagic shock.
Collapse
Affiliation(s)
- Matthew R Baucom
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio
| | | | - Adam D Price
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio
| | - Lisa England
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio
| | | | - Timothy A Pritts
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio
| | | |
Collapse
|
20
|
Maier CL, Brohi K, Curry N, Juffermans NP, Mora Miquel L, Neal MD, Shaz BH, Vlaar APJ, Helms J. Contemporary management of major haemorrhage in critical care. Intensive Care Med 2024; 50:319-331. [PMID: 38189930 DOI: 10.1007/s00134-023-07303-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 12/05/2023] [Indexed: 01/09/2024]
Abstract
Haemorrhagic shock is frequent in critical care settings and responsible for a high mortality rate due to multiple organ dysfunction and coagulopathy. The management of critically ill patients with bleeding and shock is complex, and treatment of these patients must be rapid and definitive. The administration of large volumes of blood components leads to major physiological alterations which must be mitigated during and after bleeding. Early recognition of bleeding and coagulopathy, understanding the underlying pathophysiology related to specific disease states, and the development of individualised management protocols are important for optimal outcomes. This review describes the contemporary understanding of the pathophysiology of various types of coagulopathic bleeding; the diagnosis and management of critically ill bleeding patients, including major haemorrhage protocols and post-transfusion management; and finally highlights recent areas of opportunity to better understand optimal management strategies for managing bleeding in the intensive care unit (ICU).
Collapse
Affiliation(s)
- Cheryl L Maier
- Department of Pathology and Laboratory Medicine, Center for Transfusion and Cellular Therapies, Emory University School of Medicine, Atlanta, GA, USA
| | - Karim Brohi
- Centre for Trauma Sciences, Queen Mary University of London, London, UK
| | - Nicola Curry
- Oxford Haemophilia and Thrombosis Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- Nuffield Department of Clinical and Laboratory Sciences, Radcliffe Department of Medicine, Oxford University, Oxford, UK
| | - Nicole P Juffermans
- Department of Intensive Care and Laboratory of Translational Intensive Care, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Lidia Mora Miquel
- Department of Anaesthesiology, Intensive Care and Pain Clinic, Vall d'Hebron Trauma, Rehabilitation and Burns Hospital, Autonomous University of Barcelona, Passeig de La Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Matthew D Neal
- Trauma and Transfusion Medicine Research Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Beth H Shaz
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| | | | - Julie Helms
- Service de Médecine Intensive-Réanimation, Department of Intensive Care, Nouvel Hôpital Civil, Université de Strasbourg (UNISTRA), 1, Place de L'Hôpital, 67091, Strasbourg Cedex, France.
| |
Collapse
|
21
|
Diebel LN, Liberati DM, Karadjoff A, Terasaki Y, Srour A, McPherson S. Detection of glycocalyx degradation in real time: A conceptual model of thromboelastography. Surgery 2024; 175:613-617. [PMID: 37863690 DOI: 10.1016/j.surg.2023.08.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 08/15/2023] [Accepted: 08/17/2023] [Indexed: 10/22/2023]
Abstract
BACKGROUND The endothelial glycocalyx is a critical component of the vascular barrier; its disruption after shock states may contribute to coagulopathy in a variety of conditions. Measurement of glycocalyx components in plasma have been used to index glycocalyx degradation but are not available as a point of care test. Heparanoids, such as heparan sulfate, may affect coagulation which may be detected by either thromboelastography or activated clotting time. METHODS Endothelial glycocalyx components syndecan-1 and heparan sulfate were added to blood samples at clinically relevant concentrations. Thromboelastography values included clot reaction time, clot amplification and fibrinogen values, and maximum clot strength (maximum amplitude, platelets). The heparinase thromboelastography cartridge was used to detect a heparin-like effect. The activated clotting time test was performed subsequently using the heparan sulfate blood samples to compare a standard coagulation test with thromboelastography clot reaction times. RESULTS Both thromboelastography clot reaction time (with comparison to heparinase) and activated clotting time were useful to detect effects of coagulation. Thromboelastography also detected platelet and fibrinogen abnormalities at higher heparan sulfate concentrations. Studies using thromboelastography or even activated clotting time may be useful to detect glycocalyx degradation after shock states and may guide clinical decision making. CONCLUSION Thromboelastography and or activated clotting time may be useful to detect glycocalyx degradation as a point of care test in patients in the acute setting. Additionally, these assays may detect previous undisclosed coagulopathy due to glycocalyx degradation.
Collapse
Affiliation(s)
- Lawrence N Diebel
- Michael and Marian Ilitch Department of Surgery, Wayne State University, Detroit, MI.
| | - David M Liberati
- Michael and Marian Ilitch Department of Surgery, Wayne State University, Detroit, MI
| | - Alison Karadjoff
- Michael and Marian Ilitch Department of Surgery, Wayne State University, Detroit, MI
| | - Yusuke Terasaki
- Michael and Marian Ilitch Department of Surgery, Wayne State University, Detroit, MI
| | - Ali Srour
- Michael and Marian Ilitch Department of Surgery, Wayne State University, Detroit, MI
| | - Steve McPherson
- Michael and Marian Ilitch Department of Surgery, Wayne State University, Detroit, MI
| |
Collapse
|
22
|
Ho JW, Dawood ZS, Taylor ME, Liggett MR, Jin G, Jaishankar D, Nadig SN, Bharat A, Alam HB. THE NEUROENDOTHELIAL AXIS IN TRAUMATIC BRAIN INJURY: MECHANISMS OF MULTIORGAN DYSFUNCTION, NOVEL THERAPIES, AND FUTURE DIRECTIONS. Shock 2024; 61:346-359. [PMID: 38517237 DOI: 10.1097/shk.0000000000002307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2024]
Abstract
ABSTRACT Severe traumatic brain injury (TBI) often initiates a systemic inflammatory response syndrome, which can potentially culminate into multiorgan dysfunction. A central player in this cascade is endotheliopathy, caused by perturbations in homeostatic mechanisms governed by endothelial cells due to injury-induced coagulopathy, heightened sympathoadrenal response, complement activation, and proinflammatory cytokine release. Unique to TBI is the potential disruption of the blood-brain barrier, which may expose neuronal antigens to the peripheral immune system and permit neuroinflammatory mediators to enter systemic circulation, propagating endotheliopathy systemically. This review aims to provide comprehensive insights into the "neuroendothelial axis" underlying endothelial dysfunction after TBI, identify potential diagnostic and prognostic biomarkers, and explore therapeutic strategies targeting these interactions, with the ultimate goal of improving patient outcomes after severe TBI.
Collapse
Affiliation(s)
- Jessie W Ho
- Department of Surgery, Division of Trauma Surgery and Critical Care, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Zaiba Shafik Dawood
- Department of Surgery, Division of Trauma Surgery and Critical Care, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Meredith E Taylor
- Department of Surgery, Division of Organ Transplant, and Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University Chicago, Illinois
| | - Marjorie R Liggett
- Department of Surgery, Division of Trauma Surgery and Critical Care, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Guang Jin
- Department of Surgery, Division of Trauma Surgery and Critical Care, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Dinesh Jaishankar
- Department of Surgery, Division of Organ Transplant, and Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University Chicago, Illinois
| | - Satish N Nadig
- Department of Surgery, Division of Organ Transplant, and Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University Chicago, Illinois
| | - Ankit Bharat
- Department of Surgery, Division of Thoracic Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Hasan B Alam
- Department of Surgery, Division of Trauma Surgery and Critical Care, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| |
Collapse
|
23
|
Rajan R, Hanifah M, Mariappan V, Anand M, Balakrishna Pillai A. Soluble Endoglin and Syndecan-1 levels predicts the clinical outcome in COVID-19 patients. Microb Pathog 2024; 188:106558. [PMID: 38272329 DOI: 10.1016/j.micpath.2024.106558] [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: 11/02/2023] [Revised: 01/21/2024] [Accepted: 01/22/2024] [Indexed: 01/27/2024]
Abstract
Endothelial instability is reported to be involved in the pathogenesis of COVID-19. The mechanism that regulates the endothelial dysfunction and disease virulence is not known. Studies on proteins that are released into circulation by activated endothelial cells may provide some means to understand the disease manifestation. The study investigated the circulating levels of two molecules Endoglin (Eng) and Syndecan-1 (SDC-1) that are presumed to be involved in the maintenance of endothelial integrity and their association with hypercoagulation marker in COVID-19 patients. The serum levels of Eng, SDC-1, D-mer were evaluated using ELISA at the time of admission (DOA) and day 7 post-admission among COVID-19 patients (N = 39 with 17 moderate and 22 severe cases). Compared to the time of admission, there was an increase in sEng and sSDC1 levels in all COVID-19 cases on day 7 post admission. The serum levels of sEng and sSDC-1 was significantly (P ≤ 0.001 & P ≤ 0.01 respectively) elevated in severe cases including the four deceased group compared to moderate cases on day 7 post admission. Further, the study molecules showed a strong positive association (P ≤ 0.001) with the hypercoagulation marker D-mer. The results show an early shedding of the endothelial proteins sEng and sSDC-1 into circulation as a host response to the viral infection during the febrile phase of infection. Increased levels of sEng and sSDC-1 along with D-mer could be beneficial in predicting COVID-19 disease severity.
Collapse
Affiliation(s)
- Remya Rajan
- Department of General Medicine, Mahatma Gandhi Medical College and Research Institute (MGMCRI), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry, 607 402, India.
| | - Mohamed Hanifah
- Department of General Medicine, Mahatma Gandhi Medical College and Research Institute (MGMCRI), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry, 607 402, India.
| | - Vignesh Mariappan
- Mahatma Gandhi Medical Advanced Research Institute (MGMARI), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry, 607 402, India.
| | - Monica Anand
- Department of General Medicine, Mahatma Gandhi Medical College and Research Institute (MGMCRI), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry, 607 402, India.
| | - Agieshkumar Balakrishna Pillai
- Mahatma Gandhi Medical Advanced Research Institute (MGMARI), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry, 607 402, India.
| |
Collapse
|
24
|
Johansson PI, Fenger Eriksen C, Bovbjerg PE, Gaarder C, Pall M, Henriksen HH, Pedersen KH, Vigstedt M, Lange T, Næss PA, Strømgaard Andersen M, Kirkegaard H, Stensballe J. Prostacyclin in trauma patients with hemorrhagic shock: A randomized clinical trial. J Trauma Acute Care Surg 2024; 96:476-481. [PMID: 37962189 DOI: 10.1097/ta.0000000000004150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
BACKGROUND A main cause of trauma morbidity and mortality is multiple-organ failure, and endotheliopathy has been implicated. Pilot studies indicate that low-dose prostacyclin improves endothelial functionality in critically ill patients, suggesting that this intervention may improve trauma patient outcome. METHODS We conducted a multicenter, randomized, blinded, clinical investigator-initiated trial in 229 trauma patients with hemorrhagic shock who were randomized 1:1 to 72 hours infusion of the prostacyclin analog iloprost (1 ng/kg/min) or placebo. The primary outcome was the number of intensive care unit (ICU)-free days alive within 28 days of admission. Secondary outcomes included 28-day all-cause mortality and hospital length of stay. RESULTS The mean number of ICU-free days alive within 28 days was 15.64 days in the iloprost group versus 13.99 days in the placebo group (adjusted mean difference, -1.63 days [95% confidence interval (CI), -4.64 to 1.38 days]; p = 0.28). The 28-day mortality was 18.8% in the iloprost group versus 19.6% in the placebo group (odds ratio, 1.01 [95% CI, 0.51-2.0]; p = 0.97). The mean hospital length of stay was 19.96 days in the iloprost group versus 27.32 days in the placebo group (adjusted mean difference, 7.84 days [95% CI, 1.66-14.02 days], p = 0.01). CONCLUSION Iloprost did not result in a statistically significant increase in the number of ICU-free days alive within 28 days of admission, whereas it was safe and a statistically significant reduction in hospital length of stay was observed. Further research on prostacyclin in shocked trauma patients is warranted. LEVEL OF EVIDENCE Therapeutic/Care Management; Level II.
