REVIEW
published: 07 May 2021
doi: 10.3389/fvets.2021.661660
Frontiers in Veterinary Science | www.frontiersin.org 1 May 2021 | Volume 8 | Article 661660
Edited by:
Pablo Martín-Vasallo,
University of La Laguna, Spain
Reviewed by:
Ivo P. Torres Filho,
United States Army Institute of
Surgical Research, United States
Muhammad Kashif Iqbal,
Cholistan University of Veterinary and
Animal Sciences, Pakistan
*Correspondence:
Lisa Smart
Specialty section:
This article was submitted to
Comparative and Clinical Medicine,
a section of the journal
Frontiers in Veterinary Science
Received: 31 January 2021
Accepted: 16 March 2021
Published: 07 May 2021
Citation:
Smart L and Hughes D (2021) The
Effects of Resuscitative Fluid Therapy
on the Endothelial Surface Layer.
Front. Vet. Sci. 8:661660.
doi: 10.3389/fvets.2021.661660
The Effects of Resuscitative Fluid
Therapy on the Endothelial Surface
Layer
Lisa Smart
1
*
and Dez Hughes
2
1
School of Veterinary Medicine, College of Science, Health, Engineering and Education, Murdoch University, Murdoch, WA,
Australia,
2
Department of Veterinary Clinical Sciences, Faculty of Veterinary and Agricultural Sciences, Melbourne Veterinary
School, Werribee, VIC, Australia
The goal of resuscitative fluid therapy is to rapidly expand circulating blood volume
in order to restore tissue perfusion. Although this therapy often serves to improve
macrohemodynamic parameters, it c an be associated with adverse effects on the
microcirculation and endothelium. The endothelial surface layer (ESL) provides a
protective barrier over the endothelium and is important for regulating transvascular fluid
movement, vasomotor tone, coagulation, and inflammation. Shedding or thinning of the
ESL can promote interstitial edema and inflammation and may cause microcirculatory
dysfunction. The pathophysiologic perturbations of critical illness and rapid, large-volume
fluid therapy both cause shedding or thinning of the ESL. Research suggests that
restricting the volume of crystalloid, or “clear” fluid, may preserve some ESL integrity and
improve outcome based on animal experimental models and preliminary clinical trials in
people. This narrative review critica lly evaluates the evidence for the detrimental effects
of resuscitative fluid therapy on the ESL and provides suggestions for future research
directions in this field.
Keywords: endothelium, glycocalyx, shock, fluid therapy, crystalloid, colloid, endothelial surface layer
INTRODUCTION
The importance of microcirculatory function and health of the endothelium has become a large
area of interest for criticalists in the last two decades. Ov er this time, fluid resuscitation strategies
have experienced a shift in perspective. Aggressive “clear” fluid resuscitation was once considered
vital for stabilization of macrohemodynamic variables. However, in certain patient cohorts this
approach has now been shown to either not improve outcome or worsen outcome (
14). Though
fast administration of fluid often serves to normalize the commonly measured clinical parameters
during shock, microcirculation may not necessarily benefit from this therapy. There is growing
evidence that fluid resuscitation may actually harm t he endothelium by modifying, or shedding,
the endothelial surface layer (ESL).
The ESL includes a structural scaffold, the endothelial glycocalyx (EG), and associated molecules
suspended within a plasma layer. We use the term EG when referring to specific components of the
structure, or biomarkers of these components, whereas ESL is used when referring to the layer as a
whole. In regards to shedding of the ESL, the term ESL will be used when the evidence supports loss
of the whole layer and the term EG used when the evidence only supports that isolated components
have been shed. Use of the term glycocalyx refers to any type of cell surface glycocalyx and is
not restricted to the endothelium, such as during discussion of glycocalyx shedding biomarkers.
