Skip to main content
Advertisement
  • Loading metrics

Protecting the endothelial glycocalyx in COVID-19

Introduction

COVID-19 is a respiratory infection caused by the highly transmissible SARS-CoV-2. While most infected patients exhibit mild to moderate symptoms, a minority may develop severe vascular damage, which can be fatal. One example of this damage is disruption to the endothelial glycocalyx (EG) layer. EG alterations have been implicated in microcirculation changes and severe endothelial dysfunction in COVID-19 [1,2]. Current evidence suggests that host–pathogen interactions, such as hyperimmune activation or persistent presence of viral antigens, are associated with EG damage and may potentially bring about continuous endothelial dysfunction even after clinical recovery. Notably, the glycocalyx is also present on the epithelium, but we will focus on the EG and its implication on vascular health in COVID-19. Additionally, we will propose possible mediators that are linked with EG damage and suggest therapeutic agents that could aid EG restoration and protection.

Damage to the endothelial glycocalyx layer may affect vascular health

The EG layer, composed of a network of membrane-bound filament-like glycoproteins lining the luminal surface of the endothelium, functions to maintain vascular haemostasis and modulate leukocyte–endothelial interactions, blood flow, and vascular permeability (reviewed in [3]). These glycoproteins include heparan sulfate (HS), syndecan and hyaluronan, which are susceptible to inflammatory insults. Disruption to these glycoproteins is hypothesised to precede endothelial injury and weakens the structural integrity of the vasculature. These disruptions may give rise to increased pulmonary permeability, disrupt shear stress sensing of the endothelial cells, and affect physiological flow (Fig 1). In support, observational studies have shown that a thinner vascular EG, quantified by video imaging, correlates with significant reductions in vascular density and reduced red blood cell velocity, which is associated with increased disease severity [1,2]. Additionally, severe COVID-19 cases exhibit increased circulation of EG proteoglycans (syndecan-1, HS, and hyaluronan) and markers of endothelial injury (vascular endothelial growth factors, angiopoietin-1, and soluble thrombomodulin), compared to non-severe and uninfected cases [1,2,4,5]. Together, these findings suggest that in severe COVID-19, endothelial injury is closely linked to the integrity of the EG, and damage to the endothelial lining may lead to severe vascular complications (reviewed in [6]). Interestingly, endothelial dysfunction may persist in some recovered individuals, as shown by increased levels of circulating endothelial cells and syndecan-1 shedding [5,7]. This has been attributed to unresolved viral presence and/or inflammation, contributing to prolonged EG damage and dislodgement of endothelial cells from injured blood vessels [79]. The exact cause of EG damage or prolonged damage in COVID-19 remains to be determined, but current evidence suggests that host–pathogen interactions may play a role in EG injury.

thumbnail
Fig 1. Diagram depicting the lower respiratory tract, uninfected alveolus (left panel) and infected alveolus (right panel).

Uninfected condition (left panel), in an uninfected condition, the EG regulates the mechanotransduction of flow-induced shear stress in the vascular lumen, promoting a homeostatic environment. SARS-CoV-2 infection (right panel), SARS-CoV-2 infection activates the immune defence to eliminate the pathogen. In the alveolus, activated alveolar macrophages release inflammatory cytokines, driving a local inflammatory response. If unresolved, this inflammation may damage the alveolus. Damaged to the (EG) lining promotes neutrophil migration to the site of infection. Activated neutrophils release antimicrobials such as myeloperoxidase (MPO) and neutrophil extracellular traps, which can damage the glycocalyx. Other sheddases including matrix metalloproteinase (MMP), heparanase, and hyaluronidase, are also released and may further disrupt the glycocalyx integrity. These events damage the pulmonary vasculature, which can lead to fluid buildup in the lungs (pulmonary edema) and respiratory failure. Importantly, a damaged EG also affects mechanotransduction in the circulatory system, potentially altering endothelial cell functions in various organs. This may give rise to multiorgan failure, which is fatal in severe COVID-19 cases. Created with Biorender.

