Skip to main content

Plasma for prevention and treatment of glycocalyx degradation in trauma and sepsis

Abstract

The endothelial glycocalyx, a gel-like layer that lines the luminal surface of blood vessels, is composed of proteoglycans, glycoproteins, and glycosaminoglycans. The endothelial glycocalyx plays an essential role in vascular homeostasis, and its degradation in trauma and sepsis can lead to microvascular dysfunction and organ injury. While there are no proven therapies for preventing or treating endothelial glycocalyx degradation, some initial literature suggests that plasma may have a therapeutic role in trauma and sepsis patients. Overall, the literature suggesting the use of plasma as a therapy for endothelial glycocalyx degradation is non-clinical basic science or exploratory. Plasma is an established therapy in the resuscitation of patients with hemorrhage for restoration of coagulation factors. However, plasma also contains other bioactive components, including sphingosine-1 phosphate, antithrombin, and adiponectin, which may protect and restore the endothelial glycocalyx, thereby helping to maintain or restore vascular homeostasis. This narrative review begins by describing the endothelial glycocalyx in health and disease: we discuss the overlapping disease mechanisms in trauma and sepsis that lead to its damage and introduce plasma transfusion as a potential therapy for prevention and treatment of endothelial glycocalyx degradation. Second, we review the literature on plasma as an exploratory therapy for endothelial glycocalyx degradation in trauma and sepsis. Third, we discuss the safety of plasma transfusion by reviewing the adverse events associated with plasma and other blood product transfusions, and we examine modern transfusion precautions that have enhanced the safety of plasma transfusion. We conclude that the literature proposes that plasma may have the potential to prevent and treat endothelial glycocalyx degradation in trauma and sepsis, indicating the need for further research.

Introduction

Trauma and sepsis are leading causes of morbidity and mortality [1,2,3]. Traumatic hemorrhage and sepsis share the common pathophysiology of endothelial glycocalyx (hereafter referred to as glycocalyx) degradation and endotheliopathy, for which plasma transfusion is emerging as a possible therapy, and additionally as preventative intervention that spares the glycocalyx from degradation [4,5,6]. Plasma transfusion is commonly used for the replenishment of coagulation factors and as a component of a balanced massive transfusion protocol in the setting of hemorrhage, alongside red blood cells and platelets [7,8,9,10,11,12]. Yet, plasma, the non-cellular component of blood, also contains numerous other biologically-active components that influence homeostatic and pathogenic pathways other than coagulation. These bioactive components, which include sphingosine-1 phosphate, antithrombin, and adiponectin, may prevent and restore damage to the glycocalyx, reduce endothelial cell permeability and leukocyte adhesion, and decrease inflammation in critical illnesses [13,14,15,16,17,18,19,20,21,22]. This narrative review aims to demonstrate the evidence for the hypothesis that plasma is a potential therapeutic, and glycocalyx sparing, agent in trauma and sepsis (beyond treatment of coagulopathy), through its potential protective and restorative effects on the glycocalyx.

The role of the glycocalyx in health

The vascular endothelium forms the intimal surface of blood vessels [23]. The luminal surface of the endothelium is coated with the glycocalyx, an apical matrix composed of membrane bound proteoglycans and their associated glycosaminoglycans (GAGs), glycoproteins, and plasma proteins (Fig. 1) [5, 24]. Proteoglycans are glycosylphosphatidylinositol (GPI) anchored (e.g. glypicans) or transmembrane (e.g. syndecans) proteins that are covalently bound to sulfated glycosaminoglycans (such as heparan sulfate and chondroitin sulfate) which extend into the vascular lumen [25, 26]. By virtue of their negative charge, heparan sulfate and chondroitin sulfate electrostatically interact with plasma proteins [27]. Hyaluronan is not sulfated and sequesters water, which stabilizes the gel like structure, and binds to the glycoprotein CD44 [28]. Playing a crucial role in maintaining vascular health, the glycocalyx helps to regulate various homeostatic functions such as blood vessel tone, leukocyte adhesion to endothelial cells, blood clot formation, and maintenance of the endothelial barrier [5, 24, 29,30,31,32,33,34,35].

Fig. 1
figure 1

A The healthy endothelial glycocalyx. B Degradation of the endothelial glycocalyx caused by sympatho-adrenal hyperactivation pathophysiology seen in trauma and sepsis. This leads to endothelial cell activation, glycocalyx degradation, and vascular leakage. MMPs (matrix metalloproteinase) and heparanase are shown degrading the endothelial glycocalyx. C Potential mechanisms by which plasma transfusion can restore the damaged glycocalyx, including inhibition of MMPs by sphingosine-1 phosphate, the glycocalyx-stabilizing effect of albumin, and the endothelial-protective effects of adiponectin and antithrombin

Glycocalyx degradation in sepsis

Sepsis, the injurious systemic response to infection, often leads to significant endothelial cell activation and shedding of the glycocalyx (Fig. 1). During sepsis, the innate immune response triggers endothelial cells to release adhesion molecules, chemoattractants, vasoactive compounds, and inflammatory cytokines, occurring in parallel with sympathoadrenal hyperactivation [4, 36]. This leads to degradation of the endothelial glycocalyx through endothelial cell activation and the action of proteases, such as matrix metalloproteinases (MMPs), and glycosaminoglycan-degrading enzymes, such as heparanase [37,38,39,40,41]. This shedding contributes to the local microcirculatory dysfunction characteristic of sepsis, marked by vascular leak, tissue swelling, hypotension, and reduced oxygen supply to tissues, which leads to organ injury [36, 42].

The overlapping pathophysiology of sepsis and trauma

Similar to sepsis, traumatic hemorrhage also involves a systemic disruptive inflammatory response that damages the glycocalyx, leading to organ dysfunction and worsening disease, which has been termed the Endotheliopathy of Trauma (EoT) [4, 6, 43, 44]. Both sepsis and trauma are syndromes characterized by sympatho-adrenal hyperactivation, leading to endothelial cell activation and glycocalyx degradation (endotheliopathy). This unifying pathophysiology of critical illness, termed Shock Induced Endotheliopathy (SHINE) by Johansson et al., is proposed to be conserved across acute critical illnesses and evolutionarily adapted [4]. SHINE is associated within glycocalyx shedding, capillary leakage, microvascular thrombosis, and organ failure [4]. Interestingly, trauma patients with endothelial glycocalyx degradation, as quantified by elevated circulating syndecan-1 levels after injury, have a higher likelihood of developing sepsis compared to those with normal syndecan-1 levels, suggesting convergent pathogenic mechanisms in these two critical illnesses characterized by endotheliopathy [45].