Collapse
Affiliation(s)
- Pär I Johansson
- From the CAG Center for Endotheliomics (P.I.J.), Copenhagen University Hospital-Rigshospitalet; Department of Clinical Medicine (P.I.J.), University of Copenhagen, Copenhagen; Department of Anesthesiology (C.F.E., M.S.A.), Aarhus University Hospital, Aarhus; Department of Orthopedic Surgery (P.E.B.), Odense University Hospital, Odense, Denmark; Department of Traumatology (C.G., P.A.N.), Oslo University Hospital; Institute of Clinical Medicine (C.G., P.A.N.), University of Oslo, Oslo, Norway; Department of Anesthesiology (M.P.) and Intensive Care V (M.P.), Odense University Hospital, Odense; CAG Center for Endotheliomics (H.H.H., K.H.P., M.V.), Copenhagen University Hospital-Rigshospitalet; Section of Biostatistics (T.L.), University of Copenhagen, Copenhagen; Research Center for Emergency Medicine (H.K.), Aarhus University Hospital; Aarhus University (H.K.), Aarhus; and CAG Center for Endotheliomics (J.S.), and Department of Anesthesiology (J.S.), Department of Anesthesiology, Centre of Head and Orthopedics (J.S.), Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Wang J, Wu Y. Mass intraoperative endothelial glycocalyx shedding affects postoperative systemic inflammation response. BMC Anesthesiol 2024; 24:76. [PMID: 38408904 PMCID: PMC10895804 DOI: 10.1186/s12871-024-02459-z] [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: 07/31/2023] [Accepted: 02/17/2024] [Indexed: 02/28/2024] Open
Abstract
BACGROUND Off-pump coronary artery bypass graft (OPCABG) has a high incidence of postoperative systemic inflammation response syndrome (SIRS), and perioperative endothelial glycocalyx layer (EGL) disruption can be one of the predisposing factors. We hypothesized that EGL shedding happened earlier in OPCABG which can influence on postoperative SIRS, and sevoflurane might preserve EGL better than propofol. METHODS We randomly allocated 50 patients undergoing OPCABG to receive either sevoflurane-sufentanil or propofol-sufentanil anesthesia. Plasma syndecan-1, heparan sulfate (HS), atrial natriuretic peptide (ANP), IL-6, and cardiac troponin I (cTnI) were measured. Blood samples were collected at 6 timepoints: induction (T1), before grafting (T2), after grafting(T3), surgery done (T4), postoperative day1 (POD1,T5) and POD2 (T6). SIRS criteria and sequential organ failure assessment (SOFA) score were examined. RESULTS There were neither differences of syndecan-1, HS, IL-6 nor of SIRS criteria or SOFA score between the sevoflurane and propofol groups. All patients were pooled as a single group for further statistical analyses, plasma syndecan-1 (P < 0.001) and IL-6 (P < 0.001) increased significantly as a function of time; syndecan-1 increasing correlated significantly with the duration of coronary graft anastomosis (r = 0.329, P = 0.026). Syndecan-1(T3) correlated significantly with ANP(T3) (r = 0.0.354, P = 0.016) and IL-6 (T5) (r = 0.570, P < 0.001). The maximum value of IL-6 correlated significantly with SIRS (r = 0.378, P = 0.010), the maximum value of SOFA score (r = 0.399, P = 0.006) and ICU days (r = 0.306, P = 0.039). The maximum value of SOFA score correlated significantly with the occurrence of SIRS (r = 0.568, P < 0.001) and ICU days (r = 0.338, P = 0.022). CONCLUSIONS OPCABG intraoperative early EGL shedding caused of grafts anastomosis greatly affected postoperative SIRS and SOFA score, sevoflurane did not clinically preserve EGL better. TRIAL REGISTRATION ChiCTR-IOR-17012535. Registered on 01/09/2017.
Collapse
Affiliation(s)
- JiaWan Wang
- Department of Anesthesiology, Beijing Chaoyang Hospital Affiliated to Capital Medical University, Beijing, China
| | - Yan Wu
- Department of Anesthesiology, Beijing Chaoyang Hospital Affiliated to Capital Medical University, Beijing, China.
| |
Collapse
|
26
|
Cralley AL, Moore EE, LaCroix I, Schaid TJ, Thielen O, Hallas W, Hom P, Mitra S, Kelher M, Hansen K, Cohen M, Silliman C, Sauaia A, Fox CJ. RESUSCITATIVE ENDOVASCULAR BALLOON OCCLUSION OF THE AORTA: ZONE 1 REPERFUSION-INDUCED COAGULOPATHY. Shock 2024; 61:322-329. [PMID: 38407818 PMCID: PMC10955717 DOI: 10.1097/shk.0000000000002299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
ABSTRACT Objective: We sought to identify potential drivers behind resuscitative endovascular balloon occlusion of the aorta (REBOA) induced reperfusion coagulopathy using novel proteomic methods. Background: Coagulopathy associated with REBOA is poorly defined. The REBOA Zone 1 provokes hepatic and intestinal ischemia that may alter coagulation factor production and lead to molecular pathway alterations that compromises hemostasis. We hypothesized that REBOA Zone 1 would lead to reperfusion coagulopathy driven by mediators of fibrinolysis, loss of coagulation factors, and potential endothelial dysfunction. Methods: Yorkshire swine were subjected to a polytrauma injury (blast traumatic brain injury, tissue injury, and hemorrhagic shock). Pigs were randomized to observation only (controls, n = 6) or to 30 min of REBOA Zone 1 (n = 6) or REBOA Zone 3 (n = 4) as part of their resuscitation. Thromboelastography was used to detect coagulopathy. ELISA assays and mass spectrometry proteomics were used to measure plasma protein levels related to coagulation and systemic inflammation. Results: After the polytrauma phase, balloon deflation of REBOA Zone 1 was associated with significant hyperfibrinolysis (TEG results: REBOA Zone 1 35.50% versus control 9.5% vs. Zone 3 2.4%, P < 0.05). In the proteomics and ELISA results, REBOA Zone 1 was associated with significant decreases in coagulation factor XI and coagulation factor II, and significant elevations of active tissue plasminogen activator, plasmin-antiplasmin complex complexes, and syndecan-1 (P < 0.05). Conclusion: REBOA Zone 1 alters circulating mediators of clot formation, clot lysis, and increases plasma levels of known markers of endotheliopathy, leading to a reperfusion-induced coagulopathy compared with REBOA Zone 3 and no REBOA.
Collapse
Affiliation(s)
| | - Ernest E Moore
- Department of Surgery, University of Colorado, Aurora, CO USA
- Ernest E Moore Shock Trauma Center at Denver Health Medical Center Surgery, Denver, CO USA
| | - Ian LaCroix
- Department of Proteomics and Metabolomics, University of Colorado, Aurora, CO USA
| | - TJ Schaid
- Department of Surgery, University of Colorado, Aurora, CO USA
| | - Otto Thielen
- Department of Surgery, University of Colorado, Aurora, CO USA
| | - William Hallas
- Department of Surgery, University of Colorado, Aurora, CO USA
| | - Patrick Hom
- Department of Surgery, University of Colorado, Aurora, CO USA
| | | | | | - Kirk Hansen
- Department of Proteomics and Metabolomics, University of Colorado, Aurora, CO USA
| | - Mitchell Cohen
- Department of Surgery, University of Colorado, Aurora, CO USA
| | - Christopher Silliman
- Vitalant Research Institute, Denver, CO USA
- Department of Pediatrics, University of Colorado, Aurora, CO USA
| | - Angela Sauaia
- Department of Health Systems, Management and Policy, School of Public Health, University of Colorado Denver, Aurora, CO, USA
| | - Charles J Fox
- Department of Vascular Surgery, University of Maryland Vascular Surgery Baltimore, MD USA
| |
Collapse
|
27
|
Nan Z, Soh S, Shim JK, Kim HB, Yang YS, Kwak YL, Song JW. Effect of 5% albumin on endothelial glycocalyx degradation during off-pump coronary artery bypass. Can J Anaesth 2024; 71:244-253. [PMID: 37989943 DOI: 10.1007/s12630-023-02652-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 07/23/2023] [Accepted: 08/08/2023] [Indexed: 11/23/2023] Open
Abstract
PURPOSE The integrity of the endothelial glycocalyx (EG), a critical player in vascular homeostasis, reportedly influences the outcomes of critically ill patients. We investigated the effect of 5% albumin, which preserved EG integrity in preclinical studies, vs balanced crystalloid solution on EG degradation in patients undergoing off-pump coronary surgery. METHODS Patients were randomized to receive either 5% albumin (N = 51) or balanced crystalloid solution (Plasma-Lyte [Baxter Incorporated, Seoul, Republic of Korea]; N = 53) for intravenous volume replacement during surgery (double-blinded). The primary outcome was plasma syndecan-1 concentration, a marker of EG degradation, measured after anesthetic induction (baseline), completion of grafting, and sternal closure. Secondary outcomes were atrial natriuretic peptide (ANP), tumour necrosis factor (TNF)-α, soluble thrombomodulin, and perioperative fluid balance. RESULTS The mean (standard deviation) fluid requirements were 833 (270) mL and 1,323 (492) mL in the albumin and Plasma-Lyte group, respectively (mean difference, -489 mL; 95% confidence interval [CI], -643 to -335; P < 0.001). Plasma syndecan-1 concentration increased after completion of grafting (median difference, 116 ng·mL-1; 95% CI, 67 to 184; P < 0.001) and sternal closure (median difference, 57 ng·mL-1; 95% CI, 36 to 80; P < 0.001) compared with those at baseline, without any intergroup differences. Atrial natriuretic peptide, TNF-α, and soluble thrombomodulin concentrations were similar between the two groups. The amount of chest tube drainage was greater in the albumin group than that in the Plasma-Lyte group (median difference, 190 mL; 95% CI, 18 to 276; P = 0.03). CONCLUSION Off-pump coronary surgery was associated with significant EG degradation. Yet, intraoperative fluid therapy with 5% albumin could not ameliorate EG degradation when compared with balanced crystalloid solution. TRIAL REGISTRATION ClinicalTrials.gov (NCT03699462); first posted 9 October 2018.