https://doi.org/10.1371/journal.ppat.1012203.g001

Proteoglycans of endothelial glycocalyx may promote spike protein interaction with endothelial cells

The spike protein on the virion is known to bind to the angiotensin converting enzyme 2 (ACE2) receptor to initiate cellular entry, and HS has been proposed to mediate initial virus-ACE2 interaction [10]. It is worth noting that the majority of cellular surfaces are covered by proteoglycans, making them likely targets for viruses like coronaviruses to exploit as attachment factors and receptors for entry [11]. For example, in laboratory settings, the interaction between spike protein and HS led to conformational changes in the virion that promoted binding to ACE2. Furthermore, SARS-CoV-2 binding is greatly reduced upon cleavage of HS [10]. This demonstrates a complex interplay, where cleaving of HS is likely to reduce the availability of HS that promotes early attachment but could also increase ACE2 expression, potentially making cells more susceptibility to direct spike protein interaction [12]. Whether increased ACE2 exposure translates to higher SARS-CoV-2 infectivity in the epithelium is unclear, but peak viral load has been correlated with disease severity [13]. Further investigations into whether synthetic HS or ACE 2 can block viral attachment, thus, reducing infection are warranted as a potential prophylaxis against coronavirus infection [14,15].

Activated host responses contribute to the damage of the endothelial glycocalyx

The specific mechanism responsible for EG degradation is not fully understood. It is postulated that elevated levels of sheddases, such as heparanase, hyaluronidase, and matrix metalloproteases (MMP) can promote EG shedding and endothelial cell activation (Fig 1) [4,1618]. Notably, heparanase and hyaluronidase are known mammalian enzymes that cleave HS and HA, respectively (reviewed in [19]). Damage to the EG exposes leukocyte-binding receptors (ICAM-1, VCAM-1), which are usually embedded within the EG. This can increase leukocyte extravasation, triggering a cascade of downstream activities that likely further damage the EG and endothelial cells [20]. To corroborate, in autopsied COVID-19 lung tissues, vascular and endothelial pathologies coupled with leukocyte infiltration have been observed [21]. This further supports the notion that uncontrolled leukocyte extravasation, along with EG and endothelial cell damage, can cause vascular injury. While in other viruses such as dengue virus, secreted viral antigen has been demonstrated to damage the EG (reviewed in [22]). However, current evidence does not support this hypothesis in SARS-CoV-2.

Activated leukocytes, such as neutrophils, release antimicrobial agents like myeloperoxidase (MPO), which has been shown to correlate with syndecan-1 shedding in COVID-19 [5]. MPO has also been proposed to bind to HS, resulting in the weakening of EG structure and subsequent damage [23]. Similarly, increased inflammatory mediators including IL-6, IL-8, and TNF-ɑ have been associated with elevated plasma concentrations of syndecan-1, hyaluronan, and HS in severe COVID-19 [4,16]. These cytokines may also act as stimuli to affect the integrity of the EG. For example, IL-6 binds to glycosaminoglycans, which can either weaken the EG structure and/or promote EG degradation via paracrine effects [24]. On the other hand, glycosaminoglycan fragments such as hyaluronan, isolated from COVID-19 subjects, have been demonstrated to bind to hyaluronan receptors on endothelial cells. This results in the activation of Rho-associated protein kinase signalling, a known pathway that triggers EG disruption [4]. Together, various factors can either directly or indirectly affect the integrity of EG. Therefore, minimising EG damage presents a potential pathway to reducing the risk of severe pathology.