Plasma transfusion as a potential therapy for endothelial glycocalyx degradation

The majority of the evidence in support of the hypothesis that plasma transfusion is a potential therapeutic agent for the treatment of glycocalyx degradation is derived primarily from studies of trauma, particularly in the context of hemorrhagic shock, where plasma is now a standard therapeutic intervention during massive transfusion [44]. In contrast, there is a relative scarcity of studies exploring plasma’s impact in sepsis. Given the shared pathophysiology of shock-induced sympatho-adrenal hyperactivation and subsequent endothelial cell activation and glycocalyx degradation across these critical illnesses, there are likely overlapping therapeutic mechanisms between the use of plasma in hemorrhagic shock and its application in other critical conditions [4]. This common pathway suggests that the benefits of plasma on the glycocalyx observed in trauma care could inform broader applications of plasma as a therapy in other critical illnesses characterized by endotheliopathy. Indeed, it is suggested that plasma is potentially beneficial in the treatment of non-hemorrhagic disease states with endotheliopathy [6]. This review will focus on studies of plasma’s impact on the glycocalyx in trauma and sepsis. Since the endotheliopathy of sepsis and trauma share similar pathophysiology, an intriguing hypothesis is that the potential benefits of plasma on the glycocalyx in trauma will translate to sepsis.

The biological mechanism of plasma transfusion’s impact on the glycocalyx

Plasma contains numerous bioactive components, several of which have proposed interaction with the glycocalyx (Fig. 1). Albumin, while overall an anionic constituent of plasma, has positively charged arginine residues that are hypothesized to interact with the negatively charged glycosaminoglycans of the glycocalyx, leading to a pleiotropic stabilizing effect on the glycocalyx [46]. Glycocalyx stabilization by albumin and other plasma proteins such as the HDL-associated apolipoprotein M is additionally mediated by endothelial delivery of sphingosine-1-phosphate (S1P), a vascular-protective lipid shown to attenuate endothelial glycocalyx shedding by inhibiting MMPs [15, 41, 47,48,49]. The actual impact of albumin in plasma on glycocalyx degradation remains questionable due to the varying concentrations of albumin in fresh frozen plasma (FFP), which are slightly below physiologic levels. However, resuscitation with plasma would be much less dilutional to albumin levels than crystalloid resuscitation. Plasma serine proteases such as antithrombin and plasma adiponectin additionally may impart protective effects on glycocalyx integrity, but further research is needed to elucidate their mechanism of action [17, 50, 51].

Plasma preparations

Plasma is available in various preparations, including FFP, thawed plasma, and dried plasma. FFP is frozen within eight hours of collection and thawed immediately prior to usage, and contains all clotting factors and proteins found in blood. Thawed plasma is previously frozen plasma that is stored at 1 to 6 °C for up to 5 days, but with a measurable decrease in FV and FVIII activity level [52,53,54]. Dried plasma includes lyophilized plasma and spray dried plasma, which have logistical advantages of not requiring freezing or refrigeration for storage [55, 56]. Unless specifically stated, throughout this review the term ‘plasma’ broadly refers to any of these preparations. Further studies are needed to determine whether different plasma preparations have similar levels of sphingosine-1-phosphate and other components that affect the glycocalyx, and if these variations impact clinical outcomes.

Plasma transfusion as a potential therapy for glycocalyx degradation in trauma: pre-clinical literature

The potential utility of plasma as a glycocalyx-protective and restorative agent for treatment of endotheliopathy is supported by trauma models of hemorrhagic shock (Supplemental Table 1). Peng et al. used a mouse model of hemorrhagic shock which was resuscitated with either FFP or Lactated Ringers (LR). Endothelial permeability was significantly lower in mice resuscitated with FFP compared to those treated with LR. Furthermore, FFP resuscitation led to a reduction in inflammation as evidenced by markedly lower levels of myeloperoxidase immunofluorescence (an indicator of neutrophil infiltration) in the FFP-treated group compared to the LR group [20]. Pati et al. used an in vitro model of human pulmonary endothelial cells (PEC) treated in a hypoxic incubator to represent hypoxia from hemorrhagic shock and then treated with FFP or LR. FFP was found to be more effective at inhibiting PEC permeability compared to LR [57]. Kozar et al. conducted a study using a rat model of hemorrhagic shock, where the rats were resuscitated with either FFP or LR. The study found that resuscitation with FFP led to early signs of glycocalyx restoration, observable under electron microscopy, starting from around two hours post-resuscitation. This was in contrast to the LR group, where no glycocalyx restoration was noted. Additionally, the thickness of the glycocalyx post-treatment was significantly greater in the FFP group compared to the LR group. Furthermore, the study assessed the expression of syndecan-1 (a proteoglycan) mRNA in the lungs. Syndecan-1 mRNA expression in the lungs was significantly lower in rats exposed to hemorrhagic shock than control rats. Resuscitation with LR further decreased this mRNA expression. However, resuscitation with FFP restored syndecan-1 mRNA expression to levels comparable to the control group, suggesting activation of mechanisms driving glycocalyx reconstitution [19]. Additional preclinical studies demonstrate that resuscitation with FFP in hemorrhagic shock models reduces glycocalyx shedding, endothelial permeability, white blood cell adhesion, and endothelial cell apoptosis, and also improves vital signs and laboratory parameters such as blood pH, lactic acid, base excess, and respiratory rate [13, 18, 22, 58,59,60]. Overall these preclinical studies suggest benefits of FFP in trauma, independent of replenishment of coagulation factors.

Plasma transfusion as a potential therapy for glycocalyx degradation in trauma: clinical literature

Expanding upon these basic science investigations, clinical studies have examined the administration of plasma to trauma patients (Supplemental Table 2). The literature supports plasma’s use for improving clinical outcomes in trauma [61,62,63,64,65]. However, there is a paucity of research examining the link between plasma’s impact on the glycocalyx to observed clinical outcomes. Therefore, there is insufficient data at this time to determine whether the link between the administration of plasma with clinical benefits is mediated through the glycocalyx.