Collapse
Affiliation(s)
- Zhengyu Nan
- Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sarah Soh
- Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jae-Kwang Shim
- Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hye Bin Kim
- Department of Anesthesiology and Pain Medicine, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Yun Seok Yang
- Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Young Lan Kwak
- Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jong Wook Song
- Department of Anesthesiology and Pain Medicine, Anesthesia and Pain Research Institute, Yonsei Cardiovascular Hospital, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
| |
Collapse
|
28
|
Buzzard L, Schreiber M. Trauma-induced coagulopathy: What you need to know. J Trauma Acute Care Surg 2024; 96:179-185. [PMID: 37828662 DOI: 10.1097/ta.0000000000004170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
ABSTRACT Trauma-induced coagulopathy (TIC) is a global inflammatory state accompanied by coagulation derangements, acidemia, and hypothermia, which occurs after traumatic injury. It occurs in approximately 25% of severely injured patients, and its incidence is directly related to injury severity. The mechanism of TIC is multifaceted; proposed contributing factors include dysregulation of activated protein C, increased tPA, systemic endothelial activation, decreased fibrinogen, clotting factor consumption, and platelet dysfunction. Effects of TIC include systemic inflammation, coagulation derangements, acidemia, and hypothermia. Trauma-induced coagulopathy may be diagnosed by conventional coagulation tests including platelet count, Clauss assay, international normalized ratio, thrombin time, prothrombin time, and activated partial thromboplastin time; viscoelastic hemostatic assays such as thrombelastography and rotational thrombelastography; or a clinical scoring system known as the Trauma Induced Coagulopathy Clinical Score. Preventing TIC begins in the prehospital phase with early hemorrhage control, blood product resuscitation, and tranexamic acid therapy. Early administration of prothrombin complex concentrate is also being studied in the prehospital environment. The mainstays of TIC treatment include hemorrhage control, blood and component transfusions, and correction of abnormalities such as hypocalcemia, acidosis, and hypothermia. LEVEL OF EVIDENCE Therapeutic/Care Management; Level III.
Collapse
Affiliation(s)
- Lydia Buzzard
- From the University of Wisconsin School of Medicine and Public Health (L.B.), Madison, Wisconsin; and Department of Surgery (L.B., M.S.), Oregon Health and Science University, Portland, Oregon
| | | |
Collapse
|
29
|
Dixon A, Kenny JE, Buzzard L, Holcomb J, Bulger E, Wade C, Fabian T, Schreiber M. Acute respiratory distress syndrome, acute kidney injury, and mortality after trauma are associated with increased circulation of syndecan-1, soluble thrombomodulin, and receptor for advanced glycation end products. J Trauma Acute Care Surg 2024; 96:319-325. [PMID: 37678154 DOI: 10.1097/ta.0000000000004096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/09/2023]
Abstract
BACKGROUND Disruption of the vascular endothelium and endothelial glycocalyx (EG) has been described after severe trauma. Plasma has been suggested to restore microvascular integrity by preservation and repair of the EG. We sought to evaluate whether plasma administered in a 1:1:1 ratio was associated with less endothelial marker circulation than a 1:1:2 ratio. METHODS This is a secondary analysis of the PROPPR trial, which investigated post-traumatic resuscitation with platelets, plasma, and red blood cells in a 1:1:1 ratio compared with a 1:1:2 ratio. Syndecan-1, soluble thrombomodulin (sTM), and receptor for advanced glycation end products (RAGE) were quantified for each treatment group on admission and at 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, and 72 hours. Patients were excluded if they did not survive longer than 3 hours or had data from fewer than two time points. RESULTS Three hundred eight patients in the 1:1:1 group and 291 in the 1:1:2 group were analyzed. There were no statistically significant differences in syndecan-1, sTM, or RAGE between treatment groups at any time point ( p > 0.05). Patients who developed acute respiratory distress syndrome, acute kidney injury, and death had significantly elevated biomarker expression at most time points when compared with patients who did not develop these sequelae ( p < 0.05). CONCLUSION Administration of FFP in a 1:1:1 ratio does not consistently affect circulation of endothelial biomarkers following significant trauma when compared with a 1:1:2 ratio. The development of post-traumatic ARDS, AKI, and death was associated with increased endothelial biomarker circulation. LEVEL OF EVIDENCE Therapeutic/Care Management; Level III.
Collapse
Affiliation(s)
- Alexandra Dixon
- From the Division of Trauma, Critical Care and Acute Care Surgery, Department of Surgery (A.D., J.E.K., L.B., M.S.), Oregon Health & Science University, Portland, Oregon; Division of Trauma and Acute Care Surgery, Department of Surgery (J.H.), University of Alabama at Birmingham, Birmingham, Alabama; Division of Trauma and Critical Care, Department of Surgery (E.B.), University of Washington, Seattle, Washington; Center for Translational Injury Research, Division of Acute Care Surgery, Department of Surgery (C.W.), University of Texas Health Science Center, Houston, Texas; and Division of Trauma and Surgical Critical Care, Department of Surgery (T.F.), University of Tennessee Health Science Center, Memphis, Tennessee
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Harriott NC, Ryan AL. Proteomic profiling identifies biomarkers of COVID-19 severity. Heliyon 2024; 10:e23320. [PMID: 38163173 PMCID: PMC10755324 DOI: 10.1016/j.heliyon.2023.e23320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 11/27/2023] [Accepted: 11/30/2023] [Indexed: 01/03/2024] Open
Abstract
SARS-CoV-2 infection remains a major public health concern, particularly for the aged and those individuals with co-morbidities at risk for developing severe COVID-19. Understanding the pathogenesis and biomarkers associated with responses to SARS-CoV-2 infection remain critical components in developing effective therapeutic approaches, especially in cases of severe and long-COVID-19. In this study blood plasma protein expression was compared in subjects with mild, moderate, and severe COVID-19 disease. Evaluation of an inflammatory protein panel confirms upregulation of proteins including TNFβ, IL-6, IL-8, IL-12, already associated with severe cytokine storm and progression to severe COVID-19. Importantly, we identify several proteins not yet associated with COVID-19 disease, including mesothelin (MSLN), that are expressed at significantly higher levels in severe COVID-19 subjects. In addition, we find a subset of markers associated with T-cell and dendritic cell responses to viral infection that are significantly higher in mild cases and decrease in expression as severity of COVID-19 increases, suggesting that an immediate and effective activation of T-cells is critical in modulating disease progression. Together, our findings identify new targets for further investigation as therapeutic approaches for the treatment of SARS-CoV-2 infection and prevention of complications of severe COVID-19.
Collapse
Affiliation(s)
- Noa C. Harriott
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles CA 90033, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles CA 90033, USA
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City IA 52240, USA
| | - Amy L. Ryan
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles CA 90033, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles CA 90033, USA
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City IA 52240, USA
| |
Collapse
|
31
|
Anand T, Crawford AE, Sjoquist M, Hashmi ZG, Richter RP, Joseph B, Richter JR. Decreased Glycocalyx Shedding on Presentation in Hemorrhaging Geriatric Trauma Patients. J Surg Res 2024; 293:709-716. [PMID: 37844411 PMCID: PMC11075129 DOI: 10.1016/j.jss.2023.09.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 08/21/2023] [Accepted: 09/04/2023] [Indexed: 10/18/2023]
Abstract
INTRODUCTION Plasma levels of syndecan-1 (Sdc-1), a biomarker of endothelial glycocalyx (EG) damage, correlate with worse outcomes in trauma patients. However, EG injury is not well characterized in injured older adults (OA). The aims of this study were to characterize Sdc-1 shedding in OA trauma patients relative to younger adults (YA) and determine associations with putative regulators of EG sheddases. METHODS We performed a secondary analysis of data from the Pragmatic, Randomized Optimal Platelet, and Plasma Ratios (PROPPR) trial, stratifying bluntly injured subjects into OA and YA groups based on upper age quartile (57 y). Plasma Sdc-1 levels were compared in OA and YA at hospital arrival through postinjury day 3, and the independent association between age and Sdc-1 level at arrival was determined after adjusting for differences in gender, shock index (SI), and pre-existing comorbidities. In a follow-up analysis, case-control matching was used to create populations of OA and YA with equivalent SI and injury severity score. Levels of Sdc-1 were compared between these matched groups, and the relationships with candidate regulators of EG shedding were assessed. RESULTS Of 680 subjects in the Pragmatic, Randomized Optimal Platelet, and Plasma Ratios trial, 350 (51%) had blunt injuries, and 92 (26.3%) of these were OA. Plasma Sdc-1 levels at arrival, 2 h, and 6 h were significantly lower in OA compared to YA (all P < 0.05). After adjusting for sex, pre-existing morbidities and SI, age was associated with decreased Sdc-1 levels at arrival. In the matched analyses, Sdc-1, high-mobility group box 1 and tissue inhibitor of metalloproteinase-2 levels were lower in OA compared to YA. Both high-mobility group box-1 and tissue inhibitor of metalloproteinase-2 significantly correlated with arrival Sdc-1 and were inversely associated with age. CONCLUSIONS This study indicates that increased age is independently associated with decreased Sdc-1 levels among patients with blunt injuries. Suppressed plasma levels of sheddases in relation to diminished Sdc-1 shedding suggest that mechanisms regulating EG cleavage may be impaired in injured older adults. These findings provide novel insight into the age-dependent impact of injury on the vascular endothelium, which could have important implications for the clinical management of older adults following trauma.
Collapse
Affiliation(s)
- Tanya Anand
- Division of Trauma, Critical Care, Burn & Emergency Surgery, Department of Surgery, University of Arizona, Tucson, Arizona. https://twitter.com/tanyaanand8
| | - Anna E Crawford
- University of Alabama at Birmingham Heersink College of Medicine, Birmingham, Alabama
| | | | - Zain G Hashmi
- Division of Trauma & Acute Care Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama; Center for Injury Science, University of Alabama at Birmingham, Birmingham, Alabama
| | - Robert P Richter
- Center for Injury Science, University of Alabama at Birmingham, Birmingham, Alabama; Division of Pediatric Critical Care Medicine, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama
| | - Bellal Joseph
- Division of Trauma, Critical Care, Burn & Emergency Surgery, Department of Surgery, University of Arizona, Tucson, Arizona
| | - Jillian R Richter
- Division of Trauma & Acute Care Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama; Center for Injury Science, University of Alabama at Birmingham, Birmingham, Alabama.
| |
Collapse
|
32
|
Iba T, Helms J, Neal MD, Levy JH. Mechanisms and management of the coagulopathy of trauma and sepsis: trauma-induced coagulopathy, sepsis-induced coagulopathy, and disseminated intravascular coagulation. J Thromb Haemost 2023; 21:3360-3370. [PMID: 37722532 PMCID: PMC10873124 DOI: 10.1016/j.jtha.2023.05.028] [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: 01/18/2023] [Revised: 04/18/2023] [Accepted: 05/12/2023] [Indexed: 09/20/2023]
Abstract
Disseminated intravascular coagulation can occur due to different causes but commonly following sepsis. Trauma-induced coagulopathy (TIC) occurs on hospital arrival in approximately 25% of seriously injured patients who initially presents with impaired hemostasis and a bleeding phenotype that can later progress to a prothrombotic phase. Following traumatic injury, ineffective hemostasis is driven by massive blood loss, tissue damage, and hyperfibrinolysis. This initial impaired hemostasis continues until surgical or other management strategies not only to stop the causes of hemorrhage but also progresses to a prothrombotic and hypofibrinolytic state, also termed fibrinolytic shutdown. Prothrombotic progression is also promoted by inflammatory mediator release, endothelial injury, and platelet dysregulation, which is commonly seen in sepsis with increased mortality. Unlike TIC, the early phase of sepsis is frequently complicated by multiorgan dysfunction described as sepsis-induced coagulopathy (SIC) that lacks a hemorrhagic phase. The phenotypes of SIC and TIC are different, especially in their initial presentations; however, patients who survive TIC may also develop subsequent infections and potentially sepsis and SIC. Although the pathophysiology of SIC and TIC are different, endothelial injury, dysregulated fibrinolysis, and coagulation abnormalities are common. Management includes treatment of the underlying cause, tissue injury vs infection is critical, and supportive therapies, such as hemostatic resuscitation and circulatory support are essential, and adjunct therapies are recommended in guidelines. Based on clinical studies and certain guidelines, additional therapies include tranexamic acid in the limited timing of initial traumatic injury and anticoagulants, such as antithrombin and recombinant thrombomodulin in disseminated intravascular coagulation.