Smart and Hughes Fluids and Endothelial Surface Layer
Resuscitative fluid t h erapy includes a range of fluid choices,
including isotonic crystalloid fluid, synthetic colloid fluid, and
hyperosmolar crystalloid fluid, with the former being the
most common type of fluid used (5). In general, due to the
pharmacodynamic properties of these fluids in either healthy
subjects or shock models (
69), the volume of fluid is “large for
isotonic crystalloid (at least a quarter to half of estimated blood
volume), “moderate for synthetic colloid (an eighth to quarter
of estimated blood volume), and “small” for hyperosmolar
crystalloids. All three types of fluid resuscitation will be covered
in this review with a particular focus on i sotonic “large-volume
crystalloid fluid therapy due to t he evidence for its effects on
the ESL. A summary of the evidence reviewed in this article is
provided in Box 1.
STRUCTURE AND FUNCTION OF THE
ENDOTHELIAL SURFACE LAYER
Basic Structure and Function
Most cells in the body are covered in a protective layer of
carbohydrate scaffold, which houses many different molecules
that serve a variety of functions. This surface layer is called
a glycocalyx. The general structure of the glycocalyx has
similarities between cell ty pes, with only small variations
in individual proteins or carbohydrate molecules. The EG
coats the luminal surface of the endothelium and is a vital
structure for cell signaling and transvascular permeability. It
is composed of proteoglycans, glycosaminoglycans (GAGs),
and glycoproteins (Figure 1). Together with mobile or soluble
components, such as albumin, this compromises the ESL.
Proteoglycans are large molecules that have a cytoplasmic,
transcellular, and extracellular domain (syndecan) or are attached
by a glycosylphosphatidylinositol anchor (glypican-1) (Figure 1)
(
1012). Their extracellular component, or ectodomain, is
covered by GAG side-chains and performs important roles
that assist with cell to cell, or cell to matrix, interactions (13).
These structures provide a structural scaffold for the ESL, in
which many other molecules are housed. The sulfated GAGs
attached to proteoglycans include heparan, chondroitin, and
dermatan sulfate. Heparan sulfate is the most abundant GAG
on syndecans and glypican-1, which is why proteoglycans are
often referred to as heparan sulfate proteoglycans. An additional
GAG, hyaluronan, is not typically associated with a proteoglycan;
instead, it is attached to the endothelium via receptors such as
CD44 or other GAG molecules (Figure 1) (14, 15). Hyaluronan
is a long GAG of varying lengths that weaves its way throug h the
tall “forest” of proteoglycans, with their sulfated GAG “branches”.
These GAG chains contribute to the b a rrier function of the ESL
of excluding large molecules (16). Glycoproteins reside on the
luminal surface of the endothelium, hidden within the ESL, and
include a dhesion molecules such as integrins and selectins (17).
Glycoproteins play an important role in leucocyte trafficking
during states of inflammation (
18); many of their functions
are only initiated once the ESL has been shed or thinned.
Finally, mobile components residing within the forest of the ESL
include proteins, such as albumin, and anticoagulants, such as
BOX 1 | Summary of the evidence for effects of “clear” uid therapy on
endothelial surface layer (ESL) shedding or modication.
Proposed mechanisms of ESL shedding
Dilution of plasma proteins
Release of natriuretic peptides
Inflammatory cytokine release (certain fluid types)
Possible downstream effects
Exacerbation of inflammation
Microcirculatory dysfunction
Increased vascular permeability
Increased interstitial edema
Prothrombotic conditions
Proposed strategies that may mitigate ESL shedding
Reduction in dose of clear fluids
Slowing down administration of resuscitative fluids
“Earlier” vasopressor therapy for vasodilatory shock
Adjunctive protein administration (such as plasma)
These proposed strategies are not based on evidence from clinical veterinary
research and require further investigation. These guidelines are opinion of
the authors only, after considering the breadth of evidence available. It is
currently unknown if “glycoprotective” strategies benefit patients. Therapy
should always be tailored to individual patient needs, with prioritization of
reestablishing adequate perfusion and a thorough cost to benefit analysis.
antithrombin and tissue factor pathway inhibitor (15, 19). The
presence of plasma proteins are likely important for maintaining
the normal structure and permeability of the ESL, as well as
providing an anticoagulated surface at the blood interface.