One study that highlights the potential benefit of plasma on the endothelial glycocalyx in a clinical setting is a secondary analysis of the Prehospital Air Medical Plasma (PAMPer) trial [12, 66]. PAMPer showed a significant decrease in 30-day mortality for trauma patients at risk of hemorrhagic shock who were randomized to receive plasma compared to those who did not (23.2% vs. 33.0%, p < 0.03). In the secondary analysis of PAMPer, Gruen et al. assayed endothelial, glycocalyx and inflammatory markers from 405 trauma patients enrolled in this trial. Plasma administration was associated with a decrease in endothelial and glycocalyx biomarkers (syndecan-1, thrombomodulin, and vascular endothelial growth factor) and decreased pro-inflammatory mediators (IL-6, TNF-α, and MCP-1) in a subgroup of patients with greater injury severity scores and a higher incidence of blunt trauma. Further research is needed to examine if any part of the clinical benefits of plasma in trauma are mediated through the glycocalyx.

Glycocalyx degradation in sepsis

Pre-clinical and clinical studies have established a key role for endothelial glycocalyx dysfunction in the pathophysiology of sepsis (Supplemental Tables 3 and 4). Additionally, increased levels of endothelial glycocalyx degradation products in blood samples correlate with the severity and mortality of sepsis [67,68,69,70]. There is also a link between sepsis-induced endothelial glycocalyx dysfunction and organ dysfunction, such as acute lung injury, acute respiratory distress syndrome, intestinal dysfunction, and acute kidney injury [37, 38, 71,72,73]. Considering the connections between endothelial glycocalyx dysfunction and increased morbidity, mortality, and organ dysfunction in sepsis, strategically targeting the endothelial glycocalyx is emerging as a potential therapeutic strategy in sepsis management.

Plasma transfusion as a potential therapy for glycocalyx degradation in sepsis: pre-clinical literature

Based on the initial data in support of the hypothesis that plasma resuscitation improves outcomes in traumatic hemorrhagic shock in part through the attenuation of endothelial injury, which is pathophysiologically similar to the endothelial injury seen in sepsis, several basic science studies have examined the impact of plasma on endothelial glycocalyx degradation in sepsis (Supplemental Table 3). Chang et al. conducted a study comparing rat FFP versus normal saline resuscitation of a rat cecal ligation and puncture model of polymicrobial sepsis. Resuscitation with FFP in this study was associated with increased survival at 48 h [74]. FFP resuscitation, as opposed to normal saline resuscitation, was linked to a reduced increase in post-resuscitation blood levels of epinephrine, norepinephrine, IL-6, and syndecan-1. Furthermore, FFP resuscitation compared to normal saline resuscitation had significantly improved PO2/FiO2 ratio, and decreased lung wet-to-dry weight ratio, which suggests less pulmonary edema in the plasma resuscitation group [74]. Conversely, Barry et al. used a mouse model of sepsis and randomized mice to receive either lyophilized plasma or LR, and found no significant difference in mortality or post resuscitation plasma biomarkers of inflammation between treatment groups. However, lyophilized plasma resuscitation did induce a decrease in pulmonary inflammatory gene expression. While the work of Barry et al. seemingly contradicts that of Chang et al., several limitations in Barry et al.’s study need consideration, including the use of human lyophilized plasma in a mouse model, the use of lyophilized plasma instead FFP, and an additional freeze–thaw cycle of the lyophilized plasma; further research is therefore needed.

Sheck et al. studied the impact of FFP on the endothelial glycocalyx by measuring syndecan-1 levels of patients post-operative from major abdominal surgery who received plasma compared to those who did not receive FFP. Circulating syndecan-1 levels were lower in patients who underwent major abdominal surgery and were transfused with FFP compared to those who were not transfused. The authors complemented this observational human study by performing mechanistic studies using human pulmonary microvascular endothelial cells. In this model, FFP treatment attenuated LPS-induced pulmonary endothelial permeability in vitro, as assessed using a FITC-dextran transwell assay [75]. In addition, adding FFP to cells exposed to LPS was associated with a significant attenuation in endothelial activation, as demonstrated by a decrease in endothelial gene expression of ICAM-1, VCAM-1, and ANG-2 [75]. Unexpectedly, FFP treatment increased LPS-induced leukocyte adhesion to endothelial cells in vitro. Taken together, this study showed that FFP decreased biomarkers of endothelial glycocalyx degradation in post-operative patients, and showed that in an in vitro model of endothelial cells FFP exposure after LPS exposure improved endothelial cell barrier function [76].

Plasma transfusion as a potential therapy for glycocalyx degradation in sepsis: clinical literature (or lack thereof)

The literature evaluating the clinical use of plasma as a therapy and restorative agent for glycocalyx degradation in human sepsis patients is quite limited (Supplemental Table 4), precluding any conclusion to be drawn. In a prospective substudy of a randomized clinical trial of 33 critically ill, non-bleeding coagulopathic patients, Straat et al. measured biomarkers of endothelial function and inflammatory cytokines before and after elective FFP transfusion prior to an invasive procedure [77]. About half of the patients in this cohort had sepsis (n = 15, 45%), and the median SOFA score was 11 (IQR 10–14). FFP administration led to a significant decrease in circulating TNF-ɑ (11.3 pg/ml vs 2.3 pg/ml, p < 0.01) and syndecan-1 levels (675 pg/ml vs 565 pg/ml, p < 0.01) [77]. These findings are hypothesis-generating that FFP administration decreases inflammation and glycocalyx shedding in non-bleeding, coagulopathic, critically ill patients. Studies examining the impact of plasma on clinical outcomes in sepsis show mixed results, are limited, and are inconclusive [78, 79].

Safety of plasma transfusion

Adverse events reported with plasma transfusions include allergic reactions, transfusion associated lung injury (TRALI), transfusion associated circulatory overload (TACO), and infection [80, 81]. However, hemovigilance methods have made transfusions safer. The incidence of TRALI has decreased since the implementation of modern plasma collection techniques such as preferential use of male donor plasma and leukocyte antibody screening [82,83,84]. Pathogen inactivation methods such as solvent and detergent treatment and amotosalen plus UVA light have proven to be safe for inactivating numerous infectious pathogens from blood products [85, 86].

Plasma contains coagulation factors, and there is a risk that plasma transfusion could lead to thromboembolism. A retrospective study of 381 trauma patients receiving FFP and packed red blood cells (PRBCs) showed that each unit of FFP transfused increased the risk of venous thromboembolism in patients who received fewer than 4 units of PRBCs [87]. The Pragmatic Randomized Optimal Platelet and Plasma Ratios (PROPPR) trial transfused trauma patients with either a 1:1:1 or 1:1:2 ratio of plasma, platelets, and red blood cells. In this trial of 680 patients, those in the 1:1:1 group received more plasma than those in the 1:1:2 group (5 units vs. 7 units, p < 0.01). However, there was no increase in venous thromboembolism or acute respiratory distress syndrome between the groups [64]. Further research is needed to better understand the exact risk that plasma transfusion has on thromboembolism, and any patients enrolled in studies of plasma should be monitored for thromboembolism.