Collapse
Affiliation(s)
- Toshiaki Iba
- Department of Emergency and Disaster Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan.
| | - Julie Helms
- Strasbourg University (UNISTRA); Strasbourg University Hospital, Medical Intensive Care Unit - NHC; INSERM (French National Institute of Health and Medical Research), Strasbourg, France
| | - Matthew D Neal
- Trauma and Transfusion Medicine Research Center, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jerrold H Levy
- Department of Anesthesiology, Critical Care, and Surgery, Duke University School of Medicine, Durham, North Carolina, USA. https://twitter.com/JerroldLevy
| |
Collapse
|
33
|
Juusela A, Jung E, Gallo DM, Bosco M, Suksai M, Diaz-Primera R, Tarca AL, Than NG, Gotsch F, Romero R, Tinnakorn Chaiworapongsa. Maternal plasma syndecan-1: a biomarker for fetal growth restriction. J Matern Fetal Neonatal Med 2023; 36:2150074. [PMID: 36597808 PMCID: PMC10291740 DOI: 10.1080/14767058.2022.2150074] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 11/14/2022] [Indexed: 01/05/2023]
Abstract
OBJECTIVE The identification of fetal growth disorders is an important clinical priority given that they increase the risk of perinatal morbidity and mortality as well as long-term diseases. A subset of small-for-gestational-age (SGA) infants are growth-restricted, and this condition is often attributed to placental insufficiency. Syndecan-1, a product of the degradation of the endothelial glycocalyx, has been proposed as a biomarker of endothelial damage in different pathologies. During pregnancy, a "specialized" form of the glycocalyx-the "syncytiotrophoblast glycocalyx"-covers the placental villi. The purpose of this study was to determine whether the concentration of maternal plasma syndecan-1 can be proposed as a biomarker for fetal growth restriction. STUDY DESIGN A cross-sectional study was designed to include women with normal pregnancy (n = 130) and pregnant women who delivered an SGA neonate (n = 50). Doppler velocimetry of the uterine and umbilical arteries was performed in women with an SGA fetus at the time of diagnosis. Venipuncture was performed within 48 h of Doppler velocimetry and plasma concentrations of syndecan-1 were determined by a specific and sensitive immunoassay. RESULTS (1) Plasma syndecan-1 concentration followed a nonlinear increase with gestational age in uncomplicated pregnancies (R2 = 0.27, p < .001); (2) women with a pregnancy complicated with an SGA fetus had a significantly lower mean plasma concentration of syndecan-1 than those with an appropriate-for-gestational-age fetus (p = .0001); (3) this difference can be attributed to fetal growth restriction, as the mean plasma syndecan-1 concentration was significantly lower only in the group of women with an SGA fetus who had abnormal umbilical and uterine artery Doppler velocimetry compared to controls (p = .00071; adjusted p = .0028). A trend toward lower syndecan-1 concentrations was also noted for SGA with abnormal uterine but normal umbilical artery Doppler velocimetry (p = .0505; adjusted p = .067); 4) among women with an SGA fetus, those with abnormal umbilical and uterine artery Doppler findings had a lower mean plasma syndecan-1 concentration than women with normal Doppler velocimetry (p = .02; adjusted p = .04); 5) an inverse relationship was found between the maternal plasma syndecan-1 concentration and the umbilical artery pulsatility index (r = -0.5; p = .003); and 6) a plasma syndecan-1 concentration ≤ 850 ng/mL had a positive likelihood ratio of 4.4 and a negative likelihood ratio of 0.24 for the identification of a mother with an SGA fetus who had abnormal umbilical artery Doppler velocimetry (area under the ROC curve 0.83; p < .001). CONCLUSION Low maternal plasma syndecan-1 may reflect placental diseases and this protein could be a biomarker for fetal growth restriction. However, as a sole biomarker for this condition, its accuracy is low.
Collapse
Affiliation(s)
- Alexander Juusela
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Eunjung Jung
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Dahiana M. Gallo
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Mariachiara Bosco
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Manaphat Suksai
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Ramiro Diaz-Primera
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Adi L. Tarca
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Computer Science, Wayne State University College of Engineering, Detroit, Michigan, USA
| | - Nandor Gabor Than
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
- Maternity Private Clinic, Budapest, Hungary
| | - Francesca Gotsch
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, Michigan, USA
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan, USA
- Detroit Medical Center, Detroit, Michigan, USA
| | - Tinnakorn Chaiworapongsa
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| |
Collapse
|
34
|
Cakir MU, Yavuz-Aksu B, Aksu U. Hypervolemia suppresses dilutional anaemic injury in a rat model of haemodilution. J Transl Int Med 2023; 11:393-400. [PMID: 38130643 PMCID: PMC10732576 DOI: 10.2478/jtim-2022-0045] [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: 11/16/2022] Open
Abstract
Background and Objectives Haemodilution leads to complications in clinical practice. It is exactly unknown whether this damage is caused by the fluid or by the stretching of the vascular bed. We aimed to compare two different haemodilution techniques at the same anaemic level. Methods Normovolemic or hypervolemic haemodilution was performed on twelve adult male Wistar rats. In the normovolemic procedure, blood was withdrawn and instantaneously administered with similar amounts of 6% hydroxyethyl starch (HES 130/0.4). Fluid was administered without withdrawing blood in the hypervolemic procedure. In both models, a 25% haematocrit level was targeted and kept at this level for 90 min to deepen the anaemia effect. Besides haemodynamics measurement, renal function (creatinine, blood urea nitrogen) and injury (tissue norepinephrine, malondialdehyde) were evaluated. Also, systemic hypoxia (lactate), oxidative stress (malondialdehyde, ischaemia-modified albumin), inflammation (tumour necrosis factor-alpha [TNF-α]), osmotic stress, adrenal stress (norepinephrine, epinephrine), and vascular stretching (atrial natriuretic peptide [ANP]) were assessed. Results Arterial pressure in the normovolemic group was lower than in the hypervolemic group. Serum creatinine, blood urea nitrogen, and lactate levels were higher in the normovolemic group. Tissue norepinephrine and malondialdehyde levels were higher in the normovolemic group. Serum ANP, malondialdehyde, ischaemia-modified albumin, free haemoglobin, syndecan-1, and TNF-α were higher in both groups compared to respective baseline. Conclusions Normovolemic haemodilution may lead to hypoxic kidney injury. The hypervolemic state may be advantageous if fluid is to be administered. Thus, the effect of the fluid itself can be relatively masked.
Collapse
Affiliation(s)
- Muzaffer Utku Cakir
- Department of Biology, Faculty of Science, University of Istanbul, Istanbul34134, Turkey
| | - Berna Yavuz-Aksu
- Duzen Laboratory Group, Biochemistry Section, Istanbul34394, Turkey
| | - Ugur Aksu
- Department of Biology, Faculty of Science, University of Istanbul, Istanbul34134, Turkey
| |
Collapse
|
35
|
Gomez Builes JC, Baker AJ, Callum J, Barahi S, Bai J, Karkouti K, Nisenbaum R, Sholzberg M. Evaluation of the association of factor XIII at hospital arrival and outcomes in a cohort of severely injured patients. J Thromb Haemost 2023; 21:3085-3098. [PMID: 37453456 DOI: 10.1016/j.jtha.2023.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 07/03/2023] [Accepted: 07/05/2023] [Indexed: 07/18/2023]
Abstract
BACKGROUND Severe traumatic bleeding depletes coagulation factor XIII (FXIII) and fibrinogen. However, the role of FXIII level in bleeding-related outcomes is unknown. OBJECTIVES To evaluate the association between FXIII levels at hospital arrival and critical administration threshold (≥3 red blood cell units in 1 hour within the first 24 hours), bleeding-related outcomes, death, and baseline characteristics. METHODS A retrospective cohort study was conducted in severely injured adult patients (Injury Severity Score of ≥22 or ≥2 red blood cell units transfused in 24 hours) admitted to a level 1 trauma center. Clinical and laboratory data were collected. Baseline FXIII antigen levels were measured in banked patient plasma. Multivariable logistic and linear regression models were used to estimate the association between FXIII levels, outcomes, and baseline characteristics. RESULTS Three hundred sixty-four of 1730 subjects admitted during a 2-year period were analyzed. Median age was 44 years (IQR, 27-62 years), and median Injury Severity Score was 29 (IQR, 22-34). FXIII levels were not associated with critical administration threshold (odds ratio [OR], 1.06; 95% CI, 0.97-1.17) or death (OR, 0.98; 95% CI, 0.90-1.07). FXIII was associated with major bleeding (OR, 1.10; 95% CI, 1.02-1.2) and massive transfusion (OR, 1.25; 95% CI, 1.08-1.44). Lower baseline FXIII levels were associated with arrival from a referring hospital (FXIII level, -0.07 U/mL; 95% CI, -0.11 to -0.03), hemoglobin (FXIII level, -0.05 U/mL; 95% CI, -0.07 to -0.03), fibrinogen level (FXIII level, -0.05 U/mL; 95% CI, -0.08 to -0.02), and platelet count (FXIII level, -0.02 U/mL; 95% CI, -0.04 to -0.008). CONCLUSIONS Baseline FXIII levels in severely injured patients were inconsistently associated with bleeding-related outcomes and mortality. However, their association with major bleeding warrants further investigation of the role of FXIII in massively transfused patients with trauma.