Protecting the endothelial glycocalyx may reduce disease severity

A damaged EG contributes to impaired vascular health, and several classes of interventions have the potential to minimise COVID-19-associated EG damage (Table 1). In vitro studies have shown that neutralising antibodies against the spike protein block binding of SARS-CoV-2 on HS and ACE2, thereby reducing infection and inflammation [25,26]. Additionally, low molecular weight heparin (LMWH) was shown to bind to the spike protein, which led to conformational change, that interfered with virus attachment to HS and reduced viral entry [25,27]. Moreover, LMWH was associated with a significant reduction in IL-6 levels in COVID-19 patients and appeared to have inhibitory properties against heparanase [16,28]. Similarly, tocilizumab, an IL-6 receptor blocker has been demonstrated to improve COVID-19 outcomes [29]. Treatment with tocilizumab protected against EG degradation in vivo, suggesting that reducing IL-6 function may reduce downstream inflammatory pathways that are associated with EG degradation [30]. Dexamethasone, a corticosteroid that has anti-inflammatory properties, is used in COVID-19 treatment. In septic mouse models, dexamethasone significantly reduced MMP activities and protected against EG damage [31]. In a COVID-19 clinical study, dexamethasone treatment was associated with significant reductions in C-reactive protein and markers of endothelial injury. However, whether the improved outcomes were due to MMP reduction that protected against EG degradation in COVID-19 remains uncertain [32]. Overall, reducing infection and inflammation is likely to protect against EG degradation [4,5,17,25].

thumbnail
Table 1. Potential compounds that may either reduce and/or restore the endothelial glycocalyx.

https://doi.org/10.1371/journal.ppat.1012203.t001

Possible therapies that restore the endothelial glycocalyx

Complementary therapies that minimise inflammatory-induced EG degradation are therapeutics that aid the regeneration of EG (Table 1). In COVID-19, treatment with a tyrosine kinase inhibitor, imatinib, has been demonstrated to improve endothelial barrier functions [33]. In an in vitro model, treating compromised endothelial cells with imatinib was shown to improve EG thickness and restore EG functions, although the precise mechanism remains to be determined [34]. Extracts from marine algae, fucoidan and rhamnan sulfate (RS), which are safe in humans [35,36], have been shown to restore EG both in vivo and in vitro. Notably, fucoidan, an HS mimetic, reduced endothelial activation and promoted EG restoration on endothelial cells previously treated with COVID-19 serum [37]. Fucoidan is proposed to contain amino acids capable of stimulating HS and hyaluronan synthesis (reviewed in [38]). Additionally, fucoidan improved EG thickness in aged mice (where thinning of EG is common due to aging) and increased nitric oxide (NO) bioavailability [39]. NO is a potent vasodilator that promotes blood flow and reduced NO production increases the risk of endothelial dysfunction (reviewed in [40]). Similarly, RS, another HS mimetic, limited leukocyte adhesion, increased HS coverage on endothelial cells and promoted EG regeneration in a study on vascular inflammation [41]. Sulodexide, a mixture of heparin and dermatan sulfate, is used to treat peripheral vascular disease. Administrating sulodexide to septic mice restored EG and reduced vascular permeability [42]. Sulodexide was also reported to reduce hyaluronidase activity in type 2 diabetic subjects, which was associated with increased EG regeneration [43]. Besides these repurposed compounds, fluid therapy with fresh frozen plasma (FFP) and albumin are often used in blood volume resuscitation in various vascular conditions. For instance, FFP contains most of the plasma proteins needed for EG maintenance, whereas albumin is a major component in plasma and both interventions have been shown to improve EG thickness in septic conditions [44]. Additionally, albumin carries sphingosine-1-phosphate, a bioactive lipid metabolite that has been demonstrated to stabilise and prevent EG shedding (reviewed in [44]). Despite these promising therapies to aid the restoration of the EG, a gold standard treatment has yet to be translated clinically.

Conclusions

Available evidence suggests that EG disruption contributes to severe COVID-19 outcomes. The therapeutic candidates presented here offer promising avenues for developing interventions that can protect and restore the EG. Further studies are urgently needed to evaluate the efficacy and safety of these candidates, whether alone or in combination, to prepare for future outbreaks and improve patient outcomes for future outbreaks.