During the COVID-19 pandemic, convalescent plasma, which is FFP with specific antibodies from those convalescing from disease, was extensively studied as a potential therapy. A Cochrane systematic review and meta-analysis, which included 33 randomized controlled trials, indicates that plasma has little to no impact on the risk of serious adverse events with moderate certainty [88]. This Cochrane review also showed no mortality benefit from plasma. However, due to differences in the pathophysiologies between COVID-19 and bacterial sepsis, further research of plasma in the context of bacterial sepsis is warranted.

Conclusion

In conclusion, this narrative review demonstrates that there is initial evidence supporting the hypothesis that plasma reduces glycocalyx degradation in pre-clinical studies and biomarkers in clinical studies of trauma and sepsis. Furthermore, this review concludes that, at this time, there is a lack of evidence to prove whether plasma’s impact on clinical outcomes is mediated through the glycocalyx. However, this remains an intriguing direction for future research. Although the majority of data in support of this hypothesis originates from trauma-related studies, the shared pathophysiology of sepsis and trauma suggests that plasma’s beneficial effects on the glycocalyx in trauma might extend to sepsis. Further research, especially in the context of sepsis, is needed to fully understand the therapeutic potential and risks of plasma before it is used clinically for the treatment of glycocalyx degradation. It is also crucial to further study S1P, albumin, antithrombin, and adiponectin, to further elucidate the biologic mechanisms through which these components impact the glycocalyx, and to explore methods for enriching these components.

References

  1. World Health Organization. Injuries and violence. Accessed December 9, 2023. https://www.who.int/news-room/fact-sheets/detail/injuries-and-violence

  2. Fleischmann C, Scherag A, Adhikari NKJ, et al. Assessment of global incidence and mortality of hospital-treated sepsis current estimates and limitations. Am J Respir Crit Care Med. 2016;193(3):259–72. https://doi.org/10.1164/rccm.201504-0781OC.

    Article  CAS  PubMed  Google Scholar 

  3. Vincent JL, Marshall JC, Namendys-Silva SA, et al. Assessment of the worldwide burden of critical illness: the intensive care over nations (ICON) audit. Lancet Respir Med. 2014;2(5):380–6. https://doi.org/10.1016/S2213-2600(14)70061-X.

    Article  PubMed  Google Scholar 

  4. Johansson PI, Stensballe J, Ostrowski SR. Shock induced endotheliopathy (SHINE) in acute critical illness—a unifying pathophysiologic mechanism. Crit Care Lond Engl. 2017;21(1):25. https://doi.org/10.1186/s13054-017-1605-5.

    Article  Google Scholar 

  5. Uchimido R, Schmidt EP, Shapiro NI. The glycocalyx: a novel diagnostic and therapeutic target in sepsis. Crit Care Lond Engl. 2019;23(1):16. https://doi.org/10.1186/s13054-018-2292-6.

    Article  Google Scholar 

  6. Wu F, Chipman A, Pati S, Miyasawa B, Corash L, Kozar RA. Resuscitative strategies to modulate the endotheliopathy of trauma: from cell to patient. Shock Augusta Ga. 2020;53(5):575–84. https://doi.org/10.1097/SHK.0000000000001378.

    Article  PubMed  Google Scholar 

  7. Johansson PI, Stensballe J, Rosenberg I, Hilsløv TL, Jørgensen L, Secher NH. Proactive administration of platelets and plasma for patients with a ruptured abdominal aortic aneurysm: evaluating a change in transfusion practice. Transfusion (Paris). 2007;47(4):593–8. https://doi.org/10.1111/j.1537-2995.2007.01160.x.

    Article  Google Scholar 

  8. Borgman MA, Spinella PC, Perkins JG, et al. The ratio of blood products transfused affects mortality in patients receiving massive transfusions at a combat support hospital. J Trauma. 2007;63(4):805–13. https://doi.org/10.1097/TA.0b013e3181271ba3.

    Article  PubMed  Google Scholar 

  9. Holcomb JB, Wade CE, Michalek JE, et al. Increased plasma and platelet to red blood cell ratios improves outcome in 466 massively transfused civilian trauma patients. Ann Surg. 2008;248(3):447–58. https://doi.org/10.1097/SLA.0b013e318185a9ad.

    Article  PubMed  Google Scholar 

  10. Cotton BA, Au BK, Nunez TC, Gunter OL, Robertson AM, Young PP. Predefined massive transfusion protocols are associated with a reduction in organ failure and postinjury complications. J Trauma. 2009;66(1):41–8. https://doi.org/10.1097/TA.0b013e31819313bb. (discussion 48-49).

    Article  PubMed  Google Scholar 

  11. Shaz BH, Dente CJ, Nicholas J, et al. Increased number of coagulation products in relationship to red blood cell products transfused improves mortality in trauma patients. Transfusion (Paris). 2010;50(2):493–500. https://doi.org/10.1111/j.1537-2995.2009.02414.x.

    Article  CAS  Google Scholar 

  12. Sperry JL, Guyette FX, Brown JB, et al. Prehospital plasma during air medical transport in trauma patients at risk for hemorrhagic shock. N Engl J Med. 2018;379(4):315–26. https://doi.org/10.1056/NEJMoa1802345.

    Article  PubMed  Google Scholar 

  13. Torres LN, Sondeen JL, Ji L, Dubick MA, Torres FI. Evaluation of resuscitation fluids on endothelial glycocalyx, venular blood flow, and coagulation function after hemorrhagic shock in rats. J Trauma Acute Care Surg. 2013;75(5):759–66. https://doi.org/10.1097/TA.0b013e3182a92514.

    Article  CAS  PubMed  Google Scholar 

  14. Zhang L, Zeng M, Fan J, Tarbell JM, Curry FRE, Fu BM. Sphingosine-1-phosphate maintains normal vascular permeability by preserving endothelial surface glycocalyx in intact microvessels. Microcirc N Y N 1994. 2016;23(4):301–10.