Collapse
Affiliation(s)
- Johana Carolina Gomez Builes
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada; Department of Anesthesia and Pain Medicine, St. Michael's Hospital, Toronto, Ontario, Canada; Department of Anesthesia and Pain Medicine, University of Toronto, Toronto, Ontario, Canada. https://twitter.com/cgomezbuiles
| | - Andrew J Baker
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada; Department of Anesthesia and Pain Medicine, St. Michael's Hospital, Toronto, Ontario, Canada; Department of Anesthesia and Pain Medicine, University of Toronto, Toronto, Ontario, Canada; Interdepartmental Division of Critical Care, University of Toronto, Toronto, Ontario, Canada; Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
| | - Jeannie Callum
- Department of Pathology and Molecular Medicine, Kingston Health Sciences Centre, Kingston, Ontario, Canada; Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - Sunti Barahi
- Department of Anesthesia and Pain Medicine, St. Michael's Hospital, Toronto, Ontario, Canada; Department of Anesthesia and Pain Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Johnny Bai
- Department of Anesthesia and Pain Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Keyvan Karkouti
- Department of Anesthesia and Pain Medicine, University of Toronto, Toronto, Ontario, Canada; Department of Anesthesia and Pain Management, University Health Network, Sinai Health System, and Women's College Hospital, Toronto, Ontario, Canada
| | - Rosane Nisenbaum
- Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada; MAP Centre for Urban Health Solutions, Toronto, Ontario, Canada; Applied Health Research Centre, Toronto, Ontario, Canada; Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Michelle Sholzberg
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada; Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada; Department of Hematology, St. Michael's Hospital, Toronto, Ontario, Canada; Institute of Health Policy, Management, and Evaluation, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
36
|
Baucom MR, Wallen TE, Price AD, Smith MP, Kopchak M, MacKinnon A, Weissman N, Schuster RM, Pritts TA, Goodman MD. Predictive Value of Early Inflammatory Markers in Trauma Patients Based on Transfusion Status. J Surg Res 2023; 291:691-699. [PMID: 37562231 PMCID: PMC10530444 DOI: 10.1016/j.jss.2023.07.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 07/02/2023] [Accepted: 07/07/2023] [Indexed: 08/12/2023]
Abstract
INTRODUCTION Seven key inflammatory biomarkers were recently found to be associated with the risk of mortality in a multicenter study of massively transfused patients. The aim of this prospective single-center study was to determine which of these early inflammatory markers could predict 30-d mortality among all critically injured trauma patients. METHODS Serum samples were collected at 6, 24, and 72 h from 238 consecutive patients admitted to the intensive care unit following traumatic injury. Inflammatory markers syndecan-1, eotaxin, IL-1ra, IL-6, IL-8, IL-10, IP-10, and MCP-1 were analyzed via multiplex enzyme-linked immunosorbent assay. Subgroup analysis was performed for patients undergoing massive transfusion (≥5 red blood cells), submassive transfusion (1-4 red blood cells), or no transfusion during the first 4 h postinjury. The primary outcome of 30-d survival was modeled as a function of each biomarker and confounders using repeat measures logistic regression. RESULTS Patients had a median age of 51.3 y [33.7, 70.2], 70.6% were male, 17.4% experienced penetrating trauma, and had a median injury severity score of 22 [14, 33]. IL-1ra, IL-8, IL-10, and MCP-1 were significantly increased during the first 72 h in nonsurvivors (n = 31). Elevated IL-1ra, IL-8, IL-10, and MCP-1 at 6 h postinjury were associated with 30-d mortality. By contrast, serum syndecan-1 and eotaxin levels were not associated with mortality at any time point. IL-8 and lactate were increased at 6 h in 30-d nonsurvivors for patients receiving submassive transfusion (n = 78). CONCLUSIONS Early evaluations of IL-1ra, IL-8, IL-10, and IP-10 within 6 h of injury are useful predictors of 30-d mortality. Subgroup analysis suggests that transfusion status does not significantly affect early inflammatory markers. LEVEL OF EVIDENCE Level III, prognostic/epidemiological.
Collapse
Affiliation(s)
- Matthew R Baucom
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio
| | - Taylor E Wallen
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio
| | - Adam D Price
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio
| | - Maia P Smith
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio
| | - Maura Kopchak
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio
| | - Andrew MacKinnon
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio
| | - Nick Weissman
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio
| | | | - Timothy A Pritts
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio
| | | |
Collapse
|
37
|
Douin DJ, Fernandez-Bustamante A. Early Fibrinogen Replacement to Treat the Endotheliopathy of Trauma: Novel Resuscitation Strategies in Severe Trauma. Anesthesiology 2023; 139:675-683. [PMID: 37815472 PMCID: PMC10575674 DOI: 10.1097/aln.0000000000004711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/11/2023]
Abstract
The authors provide a comprehensive review of the endothelial glycocalyx, the components that may be targeted to improve clinical outcomes, and the next steps for evaluation in human subjects.
Collapse
Affiliation(s)
- David J Douin
- Department of Anesthesiology, University of Colorado School of Medicine, Aurora, Colorado
| | | |
Collapse
|
38
|
Thaler S, Stöhr D, Kammerer T, Nitschke T, Hoechter DJ, Brandes F, Müller M, Groene P, Schäfer ST. Predictive value of coagulation variables and glycocalyx shedding in hospitalized COVID-19 patients - a prospective observational study. Acta Clin Belg 2023; 78:392-400. [PMID: 37092324 DOI: 10.1080/17843286.2023.2204593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 04/15/2023] [Indexed: 04/25/2023]
Abstract
OBJECTIVES Covid-19 disease causes an immense burden on the healthcare system. It has not yet been finally clarified which patients will suffer from a severe course and which will not. Coagulation disorders can be detected in many of these patients. The aim of the present study was therefore to identify variables of the coagulation system including standard and viscoelastometric tests as well as components of glycocalyx damage that predict admission to the intensive care unit. METHODS Adult patients were included within 24 h of admission. Blood samples were analyzed at hospital admission and at ICU admission if applicable. We analyzed group differences and furthermore performed receiver operator characteristics (ROC). RESULTS This study included 60 adult COVID-19 patients. During their hospital stay, 14 patients required ICU treatment. Comparing ICU and non-ICU patients at time of hospital admission, D-dimer (1450 µg/ml (675/2850) vs. 600 µg/ml (500/900); p = 0.0022; cut-off 1050 µg/ml, sensitivity 71%, specificity 89%) and IL-6 (47.6 pg/ml (24.9/85.4 l) vs. 16.1 pg/ml (5.5/34.4); p = 0.0003; cut-off 21.25 pg/ml, sensitivity 86%, specificity 65%) as well as c-reactive protein (92 mg/dl (66.8/131.5) vs. 43.5 mg/dl (26.8/83.3); p = 0.0029; cutoff 54.5 mg/dl, sensitivity 86%, specificity 65%) were higher in patients who required ICU admission. Thromboelastometric variables and markers of glycocalyx damage (heparan sulfate, hyaluronic acid, syndecan-1) at the time of hospital admission did not differ between groups. CONCLUSION General inflammatory variables continue to be the most robust predictors of a severe course of a COVID-19 infection. Viscoelastometric variables and markers of glycocalyx damage are significantly increased upon admission to the ICU without being predictors of ICU admission.
Collapse
Affiliation(s)
- Sarah Thaler
- Department of Anaesthesiology, University Hospital, Munich, Germany
| | - Dana Stöhr
- Department of Anaesthesiology, University Hospital, Munich, Germany
| | - Tobias Kammerer
- Department of Anaesthesiology, University Hospital, Munich, Germany
- Department of Anaesthesiology and Intensive Care Medicine, University Hospital of Cologne, Cologne, Germany
| | - Tobias Nitschke
- Department of Anaesthesiology, University Hospital, Munich, Germany
| | | | - Florian Brandes
- Department of Anaesthesiology, University Hospital, Munich, Germany
| | - Martin Müller
- Department of Anaesthesiology, University Hospital, Munich, Germany
| | - Philipp Groene
- Department of Anaesthesiology, University Hospital, Munich, Germany
| | - Simon T Schäfer
- Department of Anaesthesiology, University Hospital, Munich, Germany
| |
Collapse
|
39
|
Arteaga GM, Crow S. End organ perfusion and pediatric microcirculation assessment. Front Pediatr 2023; 11:1123405. [PMID: 37842022 PMCID: PMC10576530 DOI: 10.3389/fped.2023.1123405] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 09/05/2023] [Indexed: 10/17/2023] Open
Abstract
Cardiovascular instability and reduced oxygenation are regular perioperative critical events associated with anesthesia requiring intervention in neonates and young infants. This review article addresses the current modalities of assessing this population's adequate end-organ perfusion in the perioperative period. Assuring adequate tissue oxygenation in critically ill infants is based on parameters that measure acceptable macrocirculatory hemodynamic parameters such as vital signs (mean arterial blood pressure, heart rate, urinary output) and chemical parameters (lactic acidosis, mixed venous oxygen saturation, base deficit). Microcirculation assessment represents a promising candidate for assessing and improving hemodynamic management strategies in perioperative and critically ill populations. Evaluation of the functional state of the microcirculation can parallel improvement in tissue perfusion, a term coined as "hemodynamic coherence". Less information is available to assess microcirculatory disturbances related to higher mortality risk in critically ill adults and pediatric patients with septic shock. Techniques for measuring microcirculation have substantially improved in the past decade and have evolved from methods that are limited in scope, such as velocity-based laser Doppler and near-infrared spectroscopy, to handheld vital microscopy (HVM), also referred to as videomicroscopy. Available technologies to assess microcirculation include sublingual incident dark field (IDF) and sublingual sidestream dark field (SDF) devices. This chapter addresses (1) the physiological basis of microcirculation and its relevance to the neonatal and pediatric populations, (2) the pathophysiology associated with altered microcirculation and endothelium, and (3) the current literature reviewing modalities to detect and quantify the presence of microcirculatory alterations.
Collapse
Affiliation(s)
- Grace M. Arteaga
- Department of Pediatric and Adolescent Medicine, Pediatric Critical Care, Mayo Clinic, Rochester MN, United States
| | - Sheri Crow
- Department of Pediatric and Adolescent Medicine, Pediatric Critical Care, Mayo Clinic, Rochester MN, United States
| |
Collapse
|
40
|
Patterson PD, Friedman JC, Ding S, Miller RS, Martin-Gill C, Hostler D, Platt TE. Acute Effect of Night Shift Work on Endothelial Function with and without Naps: A Scoping Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:6864. [PMID: 37835134 PMCID: PMC10572584 DOI: 10.3390/ijerph20196864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/23/2023] [Accepted: 09/27/2023] [Indexed: 10/15/2023]
Abstract
We examined the breadth and depth of the current evidence investigating napping/sleeping during night shift work and its impact on non-invasive measures of endothelial function. We used a scoping review study design and searched five databases: Ovid Medline, EMBASE, Ovid APA PsycInfo, Web of Science Core Collection, and EBSCO CINAHL. We limited our search to English language and publications from January 1980 to September 2022. Our reporting adhered to the PRISMA-ScR guidance for scoping reviews. Our search strategy yielded 1949 records (titles and abstracts) after deduplication, of which 36 were retained for full-text review. Five articles were retained, describing three observational and two experimental research studies with a total sample of 110 individuals, which examined the non-invasive indicators of endothelial function in relation to the exposure to night shift work. While there is some evidence of an effect of night shift work on the non-invasive indicators of endothelial function, this evidence is incomplete, limited to a small samples of shift workers, and is mostly restricted to one measurement technique for assessing endothelial function with diverse protocols. In addition, there is no identifiable research investigating the potential benefits of napping during night shift work on non-invasive measures of endothelial function.