References

  1. 1. Stahl K, Gronski PA, Kiyan Y, Seeliger B, Bertram A, Pape T, et al. Injury to the Endothelial Glycocalyx in Critically Ill Patients with COVID-19. Am J Respir Crit Care Med. 2020;202(8):1178–81. Epub 2020/08/25 06:00. pmid:32833500.
  2. 2. Rovas A, Osiaevi I, Buscher K, Sackarnd J, Tepasse PR, Fobker M, et al. Microvascular dysfunction in COVID-19: the MYSTIC study. Angiogenesis. 2021;24(1):145–57. Epub 20201014. pmid:33058027; PubMed Central PMCID: PMC7556767.
  3. 3. Weinbaum S, Cancel LM, Fu BM, Tarbell JM. The Glycocalyx and Its Role in Vascular Physiology and Vascular Related Diseases. Cardiovasc Eng Technol. 2021;12(1):37–71. Epub 2020/09/23 06:00. pmid:32959164.
  4. 4. Queisser KA, Mellema RA, Middleton EA, Portier I, Manne BK, Denorme F, et al. COVID-19 generates hyaluronan fragments that directly induce endothelial barrier dysfunction. JCI Insight. 2021;6(17):147472. Epub 2021/07/28 06:00. pmid:34314391.
  5. 5. Teo A, Chan LLY, Cheung C, Chia PY, Ong SWX, Fong SW, et al. Myeloperoxidase inhibition may protect against endothelial glycocalyx shedding induced by COVID-19 plasma. Commun Med (Lond). 2023;3(1):62. Epub 2023/05/06 09:42. pmid:37147421.
  6. 6. Zha D, Fu M, Qian Y. Vascular Endothelial Glycocalyx Damage and Potential Targeted Therapy in COVID-19. Cells. 2022;11(12). Epub 2022/06/25 06:00. pmid:35741101.
  7. 7. Chioh FW, Fong SW, Young BE, Wu KX, Siau A, Krishnan S, et al. Convalescent COVID-19 patients are susceptible to endothelial dysfunction due to persistent immune activation. Elife. 2021;10(10):64909. Epub 2021/03/24 06:00. pmid:33752798.
  8. 8. Owusu D, Pomeroy MA, Lewis NM, Wadhwa A, Yousaf AR, Whitaker B, et al. Persistent SARS-CoV-2 RNA Shedding Without Evidence of Infectiousness: A Cohort Study of Individuals With COVID-19. J Infect Dis. 2021;224(8):1362–1371. pmid:33649773; PubMed Central PMCID: PMC7989388.
  9. 9. Goh D, Lim JCT, Fernaíndez SB, Joseph CR, Edwards SG, Neo ZW, et al. Case report: Persistence of residual antigen and RNA of the SARS-CoV-2 virus in tissues of two patients with long COVID. Front Immunol. 2022;13:939989. Epub 20220905. pmid:36131932; PubMed Central PMCID: PMC9483160.
  10. 10. Clausen TM, Sandoval DR, Spliid CB, Pihl J, Perrett HR, Painter CD, et al. SARS-CoV-2 Infection Depends on Cellular Heparan Sulfate and ACE2. Cell. 2020;183(4):1043–57 e15. Epub 2020/09/25 06:00. pmid:32970989.
  11. 11. Kim SH, Kearns FL, Rosenfeld MA, Votapka L, Casalino L, Papanikolas M, et al. SARS-CoV-2 evolved variants optimize binding to cellular glycocalyx. Cell Rep Phys Sci. 2023;4(4):101346. Epub 20230407. pmid:37077408; PubMed Central PMCID: PMC10080732.
  12. 12. Targosz-Korecka M, Kubisiak A, Kloska D, Kopacz A, Grochot-Przeczek A, Szymonski M. Endothelial glycocalyx shields the interaction of SARS-CoV-2 spike protein with ACE2 receptors. Sci Rep. 2021;11(1):12157. Epub 2021/06/11 06:00. pmid:34108510.