    CAS  Google Scholar 

  15. Adamson RH, Clark JF, Radeva M, Kheirolomoom A, Ferrara KW, Curry FE. Albumin modulates S1P delivery from red blood cells in perfused microvessels: mechanism of the protein effect. Am J Physiol Heart Circ Physiol. 2014;306(7):H1011-1017. https://doi.org/10.1152/ajpheart.00829.2013.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Lopez E, Peng Z, Kozar RA, et al. Antithrombin III contributes to the protective effects of fresh frozen plasma following hemorrhagic shock by preventing syndecan-1 shedding and endothelial barrier disruption. Shock Augusta Ga. 2020;53(2):156–63. https://doi.org/10.1097/SHK.0000000000001432.

    Article  CAS  PubMed  Google Scholar 

  17. Deng X, Cao Y, Huby MP, et al. Adiponectin in fresh frozen plasma contributes to restoration of vascular barrier function after hemorrhagic shock. Shock Augusta Ga. 2016;45(1):50–4. https://doi.org/10.1097/SHK.0000000000000458.

    Article  CAS  PubMed  Google Scholar 

  18. Haywood-Watson RJ, Holcomb JB, Gonzalez EA, et al. Modulation of syndecan-1 shedding after hemorrhagic shock and resuscitation. PLoS ONE. 2011;6(8): e23530. https://doi.org/10.1371/journal.pone.0023530.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Kozar RA, Peng Z, Zhang R, et al. Plasma restoration of endothelial glycocalyx in a rodent model of hemorrhagic shock. Anesth Analg. 2011;112(6):1289–95. https://doi.org/10.1213/ANE.0b013e318210385c.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Peng Z, Pati S, Potter D, et al. Fresh frozen plasma lessens pulmonary endothelial inflammation and hyperpermeability after hemorrhagic shock and is associated with loss of syndecan 1. Shock Augusta Ga. 2013;40(3):195–202. https://doi.org/10.1097/SHK.0b013e31829f91fc.

    Article  CAS  PubMed  Google Scholar 

  21. Nohé B, Kiefer RT, Ploppa A, Haeberle HA, Schroeder TH, Dieterich HJ. The effects of fresh frozen plasma on neutrophil-endothelial interactions. Anesth Analg. 2003;97(1):216–21. https://doi.org/10.1213/01.ane.0000065906.35792.dd.

    Article  PubMed  Google Scholar 

  22. Diebel ME, Diebel LN, Liberati DM. Protective effects of plasma products on the endothelial-glycocalyx barrier following trauma-hemorrhagic shock: Is sphingosine-1 phosphate responsible? J Trauma Acute Care Surg. 2019;87(5):1061–9. https://doi.org/10.1097/TA.0000000000002446.

    Article  CAS  PubMed  Google Scholar 

  23. Jaffe EA. Cell biology of endothelial cells. Hum Pathol. 1987;18(3):234–9. https://doi.org/10.1016/s0046-8177(87)80005-9.

    Article  CAS  PubMed  Google Scholar 

  24. Weinbaum S, Tarbell JM, Damiano ER. The structure and function of the endothelial glycocalyx layer. Annu Rev Biomed Eng. 2007;9:121–67. https://doi.org/10.1146/annurev.bioeng.9.060906.151959.

    Article  CAS  PubMed  Google Scholar 

  25. Filmus J, Capurro M, Rast J. Glypicans. Genome Biol. 2008;9(5):224. https://doi.org/10.1186/gb-2008-9-5-224.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Carey DJ. Syndecans: multifunctional cell-surface co-receptors. Biochem J. 1997;327(1):1–16. https://doi.org/10.1042/bj3270001.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Saito A, Munakata H. Analysis of plasma proteins that bind to glycosaminoglycans. Biochim Biophys Acta. 2007;1770(2):241–6. https://doi.org/10.1016/j.bbagen.2006.10.015.

    Article  CAS  PubMed  Google Scholar 

  28. Varki A, Cummings RD, Esko JD, et al editors. Essentials of glycobiology. 4th ed. New York: Cold Spring Harbor Laboratory Press; 2022.

    Google Scholar 

  29. Reitsma S, Slaaf DW, Vink H, van Zandvoort MAMJ, Oude Egbrink MGA. The endothelial glycocalyx: composition, functions, and visualization. Pflugers Arch. 2007;454(3):345–59. https://doi.org/10.1007/s00424-007-0212-8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Schött U, Solomon C, Fries D, Bentzer P. The endothelial glycocalyx and its disruption, protection and regeneration: a narrative review. Scand J Trauma Resusc Emerg Med. 2016;24:48. https://doi.org/10.1186/s13049-016-0239-y.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Roumenina LT, Rayes J, Frimat M, Fremeaux-Bacchi V. Endothelial cells: source, barrier, and target of defensive mediators. Immunol Rev. 2016;274(1):307–29. https://doi.org/10.1111/imr.12479.

    Article  CAS  PubMed  Google Scholar 

  32. Daniel AE, van Buul JD. Endothelial junction regulation: a prerequisite for leukocytes crossing the vessel wall. J Innate Immun. 2013;5(4):324–35. https://doi.org/10.1159/000348828.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Salvador B, Arranz A, Francisco S, et al. Modulation of endothelial function by Toll like receptors. Pharmacol Res. 2016;108:46–56. https://doi.org/10.1016/j.phrs.2016.03.038.

    Article  CAS  PubMed  Google Scholar 

  34. Alphonsus CS, Rodseth RN. The endothelial glycocalyx: a review of the vascular barrier. Anaesthesia. 2014;69(7):777–84. https://doi.org/10.1111/anae.12661.

    Article  CAS  PubMed  Google Scholar 

  35. Yen W, Cai B, Yang J, et al. Endothelial surface glycocalyx can regulate flow-induced nitric oxide production in microvessels in vivo. PLoS ONE. 2015;10(1):e0117133. https://doi.org/10.1371/journal.pone.0117133.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Dolmatova EV, Wang K, Mandavilli R, Griendling KK. The effects of sepsis on endothelium and clinical implications. Cardiovasc Res. 2021;117(1):60–73. https://doi.org/10.1093/cvr/cvaa070.

    Article  CAS  PubMed  Google Scholar 

  37. Lygizos MI, Yang Y, Altmann CJ, et al. Heparanase mediates renal dysfunction during early sepsis in mice. Physiol Rep. 2013;1(6):e00153. https://doi.org/10.1002/phy2.153.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Schmidt EP, Yang Y, Janssen WJ, et al. The pulmonary endothelial glycocalyx regulates neutrophil adhesion and lung injury during experimental sepsis. Nat Med. 2012;18(8):1217–23. https://doi.org/10.1038/nm.2843.