Collapse
Affiliation(s)
- Paul D. Patterson
- Department of Emergency Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Community Health Services and Rehabilitation Sciences, School of Health and Rehabilitation Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Jacob C. Friedman
- Department of Emergency Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Community Health Services and Rehabilitation Sciences, School of Health and Rehabilitation Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Samuel Ding
- Department of Emergency Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Community Health Services and Rehabilitation Sciences, School of Health and Rehabilitation Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Rebekah S. Miller
- Health Sciences Library System, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Christian Martin-Gill
- Department of Emergency Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - David Hostler
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, University at Buffalo, The State University of New York, Buffalo, NY 14214, USA;
| | - Thomas E. Platt
- Department of Community Health Services and Rehabilitation Sciences, School of Health and Rehabilitation Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
41
|
Anand T, Reyes AA, Sjoquist MC, Magnotti L, Joseph B. Resuscitating the Endothelial Glycocalyx in Trauma and Hemorrhagic Shock. ANNALS OF SURGERY OPEN 2023; 4:e298. [PMID: 37746602 PMCID: PMC10513357 DOI: 10.1097/as9.0000000000000298] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 05/20/2023] [Indexed: 09/26/2023] Open
Abstract
The endothelium is lined by a protective mesh of proteins and carbohydrates called the endothelial glycocalyx (EG). This layer creates a negatively charged gel-like barrier between the vascular environment and the surface of the endothelial cell. When intact the EG serves multiple functions, including mechanotransduction, cell signaling, regulation of permeability and fluid exchange across the microvasculature, and management of cell-cell interactions. In trauma and/or hemorrhagic shock, the glycocalyx is broken down, resulting in the shedding of its individual components. The shedding of the EG is associated with increased systemic inflammation, microvascular permeability, and flow-induced vasodilation, leading to further physiologic derangements. Animal and human studies have shown that the greater the severity of the injury, the greater the degree of shedding, which is associated with poor patient outcomes. Additional studies have shown that prioritizing certain resuscitation fluids, such as plasma, cryoprecipitate, and whole blood over crystalloid shows improved outcomes in hemorrhaging patients, potentially through a decrease in EG shedding impacting downstream signaling. The purpose of the following paragraphs is to briefly describe the EG, review the impact of EG shedding and hemorrhagic shock, and begin entertaining the notion of directed resuscitation. Directed resuscitation emphasizes transitioning from macroscopic 1:1 resuscitation to efforts that focus on minimizing EG shedding and maximizing its reconstitution.
Collapse
Affiliation(s)
- Tanya Anand
- From the Department of Surgery, Division of Trauma, Critical Care, Burns, and Emergency Surgery, The University of Arizona, Tucson, AZ
| | | | - Michael C. Sjoquist
- Department of Surgery, University of Arizona College of Medicine, Tucson, AZ
| | - Louis Magnotti
- From the Department of Surgery, Division of Trauma, Critical Care, Burns, and Emergency Surgery, The University of Arizona, Tucson, AZ
| | - Bellal Joseph
- From the Department of Surgery, Division of Trauma, Critical Care, Burns, and Emergency Surgery, The University of Arizona, Tucson, AZ
| |
Collapse
|
42
|
Appelman B, Michels EHA, de Brabander J, Peters-Sengers H, van Amstel RBE, Noordzij SM, Klarenbeek AM, van Linge CCA, Chouchane O, Schuurman AR, Reijnders TDY, Douma RA, Bos LDJ, Wiersinga WJ, van der Poll T. Thrombocytopenia is associated with a dysregulated host response in severe COVID-19. Thromb Res 2023; 229:187-197. [PMID: 37541167 DOI: 10.1016/j.thromres.2023.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 06/23/2023] [Accepted: 07/17/2023] [Indexed: 08/06/2023]
Abstract
BACKGROUND Thrombocytopenia is associated with increased mortality in COVID-19 patients. OBJECTIVE To determine the association between thrombocytopenia and alterations in host response pathways implicated in disease pathogenesis in patients with severe COVID-19. PATIENTS/METHODS We studied COVID-19 patients admitted to a general hospital ward included in a national (CovidPredict) cohort derived from 13 hospitals in the Netherlands. In a subgroup, 43 host response biomarkers providing insight in aberrations in distinct pathophysiological domains (coagulation and endothelial cell function; inflammation and damage; cytokines and chemokines) were determined in plasma obtained at a single time point within 48 h after admission. Patients were stratified in those with normal platelet counts (150-400 × 109/L) and those with thrombocytopenia (<150 × 109/L). RESULTS 6.864 patients were enrolled in the national cohort, of whom 1.348 had thrombocytopenia and 5.516 had normal platelets counts; the biomarker cohort consisted of 429 patients, of whom 85 with thrombocytopenia and 344 with normal platelet counts. Plasma D-dimer levels were not different in thrombocytopenia, although patients with moderate-severe thrombocytopenia (<100 × 109/L) showed higher D-dimer levels, indicating enhanced coagulation activation. Patients with thrombocytopenia had lower plasma levels of many proinflammatory cytokines and chemokines, and antiviral mediators, suggesting involvement of platelets in inflammation and antiviral immunity. Thrombocytopenia was associated with alterations in endothelial cell biomarkers indicative of enhanced activation and a relatively preserved glycocalyx integrity. CONCLUSION Thrombocytopenia in hospitalized patients with severe COVID-19 is associated with broad host response changes across several pathophysiological domains. These results suggest a role of platelets in the immune response during severe COVID-19.
Collapse
Affiliation(s)
- Brent Appelman
- Amsterdam UMC location University of Amsterdam, Center for Experimental and Molecular Medicine, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands.
| | - Erik H A Michels
- Amsterdam UMC location University of Amsterdam, Center for Experimental and Molecular Medicine, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Justin de Brabander
- Amsterdam UMC location University of Amsterdam, Center for Experimental and Molecular Medicine, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Hessel Peters-Sengers
- Amsterdam UMC location University of Amsterdam, Center for Experimental and Molecular Medicine, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Epidemiology and Data Science, Boelelaan 1117, Amsterdam, the Netherlands
| | - Rombout B E van Amstel
- Amsterdam UMC location University of Amsterdam, Department of Intensive Care Medicine, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Sophie M Noordzij
- Amsterdam UMC location University of Amsterdam, Center for Experimental and Molecular Medicine, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Augustijn M Klarenbeek
- Amsterdam UMC location University of Amsterdam, Center for Experimental and Molecular Medicine, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Christine C A van Linge
- Amsterdam UMC location University of Amsterdam, Center for Experimental and Molecular Medicine, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Osoul Chouchane
- Amsterdam UMC location University of Amsterdam, Center for Experimental and Molecular Medicine, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Alex R Schuurman
- Amsterdam UMC location University of Amsterdam, Center for Experimental and Molecular Medicine, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Tom D Y Reijnders
- Amsterdam UMC location University of Amsterdam, Center for Experimental and Molecular Medicine, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Renée A Douma
- Flevo Hospital, Department of Internal Medicine, Almere, the Netherlands
| | - Lieuwe D J Bos
- Amsterdam UMC location University of Amsterdam, Department of Intensive Care Medicine, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - W Joost Wiersinga
- Amsterdam UMC location University of Amsterdam, Center for Experimental and Molecular Medicine, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Amsterdam UMC location University of Amsterdam, Division of Infectious Diseases, Department of Medicine, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Tom van der Poll
- Amsterdam UMC location University of Amsterdam, Center for Experimental and Molecular Medicine, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Amsterdam UMC location University of Amsterdam, Division of Infectious Diseases, Department of Medicine, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| |
Collapse
|
43
|
Park S, Newton J, Hidjir T, Young EWK. Bidirectional airflow in lung airway-on-a-chip with matrix-derived membrane elicits epithelial glycocalyx formation. LAB ON A CHIP 2023; 23:3671-3682. [PMID: 37462986 DOI: 10.1039/d3lc00259d] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/09/2023]
Abstract
Organ-on-a-chip systems are rapidly advancing as a viable alternative to existing experimental models in respiratory research. To date, however, epithelial cell cultures within lung airway-on-a-chip devices have yet to demonstrate the presence of an epithelial glycocalyx, a thin layer of proteoglycans, glycoproteins, and glycolipids known to play an important role in regulating epithelial function. Here, we demonstrate that an airway-on-a-chip device that incorporates bidirectional flow mimicking breathing cycles in combination with an ultra-thin matrix-derived membrane (UMM) layer can generate a glycocalyx layer comprised of heparan sulfate. Results with this device and airflow system showed dramatic differences of airway epithelial cell viability and expression of tight junctions, cilia, and mucus over a wide range of flow rates when cultured under oscillatory flow. More importantly, for the first time in a microfluidic organ-on-a-chip setting, we achieved the visualization of an airflow-induced epithelial glycocalyx layer. Our experiments highlight the importance of physiological mimicry in developing in vitro models, as bidirectional airflow showed more representative mucociliary differentiation compared to continuous unidirectional airflow. Thus, the lung airway-on-a-chip platform demonstrated in this study holds great potential as a lung epithelial barrier model for studying the mechanisms of various respiratory diseases and for testing the efficacy of therapeutic candidates in the presence of bidirectional airflow and the glycocalyx.
Collapse
Affiliation(s)
- Siwan Park
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3G9, Canada.
| | - Jeremy Newton
- Department of Mechanical & Industrial Engineering, University of Toronto, Toronto, ON, M5S 3G8, Canada
| | - Tesnime Hidjir
- Department of Mechanical & Industrial Engineering, University of Toronto, Toronto, ON, M5S 3G8, Canada
| | - Edmond W K Young
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3G9, Canada.
- Department of Mechanical & Industrial Engineering, University of Toronto, Toronto, ON, M5S 3G8, Canada
| |
Collapse
|
44
|
Ying J, Zhang C, Wang Y, Liu T, Yu Z, Wang K, Chen W, Zhou Y, Lu G. Sulodexide improves vascular permeability via glycocalyx remodelling in endothelial cells during sepsis. Front Immunol 2023; 14:1172892. [PMID: 37614234 PMCID: PMC10444196 DOI: 10.3389/fimmu.2023.1172892] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 07/21/2023] [Indexed: 08/25/2023] Open
Abstract
Background Degradation of the endothelial glycocalyx is critical for sepsis-associated lung injury and pulmonary vascular permeability. We investigated whether sulodexide, a precursor for the synthesis of glycosaminoglycans, plays a biological role in glycocalyx remodeling and improves endothelial barrier dysfunction in sepsis. Methods The number of children with septic shock that were admitted to the PICU at Children's Hospital of Fudan University who enrolled in the study was 28. On days one and three after enrollment, venous blood samples were collected, and heparan sulfate, and syndecan-1 (SDC1) were assayed in the plasma. We established a cell model of glycocalyx shedding by heparinase III and induced sepsis in a mouse model via lipopolysaccharide (LPS) injection and cecal ligation and puncture (CLP). Sulodexide was administrated to prevent endothelial glycocalyx damage. Endothelial barrier function and expression of endothelial-related proteins were determined using permeability, western blot and immunofluorescent staining. The survival rate, histopathology evaluation of lungs and wet-to-dry lung weight ratio were also evaluated. Results We found that circulating SDC1 levels were persistently upregulated in the non-alive group on days 1 and 3 and were positively correlated with IL-6 levels. Receiver operating characteristic curve analysis showed that SDC1 could distinguish patients with mortality. We showed that SDC1-shedding caused endothelial permeability in the presence of heparinase III and sepsis conditions. Mechanistically, sulodexide (30 LSU/mL) administration markedly inhibited SDC1 shedding and prevented endothelial permeability with zonula occludens-1 (ZO-1) upregulation via NF-κB/ZO-1 pathway. In mice with LPS and CLP-induced sepsis, sulodexide (40 mg/kg) administration decreased the plasma levels of SDC1 and increased survival rate. Additionally, sulodexide alleviated lung injury and restored endothelial glycocalyx damage. Conlusions In conclusion, our data suggest that SDC1 predicts prognosis in children with septic shock and sulodexide may have therapeutic potential for the treatment of sepsis-associated endothelial dysfunction.