  13. 13. Stankiewicz Karita HC, Dong TQ, Johnston C, Neuzil KM, Paasche-Orlow MK, Kissinger PJ, et al. Trajectory of Viral RNA Load Among Persons With Incident SARS-CoV-2 G614 Infection (Wuhan Strain) in Association With COVID-19 Symptom Onset and Severity. JAMA Netw Open. 2022;5(1):e2142796. Epub 2022/01/11 06:00. pmid:35006245.
  14. 14. Monteil V, Kwon H, Prado P, Hagelkrüys A, Wimmer RA, Stahl M, et al. Inhibition of SARS-CoV-2 Infections in Engineered Human Tissues Using Clinical-Grade Soluble Human ACE2. Cell. 2020;181(4):905–13.e7. Epub 20200424. pmid:32333836; PubMed Central PMCID: PMC7181998.
  15. 15. Guimond SE, Mycroft-West CJ, Gandhi NS, Tree JA, Le TT, Spalluto CM, et al. Synthetic Heparan Sulfate Mimetic Pixatimod (PG545) Potently Inhibits SARS-CoV-2 by Disrupting the Spike-ACE2 Interaction. ACS Cent Sci. 2022;8(5):527–45. Epub 20220329. pmid:35647275; PubMed Central PMCID: PMC9136977.
  16. 16. Buijsers B, Yanginlar C, de Nooijer A, Grondman I, Maciej-Hulme ML, Jonkman I, et al. Increased Plasma Heparanase Activity in COVID-19 Patients. Front Immunol. 2020;11(575047):575047. Epub 2020/10/31 06:00. pmid:33123154.
  17. 17. Potje SR, Costa TJ, Fraga-Silva TFC, Martins RB, Benatti MN, Almado CEL, et al. Heparin prevents in vitro glycocalyx shedding induced by plasma from COVID-19 patients. Life Sci. 2021;276(119376):119376. Epub 2021/03/31 06:00. pmid:33781826.
  18. 18. Syed F, Li W, Relich RF, Russell PM, Zhang S, Zimmerman MK, et al. Excessive Matrix Metalloproteinase-1 and Hyperactivation of Endothelial Cells Occurred in COVID-19 Patients and Were Associated With the Severity of COVID-19. J Infect Dis. 2021;224(1):60–69. pmid:33885811; PubMed Central PMCID: PMC8083685.
  19. 19. Becker BF, Jacob M, Leipert S, Salmon AH, Chappell D. Degradation of the endothelial glycocalyx in clinical settings: searching for the sheddases. Br J Clin Pharmacol. 2015;80(3):389–402. Epub 20150522. pmid:25778676; PubMed Central PMCID: PMC4574825.
  20. 20. Schmidt EP, Yang Y, Janssen WJ, Gandjeva A, Perez MJ, Barthel L, et al. The pulmonary endothelial glycocalyx regulates neutrophil adhesion and lung injury during experimental sepsis. Nat Med. 2012;18(8):1217–23. Epub 20120722. pmid:22820644; PubMed Central PMCID: PMC3723751.
  21. 21. Ackermann M, Verleden SE, Kuehnel M, Haverich A, Welte T, Laenger F, et al. Pulmonary Vascular Endothelialitis, Thrombosis, and Angiogenesis in Covid-19. N Engl J Med. 2020;383(2):120–8. Epub 2020/05/22 06:00. pmid:32437596.
  22. 22. Teo A, Chua CLL, Chia PY, Yeo TW. Insights into potential causes of vascular hyperpermeability in dengue. PLoS Pathog. 2021;17(12):e1010065. Epub 2021/12/10 06:00. pmid:34882753.
  23. 23. Manchanda K, Kolarova H, Kerkenpaß C, Mollenhauer M, Vitecek J, Rudolph V, et al. MPO (Myeloperoxidase) Reduces Endothelial Glycocalyx Thickness Dependent on Its Cationic Charge. Arterioscler Thromb Vasc Biol. 2018;38(8):1859–67. Epub 2018/06/16 06:00. pmid:29903730.
  24. 24. Mummery RS, Rider CC. Characterization of the heparin-binding properties of IL-6. J Immunol. 2000;165(10):5671–9. Epub 2000/11/09 11:00. pmid:11067924.