    Article  CAS  PubMed  Google Scholar 

  39. Mulivor AW, Lipowsky HH. Inhibition of glycan shedding and leukocyte-endothelial adhesion in postcapillary venules by suppression of matrixmetalloprotease activity with doxycycline. Microcirc N Y N 1994. 2009;16(8):657–66. https://doi.org/10.3109/10739680903133714.

    Article  CAS  Google Scholar 

  40. Garsen M, Benner M, Dijkman HB, et al. Heparanase is essential for the development of acute experimental glomerulonephritis. Am J Pathol. 2016;186(4):805–15. https://doi.org/10.1016/j.ajpath.2015.12.008.

    Article  CAS  PubMed  Google Scholar 

  41. Zeng Y, Adamson RH, Curry FRE, Tarbell JM. Sphingosine-1-phosphate protects endothelial glycocalyx by inhibiting syndecan-1 shedding. Am J Physiol Heart Circ Physiol. 2014;306(3):H363-372. https://doi.org/10.1152/ajpheart.00687.2013.

    Article  CAS  PubMed  Google Scholar 

  42. Jedlicka J, Becker BF, Chappell D. Endothelial Glycocalyx. Crit Care Clin. 2020;36(2):217–32. https://doi.org/10.1016/j.ccc.2019.12.007.

    Article  PubMed  Google Scholar 

  43. Naumann DN, Hazeldine J, Davies DJ, et al. Endotheliopathy of trauma is an on-scene phenomenon, and is associated with multiple organ dysfunction syndrome: a prospective observational study. Shock Augusta Ga. 2018;49(4):420–8. https://doi.org/10.1097/SHK.0000000000000999.

    Article  PubMed  Google Scholar 

  44. Barry M, Pati S. Targeting repair of the vascular endothelium and glycocalyx after traumatic injury with plasma and platelet resuscitation. Matrix Biol Plus. 2022;14:100107. https://doi.org/10.1016/j.mbplus.2022.100107.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Wei S, Gonzalez Rodriguez E, Chang R, et al. Elevated syndecan-1 after trauma and risk of sepsis: a secondary analysis of patients from the pragmatic, randomized optimal platelet and plasma ratios (PROPPR) trial. J Am Coll Surg. 2018;227(6):587–95. https://doi.org/10.1016/j.jamcollsurg.2018.09.003.

    Article  PubMed  PubMed Central  Google Scholar 

  46. Aldecoa C, Llau JV, Nuvials X, Artigas A. Role of albumin in the preservation of endothelial glycocalyx integrity and the microcirculation: a review. Ann Intensive Care. 2020;10(1):85. https://doi.org/10.1186/s13613-020-00697-1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Galvani S, Sanson M, Blaho VA, et al. HDL-bound sphingosine 1-phosphate acts as a biased agonist for the endothelial cell receptor S1P1 to limit vascular inflammation. Sci Signal. 2015;8(389):ra79. https://doi.org/10.1126/scisignal.aaa2581.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Christensen PM, Liu CH, Swendeman SL, et al. Impaired endothelial barrier function in apolipoprotein M-deficient mice is dependent on sphingosine-1-phosphate receptor 1. FASEB J Off Publ Fed Am Soc Exp Biol. 2016;30(6):2351–9. https://doi.org/10.1096/fj.201500064.

    Article  CAS  Google Scholar 

  49. Christoffersen C, Obinata H, Kumaraswamy SB, et al. Endothelium-protective sphingosine-1-phosphate provided by HDL-associated apolipoprotein M. Proc Natl Acad Sci U S A. 2011;108(23):9613–8. https://doi.org/10.1073/pnas.1103187108.

    Article  PubMed  PubMed Central  Google Scholar 

  50. Chappell D, Jacob M, Hofmann-Kiefer K, et al. Antithrombin reduces shedding of the endothelial glycocalyx following ischaemia/reperfusion. Cardiovasc Res. 2009;83(2):388–96. https://doi.org/10.1093/cvr/cvp097.

    Article  CAS  PubMed  Google Scholar 

  51. Chappell D, Hofmann-Kiefer K, Jacob M, et al. TNF-alpha induced shedding of the endothelial glycocalyx is prevented by hydrocortisone and antithrombin. Basic Res Cardiol. 2009;104(1):78–89. https://doi.org/10.1007/s00395-008-0749-5.

    Article  CAS  PubMed  Google Scholar 

  52. Lamboo M, Poland DCW, Eikenboom JCJ, et al. Coagulation parameters of thawed fresh-frozen plasma during storage at different temperatures. Transfus Med Oxf Engl. 2007;17(3):182–6. https://doi.org/10.1111/j.1365-3148.2007.00729.x.

    Article  CAS  Google Scholar 

  53. Sidhu RS, Le T, Brimhall B, Thompson H. Study of coagulation factor activities in apheresed thawed fresh frozen plasma at 1–6 degrees C for five days. J Clin Apheresis. 2006;21(4):224–6. https://doi.org/10.1002/jca.20095.

    Article  PubMed  Google Scholar 

  54. Downes KA, Wilson E, Yomtovian R, Sarode R. Serial measurement of clotting factors in thawed plasma stored for 5 days. Transfusion (Paris). 2001;41(4):570. https://doi.org/10.1046/j.1537-2995.2001.41040570.x.

    Article  CAS  Google Scholar 

  55. Pusateri AE, Given MB, Schreiber MA, et al. Dried plasma: state of the science and recent developments. Transfusion (Paris). 2016;56(Suppl 2):S128-139. https://doi.org/10.1111/trf.13580.

    Article  Google Scholar 

  56. Mok G, Hoang R, Khan MW, et al. Freeze-dried plasma for major trauma - Systematic review and meta-analysis. J Trauma Acute Care Surg. 2021;90(3):589–602. https://doi.org/10.1097/TA.0000000000003012.

    Article  PubMed  Google Scholar 

  57. Pati S, Matijevic N, Doursout MF, et al. Protective effects of fresh frozen plasma on vascular endothelial permeability, coagulation, and resuscitation after hemorrhagic shock are time dependent and diminish between days 0 and 5 after thaw. J Trauma. 2010;69(Suppl 1):S55-63. https://doi.org/10.1097/TA.0b013e3181e453d4.