The EG serves a range of functions, many of which are
still being characterized. These functions include maintenance
of a surface barrier that buffers circulating leucocytes, inhibits
coagulation, regulates fluid flux, and communicates changes in
vascular wall shear forces. The sulfated GAGs on the surface of
the endothelium generate and maintain a net negative charge,
which repels similarly charged cells from the endothelial surface
(
20). Although albumin has an overall negative charge, it is
likely that the positively charged groups within the molecular
structure are what allow for incorporation of albumin into the
ESL (
21). Maintenance of this negative charge is important for
endothelial integrity; loss of negative charge on the luminal
surface of the endothelium leads to extravasation of albumin
(2224). In addition to the importance of electrostatic charge,
the ESL provides a barrier to fluid filtration, thus creating a
protein-poor sub-glycocalyx layer and maintaining a colloid
osmotic pressure (COP) gradient favoring fluid retention with in
the vasculature (20).
The traditional Starling hypothesis describes a relationship
between the intravascular and the interstitial colloid osmotic
pressure and implies the importance of this relationship in
determining net transvascular fluid filtration. Many studies
in the last four decades, however, have built a body of
evidence stating that it is mainly the low CO P in the sub-
glycocalyx area that creates the pressure gradient opposing
capillary hydrostatic pressure (
25, 26). The theory is that the
Frontiers in Veterinary Science | www.frontiersin.org 2 May 2021 | Volume 8 | Article 661660
Smart and Hughes Fluids and Endothelial Surface Layer
FIGURE 1 | The scaffold of the endothelial glycocalyx, within the endothelial surface layer, is provided by proteoglycans, syndecan (four subtypes), and glypican-1.
Glycosaminoglycans are attached to proteoglycans (e.g., heparan sulfate) or the endothelial surface (hyaluronan). Molecules suspended in the plasma of the
endothelial surface layer include proteins such as albumin. These proteins create a protein-poor sub-glycocalyx area that is important for transvascular colloid osmotic
pressure balance. For simplicity, structures within the interendothelial cleft are not represented.
sub-glycocalyx fluid space, within the inter-endothelial cleft,
has a very low concentration of macromolecules. This is due
to the high imperme ability of the ESL to macromolecules
and the rapid flow of water and solutes through the sub-
glycocalyx space (
27). In the steady state with an intact
ESL, net filtration of fluid occurs across the blood vessel
wall, with only a small limitation from intravascular COP.
Increasing intravascular COP does not , and cannot, serve to
reverse net fluid transudation nor does increasing interstitial
COP lead to an increase in fluid transudation (28, 29). A
large drop in hydrostatic pressure, such as that may occur
during circulatory shock, may reverse transvascular fluid flow
to a resorptive state; however, this effect is transient (
29).
These hypot h eses explain why fluid would not be “drawn”
out of the interstitium by increasing intravascular COP, such
as the use of synthetic colloid fluids. Rather, they can
reduce extravasation compared to crystalloid fluid. The COP
of the intravascular space, and even of the sub-glycocalyx
space, is microanatomically and physiologically distant from
the interstitial COP. However, t he fluid dynamics across
the endothelium in disease states with a denuded ESL,
removing the influence of the sub-glycocalyx lay er, is yet to be
fully characterized.
The EG also plays an important role to changing pressures
and flow within the intravascular space. Proteoglycans, especially
those with heparan sulfate chains, play an important role in
responding to changes in vascular wall shear stress or changes in
intravascular pressure (10, 30, 31). Detection of these mechanical
forces leads to morphological changes in endothelial cells and
release of nitric oxide (
3235). Shear stress can also lead to a
change in location of proteoglycans or upregulation of their cell
surface expression (
3638). It is possible that change in location
or upregulation of these molecules during microcirculatory
disturbances may also affect shedding of the extracellular
Frontiers in Veterinary Science | www.frontiersin.org 3 May 2021 | Volume 8 | Article 661660
Smart and Hughes Fluids and Endothelial Surface Layer
components. This becomes relevant when discussing biomarkers
for EG shedding below.