  25. 25. Bermejo-Jambrina M, Eder J, Kaptein TM, van Hamme JL, Helgers LC, Vlaming KE, et al. Infection and transmission of SARS-CoV-2 depend on heparan sulfate proteoglycans. EMBO J. 2021;40(20):e106765. Epub 2021/09/13 06:00. pmid:34510494.
  26. 26. Chen X, Li R, Pan Z, Qian C, Yang Y, You R, et al. Human monoclonal antibodies block the binding of SARS-CoV-2 spike protein to angiotensin converting enzyme 2 receptor. Cell Mol Immunol. 2020;17(6):647–9. Epub 20200420. pmid:32313207; PubMed Central PMCID: PMC7167496.
  27. 27. Mycroft-West CJ, Su D, Pagani I, Rudd TR, Elli S, Gandhi NS, et al. Heparin Inhibits Cellular Invasion by SARS-CoV-2: Structural Dependence of the Interaction of the Spike S1 Receptor-Binding Domain with Heparin. Thromb Haemost. 2020;120(12):1700–15. Epub 2020/12/29 06:00. pmid:33368089.
  28. 28. Shi C, Wang C, Wang H, Yang C, Cai F, Zeng F, et al. The Potential of Low Molecular Weight Heparin to Mitigate Cytokine Storm in Severe COVID-19 Patients: A Retrospective Cohort Study. Clin Transl Sci. 2020;13(6):1087–95. Epub 2020/09/04 06:00. pmid:32881340.
  29. 29. Rosas IO, Bräu N, Waters M, Go RC, Hunter BD, Bhagani S, et al. Tocilizumab in Hospitalized Patients with Severe Covid-19 Pneumonia. N Engl J Med. 2021;384(16):1503–16. Epub 2021/02/26 06:00. pmid:33631066.
  30. 30. Ikonomidis I, Pavlidis G, Katsimbri P, Lambadiari V, Parissis J, Andreadou I, et al. Tocilizumab improves oxidative stress and endothelial glycocalyx: A mechanism that may explain the effects of biological treatment on COVID-19. Food Chem Toxicol. 2020;145(111694):111694. Epub 2020/08/22 06:00. pmid:32822775.
  31. 31. Cui N, Wang H, Long Y, Su L, Liu D. Dexamethasone Suppressed LPS-Induced Matrix Metalloproteinase and Its Effect on Endothelial Glycocalyx Shedding. Mediators Inflamm. 2015;2015(10):912726. Epub 2015/07/23 06:00. pmid:26199464.
  32. 32. Kim WY, Kweon OJ, Cha MJ, Baek MS, Choi SH. Dexamethasone may improve severe COVID-19 via ameliorating endothelial injury and inflammation: A preliminary pilot study. PLoS ONE. 2021;16(7):e0254167. Epub 2021/07/03 06:00. pmid:34214123.
  33. 33. de Brabander J, Duijvelaar E, Schippers JR, Smeele PJ, Peters-Sengers H, Duitman JW, et al. Immunomodulation and endothelial barrier protection mediate the association between oral imatinib and mortality in hospitalised COVID-19 patients. Eur Respir J. 2022;60(6):00780–2022. Epub 2022/07/28 06:00. pmid:35896211.
  34. 34. Li Y, Sun Z, Zhu H, Sun Y, Shteyman DB, Markx S, et al. Inhibition of Abl Kinase by Imatinib Can Rescue the Compromised Barrier Function of 22q11.2DS Patient-iPSC-Derived Blood-Brain Barriers. Cells. 2023;12(3). Epub 2023/02/12 06:00. pmid:36766762.
  35. 35. Shimada Y, Terasawa M, Okazaki F, Nakayama H, Zang L, Nishiura K, et al. Rhamnan sulphate from green algae Monostroma nitidum improves constipation with gut microbiome alteration in double-blind placebo-controlled trial. Sci Rep. 2021;11(1):13384. Epub 20210705. pmid:34226572; PubMed Central PMCID: PMC8257721.
  36. 36. Tsai HL, Tai CJ, Huang CW, Chang FR, Wang JY. Efficacy of Low-Molecular-Weight Fucoidan as a Supplemental Therapy in Metastatic Colorectal Cancer Patients: A Double-Blind Randomized Controlled Trial. Mar Drugs. 2017;15(4). Epub 20170421. pmid:28430159; PubMed Central PMCID: PMC5408268.