Collapse
Affiliation(s)
- Jiayun Ying
- Department of Critical Care Medicine, Children’s Hospital of Fudan University, Shanghai, China
| | - Caiyan Zhang
- Department of Critical Care Medicine, Children’s Hospital of Fudan University, Shanghai, China
| | - Yaodong Wang
- Department of Critical Care Medicine, Children’s Hospital of Fudan University, Shanghai, China
| | - Tingyan Liu
- Department of Critical Care Medicine, Children’s Hospital of Fudan University, Shanghai, China
| | - Zhenhao Yu
- Department of Critical Care Medicine, Children’s Hospital of Fudan University, Shanghai, China
| | - Kexin Wang
- Institute of Pediatrics, Children’s Hospital of Fudan University, National Children’s Medical Center, and the Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Weiming Chen
- Department of Critical Care Medicine, Children’s Hospital of Fudan University, Shanghai, China
| | - Yufeng Zhou
- Institute of Pediatrics, Children’s Hospital of Fudan University, National Children’s Medical Center, and the Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- National Health Commission (NHC) Key Laboratory of Neonatal Diseases, Fudan University, Shanghai, China
- State-level Reginal Children’s Medical Center, Children’s Hospital Of Fudan University at Xiamen (Xiamen Children’s Hospital), Fujian Provincial Key Laboratory of Neonatal Diseases, Fujian, China
| | - Guoping Lu
- Department of Critical Care Medicine, Children’s Hospital of Fudan University, Shanghai, China
| |
Collapse
|
45
|
Wu F, Dorman B, Zeineddin A, Kozar RA. Fibrinogen Inhibits Metalloproteinase-9 Activation and Syndecan-1 Cleavage to Protect Lung Function in ApoE Null Mice After Hemorrhagic Shock. J Surg Res 2023; 288:208-214. [PMID: 37023568 PMCID: PMC10192037 DOI: 10.1016/j.jss.2023.02.043] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 12/05/2022] [Accepted: 02/18/2023] [Indexed: 04/08/2023]
Abstract
INTRODUCTION Obesity is associated with higher mortality following trauma, although the pathogenesis is unclear. Both obesity and trauma are associated with syndecan-1 shedding and metalloproteinase-9 (MMP-9) activation, which can adversely affect endothelial cell function. We recently demonstrated that fibrinogen stabilizes endothelial cell surface syndecan-1 to reduce shedding and maintain endothelial barrier integrity. We thus hypothesized that MMP-9 activation and syndecan-1 shedding would be exacerbated by obesity after trauma but attenuated by fibrinogen-based resuscitation. MATERIALS AND METHODS ApoE null (-/-) mice were fed a Western diet to induce obesity. Mice were subjected to hemorrhage shock and laparotomy then resuscitated with Lactated Ranger's (LR) or LR containing fibrinogen and compared to null and lean sham wild type mice. Mean arterial pressure (MAP) was monitored. Bronchial alveolar lavage protein as an indicator of permeability and lung histopathologic injury were assessed. Syndecan-1 protein and active MMP-9 protein were measured. RESULTS MAP was similar between lean sham and ApoE-/- sham mice. However, following hemorrhage, ApoE-/- mice resuscitated with fibrinogen had significantly higher MAP than LR mice. Lung histopathologic injury and permeability were increased in LR compared to fibrinogen resuscitated animals. Compared with lean sham mice, both active MMP-9 and cleaved syndecan-1 level were significantly higher in ApoE-/- sham mice. Resuscitation with fibrinogen but not lactated Ringers largely reduced these changes. CONCLUSIONS Fibrinogen as a resuscitative adjunct in ApoE-/- mice after hemorrhage shock augmented MAP and reduced histopathologic injury and lung permeability, suggesting fibrinogen protects the endothelium by inhibiting MMP-9-mediated syndecan-1 cleavage in obese mice.
Collapse
Affiliation(s)
- Feng Wu
- Shock Trauma Center, University of Maryland School of Medicine, Baltimore, Maryland
| | - Brooke Dorman
- Shock Trauma Center, University of Maryland School of Medicine, Baltimore, Maryland
| | - Ahmad Zeineddin
- Shock Trauma Center, University of Maryland School of Medicine, Baltimore, Maryland
| | - Rosemary Ann Kozar
- Shock Trauma Center, University of Maryland School of Medicine, Baltimore, Maryland.
| |
Collapse
|
46
|
Matsumoto H, Annen S, Mukai N, Ohshita M, Murata S, Harima Y, Ogawa S, Okita M, Nakabayashi Y, Kikuchi S, Takeba J, Sato N. Circulating Syndecan-1 Levels Are Associated with Chronological Coagulofibrinolytic Responses and the Development of Disseminated Intravascular Coagulation (DIC) after Trauma: A Retrospective Observational Study. J Clin Med 2023; 12:4386. [PMID: 37445421 DOI: 10.3390/jcm12134386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 06/16/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
BACKGROUND The purpose of this study was to evaluate the association between endotheliopathy represented by high levels of circulating syndecan-1 (SDC-1) and coagulofibrinolytic responses due to trauma, which can lead to disseminated intravascular coagulation (DIC). METHODS We retrospectively evaluated 48 eligible trauma patients immediately admitted to our hospital and assessed SDC-1 and coagulofibrinolytic parameters for 7 days after admission. We compared the longitudinal changes of coagulofibrinolytic parameters and SDC-1 levels between two groups (high and low SDC-1) according to median SDC-1 value on admission. RESULTS The median circulating SDC-1 level was 99.6 (61.1-214.3) ng/mL on admission, and levels remained high until 7 days after admission. Coagulofibrinolytic responses assessed by biomarkers immediately after trauma were correlated with SDC-1 elevation (thrombin-antithrombin complex, TAT: r = 0.352, p = 0.001; antithrombin, AT: r = -0.301, p < 0.001; plasmin-α2-plasmin inhibitor complex, PIC: r = 0.503, p = 0.035; tissue plasminogen activator, tPA: r = 0.630, p < 0.001). Sustained SDC-1 elevation was associated with intense and prolonged coagulation activation, impairment of anticoagulation, and fibrinolytic activation followed by inhibition of fibrinolysis, which are the primary responses associated with development of DIC in the acute phase of trauma. Elevation of circulating SDC-1 level was also associated with consumption coagulopathy and the need for transfusion, which revealed a significant association between high SDC-1 levels and the development of DIC after trauma (area under the curve, AUC = 0.845, cut-off value = 130.38 ng/mL, p = 0.001). CONCLUSIONS High circulating levels of syndecan-1 were associated with intense and prolonged coagulation activation, impairment of anticoagulation, fibrinolytic activation, and consumption coagulopathy after trauma. Endotheliopathy represented by SDC-1 elevation was associated with trauma induced coagulopathy, which can lead to the development of DIC.
Collapse
Affiliation(s)
- Hironori Matsumoto
- Department of Emergency and Critical Care Medicine, Graduate School of Medicine, Ehime University, Toon 791-0295, Ehime, Japan
| | - Suguru Annen
- Department of Emergency and Critical Care Medicine, Graduate School of Medicine, Ehime University, Toon 791-0295, Ehime, Japan
| | - Naoki Mukai
- Department of Emergency and Critical Care Medicine, Graduate School of Medicine, Ehime University, Toon 791-0295, Ehime, Japan
| | - Muneaki Ohshita
- Department of Emergency and Critical Care Medicine, Graduate School of Medicine, Ehime University, Toon 791-0295, Ehime, Japan
| | - Satoru Murata
- Department of Emergency and Critical Care Medicine, Graduate School of Medicine, Ehime University, Toon 791-0295, Ehime, Japan
| | - Yutaka Harima
- Department of Emergency and Critical Care Medicine, Graduate School of Medicine, Ehime University, Toon 791-0295, Ehime, Japan
| | - Shirou Ogawa
- Department of Emergency and Critical Care Medicine, Graduate School of Medicine, Ehime University, Toon 791-0295, Ehime, Japan
| | - Mitsuo Okita
- Department of Emergency and Critical Care Medicine, Graduate School of Medicine, Ehime University, Toon 791-0295, Ehime, Japan
| | - Yuki Nakabayashi
- Department of Emergency and Critical Care Medicine, Graduate School of Medicine, Ehime University, Toon 791-0295, Ehime, Japan
| | - Satoshi Kikuchi
- Department of Emergency and Critical Care Medicine, Graduate School of Medicine, Ehime University, Toon 791-0295, Ehime, Japan
| | - Jun Takeba
- Department of Emergency and Critical Care Medicine, Graduate School of Medicine, Ehime University, Toon 791-0295, Ehime, Japan
| | - Norio Sato
- Department of Emergency and Critical Care Medicine, Graduate School of Medicine, Ehime University, Toon 791-0295, Ehime, Japan
| |
Collapse
|
47
|
Sembajwe LF, Ssekandi AM, Namaganda A, Muwonge H, Kasolo JN, Kalyesubula R, Nakimuli A, Naome M, Patel KP, Masenga SK, Kirabo A. Glycocalyx-Sodium Interaction in Vascular Endothelium. Nutrients 2023; 15:2873. [PMID: 37447199 PMCID: PMC10343370 DOI: 10.3390/nu15132873] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 06/19/2023] [Accepted: 06/22/2023] [Indexed: 07/15/2023] Open
Abstract
The glycocalyx generally covers almost all cellular surfaces, where it participates in mediating cell-surface interactions with the extracellular matrix as well as with intracellular signaling molecules. The endothelial glycocalyx that covers the luminal surface mediates the interactions of endothelial cells with materials flowing in the circulating blood, including blood cells. Cardiovascular diseases (CVD) remain a major cause of morbidity and mortality around the world. The cardiovascular risk factors start by causing endothelial cell dysfunction associated with destruction or irregular maintenance of the glycocalyx, which may culminate into a full-blown cardiovascular disease. The endothelial glycocalyx plays a crucial role in shielding the cell from excessive exposure and absorption of excessive salt, which can potentially cause damage to the endothelial cells and underlying tissues of the blood vessels. So, in this mini review/commentary, we delineate and provide a concise summary of the various components of the glycocalyx, their interaction with salt, and subsequent involvement in the cardiovascular disease process. We also highlight the major components of the glycocalyx that could be used as disease biomarkers or as drug targets in the management of cardiovascular diseases.