Shedding
Shedding, or modification, of the ESL is a vital step after t issue
injury in order to facilitate leucocyte and platelet adherence
(
39). Shedding reproduced in cell culture or ex vivo models
leads to increased expression of adhesion molecules, increased
leucocyte adhesion, and increased cytokine production (32, 40
43). This process is likely more complex in vivo, whereby certain
disease states may have varying effects, from selective removal
of glycocalyx components to complete denudation. Although
ESL shedding is a necessary step in localized inflammation, it
is becoming clearer that systemic-wide shedding is associated
with severity of illness and poor outcome. There has also
been a growing concern in recent years that interventions
that promote ESL shedding may worsen clinical outcomes for
critically ill patients. Given that ESL shedding is associated with
exacerbating inflammation and increasing vascular permeability
in laboratory models, there is theoretical plausibility that limiting
ESL shedding may improve clinical outcome.
Shedding alters capillary perfusion, causing a decrease in
functional capillary density (44), in addition to increased
endothelial permeability (45). A decrease in functional capillary
density means that some vessels within a given area do
not have red blood cells traversing their course, consistent
with microcirculatory dysfunction. This can create regions
of tissue hypoxia. A clear association between ESL shedding
and impairment of microcirculatory blood flow is yet to be
demonstrated in either an in vivo animal model or clinical
study (46). However, given that ESL shedding via artificial
means can reduce capillary blood flow (44), it is mechanistically
plausible that shedding of the ESL in critical illness plays a
role in altered microcirculation. It has been well-demonstrated
that heterogeneity of capillary blood flow can persist in critical
illness despite normalization of macrohemodynamic variables,
such as blood pressure and cardiac output (4751). Further,
persistence of altered microcirculatory flow in critically ill
people is associated with severit y of illness and poor outcome
(47, 5153). Consequently, it appears important to identify
causes of microcirculatory dysfunction that occurs independent
of circulatory shock. More evidence is required to determine
a causal link between ESL shedding and microcirculatory
dysfunction, and whether protection of the ESL can prevent
the lat ter.
There may be downstream consequences to the release of
EG components into circulation. Shed components can stimulate
inflammation by acting as danger-associated molecular patterns
or “alarmins.” Soluble heparan sulfate molecules play a key
role in modulating inflammation, including leukocyte activation,
increasing production of cytokines, and endothelial activation
(
54, 55). Low molecular weight hyaluronan can also stimulate
production of inflammatory mediators (5659). In contrast,
shed components such as syndecan-1 and4 ectodomains can
have indirect anti-inflammatory effects by facilitating neutrophil
cytotoxicity (
60, 61). The complexity of EG components acting as
effector molecules in the systemic circulation may be analogous
to the systemic inflammatory response, where some cytokine
release is beneficial to the host response, whereas a “cytokine
storm” cre a tes pathological consequences.
It is unclear how long it takes for ESL recovery to occur.
Most evidence is based on data from in vitro cell culture or
ex vivo vascular models, which are unable to fully replicate in
vivo conditions. That being said, individual components can be
regenerated within 24 h (
37, 62), but restoration of the structure
itself can take up to 7 days (32, 63). During critical illness,
inflammation likely continues the shedding process, delaying
the repair. Further in vivo research is required using real-time
videomicroscopy or similar means in order to characterize the
temporal changes during ESL recovery.