  37. 37. Yuan L, Cheng S, Sol W, van der Velden AIM, Vink H, Rabelink TJ, van den Berg BM. Heparan sulfate mimetic fucoidan restores the endothelial glycocalyx and protects against dysfunction induced by serum of COVID-19 patients in the intensive care unit. ERJ Open Res. 2022;8(2):00652–2021. Epub 2022/05/06 06:00. pmid:35509442.
  38. 38. Karam MB, El Khoury J, Chakar C, Changotade S, Lutomski D, Naaman N, et al. Heparan-mimetics: Potential agents of tissue regeneration for bone and periodontal therapies. Med Nov Technol Devices. 2021;11:100066.
  39. 39. Machin DR, Nguyen D, Bramwell RC, Lesniewski LA, Donato AJ. Dietary Glycocalyx Precursor Supplementation Ameliorates Age-Related Vascular Dysfunction. FASEB J. 2019;33(S1):828.1–1.
  40. 40. Cyr AR, Huckaby LV, Shiva SS, Zuckerbraun BS. Nitric Oxide and Endothelial Dysfunction. Crit Care Clin. 2020;36(2):307–321. pmid:32172815; PubMed Central PMCID: PMC9015729.
  41. 41. Patil NP, Gómez-Hernández A, Zhang F, Cancel L, Feng X, Yan L, et al. Rhamnan sulfate reduces atherosclerotic plaque formation and vascular inflammation. Biomaterials. 2022;291(121865):121865. Epub 2022/11/05 06:00. pmid:36332287.
  42. 42. Song JW, Zullo JA, Liveris D, Dragovich M, Zhang XF, Goligorsky MS. Therapeutic Restoration of Endothelial Glycocalyx in Sepsis. J Pharmacol Exp Ther. 2017;361(1):115–21. Epub 2017/02/09 06:00. pmid:28167639.
  43. 43. Broekhuizen LN, Lemkes BA, Mooij HL, Meuwese MC, Verberne H, Holleman F, et al. Effect of sulodexide on endothelial glycocalyx and vascular permeability in patients with type 2 diabetes mellitus. Diabetologia. 2010;53(12):2646–55. Epub 2010/09/25 06:00. pmid:20865240.
  44. 44. Uchimido R, Schmidt EP, Shapiro NI. The glycocalyx: a novel diagnostic and therapeutic target in sepsis. Crit Care. 2019;23(1):16. Epub 2019/01/19 06:00. pmid:30654825.
  45. 45. Evans JP, Zeng C, Carlin C, Lozanski G, Saif LJ, Oltz EM, et al. Neutralizing antibody responses elicited by SARS-CoV-2 mRNA vaccination wane over time and are boosted by breakthrough infection. Sci Transl Med. 2022;14(637):eabn8057. Epub 2022/02/16 06:00. pmid:35166573.
  46. 46. Hoang TN, Pino M, Boddapati AK, Viox EG, Starke CE, Upadhyay AA, et al. Baricitinib treatment resolves lower-airway macrophage inflammation and neutrophil recruitment in SARS-CoV-2-infected rhesus macaques. Cell. 2021;184(2):460–75.e21. Epub 20201110. pmid:33278358; PubMed Central PMCID: PMC7654323.
  47. 47. Zeng Y, Adamson RH, Curry FR, Tarbell JM. Sphingosine-1-phosphate protects endothelial glycocalyx by inhibiting syndecan-1 shedding. Am J Physiol Heart Circ Physiol. 2014;306(3):H363–72. Epub 2013/11/29 06:00. pmid:24285115.
  48. 48. Al-Kuraishy HM, Batiha GE, Al-Gareeb AI, Al-Harcan NAH, Welson NN. Receptor-dependent effects of sphingosine-1-phosphate (S1P) in COVID-19: the black side of the moon. Mol Cell Biochem. 2023;478(10):2271–9. Epub 2023/01/19 06:00. pmid:36652045.