Collapse
Affiliation(s)
- Lawrence Fred Sembajwe
- Department of Medical Physiology, Makerere University College of Health Sciences, Kampala P.O. Box 7072, Uganda; (A.M.S.); (A.N.); (H.M.); (J.N.K.); (R.K.)
| | - Abdul M. Ssekandi
- Department of Medical Physiology, Makerere University College of Health Sciences, Kampala P.O. Box 7072, Uganda; (A.M.S.); (A.N.); (H.M.); (J.N.K.); (R.K.)
| | - Agnes Namaganda
- Department of Medical Physiology, Makerere University College of Health Sciences, Kampala P.O. Box 7072, Uganda; (A.M.S.); (A.N.); (H.M.); (J.N.K.); (R.K.)
| | - Haruna Muwonge
- Department of Medical Physiology, Makerere University College of Health Sciences, Kampala P.O. Box 7072, Uganda; (A.M.S.); (A.N.); (H.M.); (J.N.K.); (R.K.)
| | - Josephine N. Kasolo
- Department of Medical Physiology, Makerere University College of Health Sciences, Kampala P.O. Box 7072, Uganda; (A.M.S.); (A.N.); (H.M.); (J.N.K.); (R.K.)
| | - Robert Kalyesubula
- Department of Medical Physiology, Makerere University College of Health Sciences, Kampala P.O. Box 7072, Uganda; (A.M.S.); (A.N.); (H.M.); (J.N.K.); (R.K.)
| | - Annettee Nakimuli
- Department of Obstetrics and Gynecology, School of Medicine, Makerere University College of Health Sciences, Kampala P.O. Box 7072, Uganda;
| | - Mwesigwa Naome
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| | - Kaushik P. Patel
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198, USA;
| | - Sepiso K. Masenga
- Department of Physiological Sciences, School of Medicine and Health Sciences, Mulungushi University, Kabwe P.O. Box 80415, Zambia;
| | - Annet Kirabo
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| |
Collapse
|
48
|
J D Moreira N, Dos Santos F, Li JB, Aletti F, Irigoyen MCC, Kistler EB. Enteral administration of the protease inhibitor gabexate mesilate preserves vascular function in experimental trauma/hemorrhagic shock. Sci Rep 2023; 13:10148. [PMID: 37349360 PMCID: PMC10287748 DOI: 10.1038/s41598-023-36021-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 05/27/2023] [Indexed: 06/24/2023] Open
Abstract
Preserving vascular function is crucial for preventing multiorgan failure and death in ischemic and low-pressure states such as trauma/hemorrhagic shock (T/HS). It has recently been reported that inhibiting circulating proteases released from the bowel to the circulation during T/HS may preserve vascular function and improve outcomes following T/HS. This study aimed to evaluate the role of the serine protease inhibitor gabexate mesilate (GM) in preserving vascular function during T/HS when given enterally. We studied the vascular reactivity of mesenteric arteries from male Wistar rats treated with enteral GM (10 mg/kg) (GM-treated, n = 6) or control (Shock-control, n = 6) following (T/HS) using pressure myography. Concentration-response curves of endothelial-dependent and endothelial-independent agonists (e.g., acetylcholine, sodium nitroprusside) ranging from 10-10 to 10-5 M were performed. In a second set of experiments, ex-vivo arteries from healthy rats were perfused with plasma from shocked animals from both groups and vascular performance was similarly measured. Arteries from the GM-treated group demonstrated a preserved concentration-response curve to the α1 adrenergic agonist phenylephrine compared to arteries from Shock-control animals (- logEC50: - 5.73 ± 0.25 vs. - 6.48 ± 0.2, Shock-control vs. GM-treated, p = 0.04). When perfused with plasma from GM-treated rats, healthy arteries exhibited an even greater constriction and sensitivity to phenylephrine (- logEC50: - 6.62 ± 0.21 vs. - 7.13 ± 0.21, Shock-control vs. GM-treated, p = 0.02). Enteral GM also preserved the endothelium-dependent vascular response to agonists following T/HS and limited syndecan-1 shedding as a marker of glycocalyx compromise (41.84 ± 9 vs. 17.63 ± 3.97 ng/mL, Shock-control vs. GM-treated, p = 0.02). Syndecan-1 cleavage was correlated with plasma trypsin-like activity (r2 = 0.9611). Enteral gabexate mesilate was able to maintain vascular function in experimental T/HS, which was reflected by improved hemodynamics (mean arterial pressure 50.39 ± 7.91 vs. 64.95 ± 3.43 mmHg, Shock-control vs. GM treated, p = 0.0001). Enteral serine protease inhibition may be a potential therapeutic intervention in the treatment of T/HS.
Collapse
Affiliation(s)
- Nathalia J D Moreira
- Instituto do Coração, Hospital das ClínicasFaculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil.
| | - Fernando Dos Santos
- Department of Anesthesiology and Critical Care, University of California, San Diego, La Jolla, CA, USA
| | - Joyce B Li
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - Federico Aletti
- Universidade Federal de São Paulo, São José dos Campos, Brazil
| | - Maria Claudia C Irigoyen
- Instituto do Coração, Hospital das ClínicasFaculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Erik B Kistler
- Department of Anesthesiology and Critical Care, University of California, San Diego, La Jolla, CA, USA
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
| |
Collapse
|
49
|
Abdullah S, Ghio M, Cotton-Betteridge A, Vinjamuri A, Drury R, Packer J, Aras O, Friedman J, Karim M, Engelhardt D, Kosowski E, Duong K, Shaheen F, McGrew PR, Harris CT, Reily R, Sammarco M, Chandra PK, Pociask D, Kolls J, Katakam PV, Smith A, Taghavi S, Duchesne J, Jackson-Weaver O. Succinate metabolism and membrane reorganization drives the endotheliopathy and coagulopathy of traumatic hemorrhage. SCIENCE ADVANCES 2023; 9:eadf6600. [PMID: 37315138 PMCID: PMC10266735 DOI: 10.1126/sciadv.adf6600] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 05/10/2023] [Indexed: 06/16/2023]
Abstract
Acute hemorrhage commonly leads to coagulopathy and organ dysfunction or failure. Recent evidence suggests that damage to the endothelial glycocalyx contributes to these adverse outcomes. The physiological events mediating acute glycocalyx shedding are undefined, however. Here, we show that succinate accumulation within endothelial cells drives glycocalyx degradation through a membrane reorganization-mediated mechanism. We investigated this mechanism in a cultured endothelial cell hypoxia-reoxygenation model, in a rat model of hemorrhage, and in trauma patient plasma samples. We found that succinate metabolism by succinate dehydrogenase mediates glycocalyx damage through lipid oxidation and phospholipase A2-mediated membrane reorganization, promoting the interaction of matrix metalloproteinase 24 (MMP24) and MMP25 with glycocalyx constituents. In a rat hemorrhage model, inhibiting succinate metabolism or membrane reorganization prevented glycocalyx damage and coagulopathy. In patients with trauma, succinate levels were associated with glycocalyx damage and the development of coagulopathy, and the interaction of MMP24 and syndecan-1 was elevated compared to healthy controls.
Collapse
Affiliation(s)
- Sarah Abdullah
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA, USA
| | - Michael Ghio
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA, USA
| | | | | | - Robert Drury
- Tulane University School of Medicine, New Orleans, LA, USA
| | - Jacob Packer
- Tulane University School of Medicine, New Orleans, LA, USA
| | - Oguz Aras
- Tulane University School of Medicine, New Orleans, LA, USA
| | - Jessica Friedman
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA, USA
| | - Mardeen Karim
- Tulane University School of Medicine, New Orleans, LA, USA
| | | | | | - Kelby Duong
- Tulane University School of Medicine, New Orleans, LA, USA
| | - Farhana Shaheen
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA, USA
| | - Patrick R. McGrew
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA, USA
- University Medical Center, New Orleans, LA, USA
| | - Charles T. Harris
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA, USA
- University Medical Center, New Orleans, LA, USA
| | - Robert Reily
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA, USA
- University Medical Center, New Orleans, LA, USA
| | - Mimi Sammarco
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA, USA
| | - Partha K. Chandra
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Derek Pociask
- Tulane University School of Medicine, Center for Translational Research in Infection and Inflammation, New Orleans, LA, USA
| | - Jay Kolls
- Tulane University School of Medicine, Center for Translational Research in Infection and Inflammation, New Orleans, LA, USA
| | - Prasad V. Katakam
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Alison Smith
- Louisiana State University Health Sciences Center, New Orleans, LA, USA
- University Medical Center, New Orleans, LA, USA
| | - Sharven Taghavi
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA, USA
- University Medical Center, New Orleans, LA, USA
| | - Juan Duchesne
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA, USA
- University Medical Center, New Orleans, LA, USA
| | - Olan Jackson-Weaver
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA, USA
| |
Collapse
|
50
|
Gregersen JS, Bazancir LA, Johansson PI, Sørensen H, Achiam MP, Olsen AA. Major open abdominal surgery is associated with increased levels of endothelial damage and interleukin-6. Microvasc Res 2023; 148:104543. [PMID: 37156371 DOI: 10.1016/j.mvr.2023.104543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 04/19/2023] [Accepted: 04/25/2023] [Indexed: 05/10/2023]
Abstract
OBJECTIVE To examine changes in biomarkers of endothelial glycocalyx shedding, endothelial damage, and surgical stress following major open abdominal surgery and the correlation to postoperative morbidity. INTRODUCTION Major abdominal surgery is associated with high levels of postoperative morbidity. Two possible reasons are the surgical stress response and the impairment of the glycocalyx and endothelial cells. Further, the degree of these responses may correlate with postoperative morbidity and complications. METHODS A secondary data analysis of prospectively collected data from two cohorts of patients undergoing open liver surgery, gastrectomy, esophagectomy, or Whipple procedure (n = 112). Hemodynamics and blood samples were collected at predefined timestamps and analyzed for biomarkers of glycocalyx shedding (Syndecan-1), endothelial activation (sVEGFR1), endothelial damage (sThrombomodulin (sTM)), and surgical stress (IL6). RESULTS Major abdominal surgery led to increased levels of IL6 (0 to 85 pg/mL), Syndecan-1 (17.2 to 46.4 ng/mL), and sVEGFR1 (382.8 to 526.5 pg/mL), peaking at the end of the surgery. In contrast, sTM, did not increase during surgery, but increased significantly following surgery (5.9 to 6.9 ng/mL), peaking at 18 h following the end of surgery. Patients characterized with high postoperative morbidity had higher levels of IL6 (132 vs. 78 pg/mL, p = 0.007) and sVEGFR1 (563.1 vs. 509.4 pg/mL, p = 0.045) at the end of the surgery, and of sTM (8.2 vs. 6.4 ng/mL, p = 0.038) 18 h following surgery. CONCLUSION Major abdominal surgery leads to significantly increased levels of biomarkers of endothelial glycocalyx shedding, endothelial damage, and surgical stress, with the highest levels seen in patients developing high postoperative morbidity.
Collapse
Affiliation(s)
| | - Laser Arif Bazancir
- Department of Surgery and Transplantation, Rigshospitalet, Copenhagen University Hospital, Denmark
| | - Pär Ingemar Johansson
- Department of Clinical Immunology, Rigshospitalet, Copenhagen University Hospital, Denmark
| | - Henrik Sørensen
- Department of Anesthesiology, Rigshospitalet, Copenhagen University Hospital, Denmark
| | - Michael Patrick Achiam
- Department of Surgery and Transplantation, Rigshospitalet, Copenhagen University Hospital, Denmark
| | - August Adelsten Olsen
- Department of Surgery and Transplantation, Rigshospitalet, Copenhagen University Hospital, Denmark
| |
Collapse
|