Assessment of Shedding
Shedding of the ESL can be detected by a number of
means. Laboratory studies utilize several methods, including
measurement of circulating components of the EG (proteoglycan
ectodomains and GAGs), dete ct ion of EG components on the
surface of the endothelium, direct visualization of the ESL
via tissue fixation and microscopy, and indirect visualization
via real-time videomicroscopy. Clinical studies usually rely on
measurement of circulating components of the EG, or EG
biomarkers, in serum or plasma s a mples to assess systemic
shedding. The most frequently reported EG biomarkers are
syndecan-1, heparan sulfate, and hyaluronan. There are several
limitations to relying on this kind of assessment of the ESL.
Firstly, studies often only measure a single component of the
EG at a single point in time. That particular component may
have other sources of shedding. For example, syndecan-1 is not
only present on the surface of the endothelium but also on
epithelial cells and leucocytes (
6468). Also, highly relevant to
intravenous fluid therapy, hyaluronan is abundant throughout
the interstitium and can be “flushed through the lymphatics
back into systemic circulation (6971). It is unclear in critical
illness to what degree other sources of glycocalyx shedding
contribute to serum or plasma concentrations. Further, tissue
injury and inflammation during critical illness may upregulate
cell surface expression of these biomarkers, especially the
proteoglycans (38, 7283). Therefore, an increase in circulating
concentration may reflect an increase in cell turnover rather
than primarily cell surface shedding. Several recent studies have
demonstrated temporal differences in the shedding of several EG
components in people presenting to an emergency department
with sepsis, whereby hyaluronan increased early in treatment
whereas sydnecan-1 increased later (
8486). This raises questions
as to why some biomarkers increase earlier than others. It is
possible that the temporal differences relate to either alternative
sources of the biomarker or differences in upregulation.
Ideally, it would be best to visualize shedding of t he EG in vivo
rather than interpreting circulating biomarker concentrations.
These techniques are mostly reserved for use in laboratory
models rather than clinical use. Specialized tissue fixation
techniques can be used to directly visualize the ESL via electron
microscopy, often employed in non-survival rodent models
(
28, 8791). Given the fragility of the ESL, it is best that the
tissue is preserved via perfusion with fixative prior to de a th,
Frontiers in Veterinary Science | www.frontiersin.org 4 May 2021 | Volume 8 | Article 661660
Smart and Hughes Fluids and Endothelial Surface Layer
either perfusion of an isolated organ or t he whole body. Tissue
immersion techniques that avoid whole-body perfusion have
also been recently described (92). Anecdotally, it is challenging
to achieve quality images of intact ESL in tissue sections.
Alternatively, components of the EG may be fluorescently
labeled and visualized by confocal microscopy of cell culture
or tissue models (
93). Other laboratory techniques include dye
exclusion with intravital microscopy to estimate ESL thickness
(9496). Finally, ESL thickness may be estimated in real-time by
sidestream dark field microscopy in both large animal models
and clinical research (46, 9799). This technology approximates
ESL thick ness by measuring the perfused boundary region, which
is the region within the blood vessel that is peripheral to the
flow of red blood cells, or t h e immobile plasma layer. A lthough
this technique shows promise for future use in veterinary clinical
research (100), image acquisition can be technically difficult
and image quality is currently limited for reliably estimating
ESL t hi ckness.
MECHANISMS OF SHEDDING RELEVANT
TO FLUID THERAPY
Inflammation and Shock
In the pa tient requiring bolus fluid therapy, it is likely that
alteration of the ESL has already occurred due to the effects of
inflammatory mediators. For example, tumor necrosis factor-
α, a potent pro-inflammatory cytokine, causes reduction in
thickness of the ESL, shedding of syndecan-1,-4, and GAGs
from the endothelium, as well as upregulation of glycocalyx
components on the endothelial surface (
76, 81, 90, 101). Matrix
metalloproteinases, one of the main perpetrat ors for glycocalyx
shedding, are released by activated leucoytes (17). Tissue injury,
such as ischemia and reperfusion, and production of reactive
oxygen species also cause EG shedding (43, 102104). Activation
of coagulation may also affect the EG, as both plasmin and
thrombin can cleave syndecan ectodomains (74, 105). Relevant
to sepsis, bacterial components such as lipopolysaccharide and
chemotactic peptides also cause shedding of the EG (43, 77,
81, 106108). Therefore, it is likely that partial or complete
denudation of the ESL has already occurred before fluid therapy
is administered.