    Article  PubMed  PubMed Central  Google Scholar 

  58. Deb S, Sun L, Martin B, et al. Lactated ringer’s solution and hetastarch but not plasma resuscitation after rat hemorrhagic shock is associated with immediate lung apoptosis by the up-regulation of the Bax protein. J Trauma. 2000;49(1):47–53. https://doi.org/10.1097/00005373-200007000-00007. (discussion 53-55).

    Article  CAS  PubMed  Google Scholar 

  59. Wataha K, Menge T, Deng X, et al. Spray-dried plasma and fresh frozen plasma modulate permeability and inflammation in vitro in vascular endothelial cells. Transfusion (Paris). 2013;53(Suppl 1):80S-90S. https://doi.org/10.1111/trf.12040.

    Article  CAS  Google Scholar 

  60. Torres Filho IP, Torres LN, Salgado C, Dubick MA. Plasma syndecan-1 and heparan sulfate correlate with microvascular glycocalyx degradation in hemorrhaged rats after different resuscitation fluids. Am J Physiol Heart Circ Physiol. 2016;310(11):H1468-1478. https://doi.org/10.1152/ajpheart.00006.2016.

    Article  PubMed  Google Scholar 

  61. Spinella PC, Perkins JG, Grathwohl KW, et al. Effect of plasma and red blood cell transfusions on survival in patients with combat related traumatic injuries. J Trauma. 2008;64(2 Suppl):S69-77. https://doi.org/10.1097/TA.0b013e318160ba2f.

    Article  PubMed  Google Scholar 

  62. Brown LM, Aro SO, Cohen MJ, et al. A high fresh frozen plasma: packed red blood cell transfusion ratio decreases mortality in all massively transfused trauma patients regardless of admission international normalized ratio. J Trauma. 2011;71(2 Suppl 3):S358-363. https://doi.org/10.1097/TA.0b013e318227f152.

    Article  PubMed  PubMed Central  Google Scholar 

  63. Holcomb JB, del Junco DJ, Fox EE, et al. The prospective, observational, multicenter, major trauma transfusion (PROMMTT) study: comparative effectiveness of a time-varying treatment with competing risks. JAMA Surg. 2013;148(2):127–36. https://doi.org/10.1001/2013.jamasurg.387.

    Article  PubMed  PubMed Central  Google Scholar 

  64. Holcomb JB, Tilley BC, Baraniuk S, et al. Transfusion of plasma, platelets, and red blood cells in a 1:1:1 vs a 1:1:2 ratio and mortality in patients with severe trauma: the PROPPR randomized clinical trial. JAMA. 2015;313(5):471–82. https://doi.org/10.1001/jama.2015.12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Holcomb JB, Donathan DP, Cotton BA, et al. Prehospital transfusion of plasma and red blood cells in trauma patients. Prehosp Emerg Care. 2015;19(1):1–9. https://doi.org/10.3109/10903127.2014.923077.

    Article  PubMed  Google Scholar 

  66. Gruen DS, Brown JB, Guyette FX, et al. Prehospital plasma is associated with distinct biomarker expression following injury. JCI Insight. 2020;5(8):e135350. https://doi.org/10.1172/jci.insight.135350.

    Article  PubMed  PubMed Central  Google Scholar 

  67. Nelson A, Berkestedt I, Bodelsson M. Circulating glycosaminoglycan species in septic shock. Acta Anaesthesiol Scand. 2014;58(1):36–43. https://doi.org/10.1111/aas.12223.

    Article  CAS  PubMed  Google Scholar 

  68. Nelson A, Berkestedt I, Schmidtchen A, Ljunggren L, Bodelsson M. Increased levels of glycosaminoglycans during septic shock: relation to mortality and the antibacterial actions of plasma. Shock Augusta Ga. 2008;30(6):623–7. https://doi.org/10.1097/SHK.0b013e3181777da3.

    Article  CAS  PubMed  Google Scholar 

  69. Anand D, Ray S, Srivastava LM, Bhargava S. Evolution of serum hyaluronan and syndecan levels in prognosis of sepsis patients. Clin Biochem. 2016;49(10–11):768–76. https://doi.org/10.1016/j.clinbiochem.2016.02.014.

    Article  CAS  PubMed  Google Scholar 

  70. Ikeda M, Matsumoto H, Ogura H, et al. Circulating syndecan-1 predicts the development of disseminated intravascular coagulation in patients with sepsis. J Crit Care. 2018;43:48–53. https://doi.org/10.1016/j.jcrc.2017.07.049.

    Article  CAS  PubMed  Google Scholar 

  71. Chen S, He Y, Hu Z, et al. Heparanase mediates intestinal inflammation and injury in a mouse model of sepsis. J Histochem Cytochem. 2017;65(4):241–9. https://doi.org/10.1369/0022155417692536.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Lever R, Rose MJ, McKenzie EA, Page CP. Heparanase induces inflammatory cell recruitment in vivo by promoting adhesion to vascular endothelium. Am J Physiol Cell Physiol. 2014;306(12):C1184-1190. https://doi.org/10.1152/ajpcell.00269.2013.

    Article  CAS  PubMed  Google Scholar 

  73. LaRivière WB, Schmidt EP. The pulmonary endothelial glycocalyx in ARDS: a critical role for heparan sulfate. Curr Top Membr. 2018;82:33–52. https://doi.org/10.1016/bs.ctm.2018.08.005.

    Article  CAS  PubMed  Google Scholar 

  74. Chang R, Holcomb JB, Johansson PI, Pati S, Schreiber MA, Wade CE. Plasma resuscitation improved survival in a cecal ligation and puncture rat model of sepsis. Shock Augusta Ga. 2018;49(1):53–61. https://doi.org/10.1097/SHK.0000000000000918.

    Article  PubMed  Google Scholar 

  75. Scheck M, Velten M, Klaschik S, et al. Differential modulation of endothelial cell function by fresh frozen plasma. Life Sci. 2020;254: 117780. https://doi.org/10.1016/j.lfs.2020.117780.

    Article  CAS  PubMed  Google Scholar 

  76. Barry M, Wu F, Pati S, Chipman A, Geng H, Kozar R. Lyophilized plasma resuscitation downregulates inflammatory gene expression in a mouse model of sepsis. J Trauma Acute Care Surg. 2022;93(2S Suppl 1):S119–27. https://doi.org/10.1097/TA.0000000000003658.

    Article  CAS  PubMed  Google Scholar 

  77. Straat M, Müller MCA, Meijers JCM, et al. Effect of transfusion of fresh frozen plasma on parameters of endothelial condition and inflammatory status in non-bleeding critically ill patients: a prospective substudy of a randomized trial. Crit Care Lond Engl. 2015;19(1):163. https://doi.org/10.1186/s13054-015-0828-6.

    Article  Google Scholar 

  78. El-Nawawy AA, Elshinawy MI, Khater DM, et al. Outcome of early hemostatic intervention in children with sepsis and nonovert disseminated intravascular coagulation admitted to PICU: a randomized controlled trial. Pediatr Crit Care Med J Soc Crit Care Med World Fed Pediatr Intensive Crit Care Soc. 2021;22(3):e168–77. https://doi.org/10.1097/PCC.0000000000002578.

    Article  Google Scholar 

  79. Dietrich M, Hölle T, Lalev LD, et al. Plasma transfusion in septic shock—a secondary analysis of a retrospective single-center cohort. J Clin Med. 2022;11(15):4367. https://doi.org/10.3390/jcm11154367.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Benson AB, Moss M, Silliman CC. Transfusion-related acute lung injury (TRALI): a clinical review with emphasis on the critically ill. Br J Haematol. 2009;147(4):431–43. https://doi.org/10.1111/j.1365-2141.2009.07840.x.

    Article  PubMed  PubMed Central  Google Scholar 

  81. Pandey S, Vyas GN. Adverse effects of plasma transfusion. Transfusion (Paris). 2012;52(51):65S-79S. https://doi.org/10.1111/j.1537-2995.2012.03663.x.

    Article  CAS  Google Scholar 

  82. Chapman CE, Stainsby D, Jones H, et al. Ten years of hemovigilance reports of transfusion-related acute lung injury in the United Kingdom and the impact of preferential use of male donor plasma. Transfusion (Paris). 2009;49(3):440–52. https://doi.org/10.1111/j.1537-2995.2008.01948.x.

    Article  Google Scholar 

  83. Eder AF, Herron RM, Strupp A, et al. Effective reduction of transfusion-related acute lung injury risk with male-predominant plasma strategy in the American Red Cross (2006–2008). Transfusion (Paris). 2010;50(8):1732–42. https://doi.org/10.1111/j.1537-2995.2010.02652.x.

    Article  Google Scholar 

  84. Lucas G, Win N, Calvert A, et al. Reducing the incidence of TRALI in the UK: the results of screening for donor leucocyte antibodies and the development of national guidelines. Vox Sang. 2012;103(1):10–7. https://doi.org/10.1111/j.1423-0410.2011.01570.x.

    Article  CAS  PubMed  Google Scholar 

  85. Klein HG, Dodd RY, Dzik WH, et al. Current status of solvent/detergent-treated frozen plasma. Transfusion (Paris). 1998;38(1):102–7. https://doi.org/10.1046/j.1537-2995.1998.38198141508.x.

    Article  CAS  Google Scholar 

  86. Erickson A, Waldhaus K, David T, et al. Plasma treated with amotosalen and ultraviolet A light retains activity for hemostasis after 5 days post-thaw storage at 1 to 6o C. Transfusion (Paris). 2017;57(4):997–1006. https://doi.org/10.1111/trf.13973.

    Article  CAS  Google Scholar 

  87. Zander AL, Olson EJ, Van Gent JM, et al. Does resuscitation with plasma increase the risk of venous thromboembolism? J Trauma Acute Care Surg. 2015;78(1):39–44. https://doi.org/10.1097/TA.0000000000000480.

    Article  CAS  PubMed  Google Scholar 

  88. Iannizzi C, Chai KL, Piechotta V, et al. Convalescent plasma for people with COVID-19: a living systematic review. Cochrane Database Syst Rev. 2023;2(2):CD013600. https://doi.org/10.1002/14651858.CD013600.pub5.

    Article  PubMed  Google Scholar 

  89. Pati S, Peng Z, Wataha K, Miyazawa B, Potter DR, Kozar RA. Lyophilized plasma attenuates vascular permeability, inflammation and lung injury in hemorrhagic shock. PLOS ONE. 2018;13(2):e0192363.

    Article  PubMed  PubMed Central  Google Scholar 

  90. Diebel LN, Martin JV, Liberati DM. Microfluidics: A high-throughput system for the assessment of the endotheliopathy of trauma and the effect of timing of plasma administration on ameliorating shock-associated endothelial dysfunction. J Trauma Acute Care Surg. 2018;84(4):575–82. https://doi.org/10.1097/TA.0000000000001791.

    Article  PubMed  Google Scholar 

  91. Nelson A, Statkevicius S, Schött U, Johansson PI, Bentzer P. Effects of fresh frozen plasma, Ringer’s acetate and albumin on plasma volume and on circulating glycocalyx components following haemorrhagic shock in rats. Intensive Care Med Exp. 2016;4(1):6. https://doi.org/10.1186/s40635-016-0080-7.

    Article  PubMed  PubMed Central  Google Scholar 

  92. Potter DR, Baimukanova G, Keating SM, et al. Fresh frozen plasma and spray-dried plasma mitigate pulmonary vascular permeability and inflammation in hemorrhagic shock. J Trauma Acute Care Surg. 2015;78(6 Suppl 1):S7–17. https://doi.org/10.1097/TA.0000000000000630.

    Article  CAS  PubMed  Google Scholar 

  93. Moore HB, Moore EE, Chapman MP, et al. Plasma-first resuscitation to treat haemorrhagic shock during emergency ground transportation in an urban area: a randomised trial. Lancet Lond Engl. 2018;392(10144):283–91. https://doi.org/10.1016/S0140-6736(18)31553-8.

    Article  Google Scholar 

Download references

Acknowledgements

Figure 1 was prepared by Rebecca Windmueller, PhD with guidance from MSK, EPS, and NIS.

Funding

MSK is supported by the National Heart, Lung, and Blood Institute of the National Institutes of Health (grant number T32HL155020). The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed substantially to this work. MSK and NK drafted the manuscript. EPS provided basic science expertise. NIS, IJS, AAG, and EPS provided clinical expertise. All authors reviewed the manuscript and approved the manuscript before submission.

Corresponding author

Correspondence to M. S. Kravitz.

Ethics declarations

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kravitz, M.S., Kattouf, N., Stewart, I.J. et al. Plasma for prevention and treatment of glycocalyx degradation in trauma and sepsis. Crit Care 28, 254 (2024). https://doi.org/10.1186/s13054-024-05026-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13054-024-05026-7