Shedding of the EG in states of critical illness, such as shock,
has been demonstrated in animal models before any fluid therapy
has commenced. In mice, hemorrhagic shock was associated with
a thinner pulmonary ESL, as measured by electron microscopy,
and downregulation of syndecan-1 on the endothelial surface,
compared to a sham model (
109, 110). In a canine hemorrhagic
shock model comparing different fluid interventions (111),
plasma hyaluronan concentration was significantly increased
across treatment groups after atraumatic blood removal, but
before fluid, compared to baseline (unpublished analysis). An
increase in plasma hyaluronan concentration has also been
demonstrated in a rodent sepsis model, 4 h after Escherichia
coli lipopolysaccharide infusion, where blood pressure was
maintained by norepinephrine administration (
112). There are
limited data on EG shedding in the clinical setting before
fluid therapy; however, serum syndecan-1 concentrations above
healthy control levels has been demonstrated pre-hospital in
human trauma patients, though the temporal relationship to fluid
administration by first responders is unknown (
113).
Hemodilution
Bearing in mind that alterations to the ESL may already
exist in critically ill patients, there is growing evidence that
administration of “clear” fluids may propagate ESL shedding.
Several clinical studies in critically ill people have identified
an association between volume of fluid administered and
EG biomarker concentrations, including increased hyaluronan
(
84), syndecan (114), and heparan sulfate concentrations (115).
Putting aside the confounders of inflammation and severity
of illness, bolus fluid therapy may have a direct effect on the
ESL via hemodilution and production of natriuretic peptides.
Ex vivo vascular models provide important data in regards
to this issue, as they exclude the effects of the varying
pharmacodynamic properties of fluid types administered in vivo.
These vascular models have shown that dilution of plasma with
crystalloid reduces ESL thickness. Notably, a mathematica l model
derived from meta-analysis of several studies demonstrated
that dilution of blood with fluid reduced vascular resistance,
independent of hematocrit and COP (116). Also, the decrease in
resistance caused by saline was reduced in magnitude when the
vasculature was pre-treated with heparinase. Heparinase sheds
heparan sulfate side-chains from proteoglyc ans, thus th inning
the glycocalyx (117). From this, it may be inferred that the
decrease in resistance caused by saline infusion is due to not
only changes in hemorrheology but also loss of the ESL. Further,
several studies have shown that perfusion of blood vessels with
crystalloid solution incre ases vascular permeability. One such
study showed an inverse linear relationship between albumin
concentration in the perfusate and hydraulic conductivity,
or permeability, of vasculature (
118). Return to baseline
permeability was not achieved by simply increasing the albumin
concentration back to baseline levels; it required a much higher
albumin concentration. This implies modification of the ESL
and its affinity for albumin after crystalloid infusion. Further
experiments showed that perfusion of blood vessels with plasma
was more effective at restoring normal vas cular permeability
after crystalloid infusion (119), compared with an albumin
solution, indicating that substances within plasma are import ant
for maintaining normal transvascular fluid flux. A more recent
study showed that infusion of guinea pig coronary vasculature
with 0.9% saline significantly increased fluid extravasation,
compared to low molecular weight (LMW) hydroxyethyl starch
(HES) (Voluven
R
) and albumin solution (
45). Pretreatment
with heparinase combined with saline infusion did not yield
higher fluid extravasation; stripping the EG before infusion was
just as detrimental as saline infusion alone. Interestingly, no
differences between fluid treatments in the appearance of the ESL
were appreciated using electron microscopy. Finally, a murine
hemorrhagic shock model compared 15 mL/kg of either fresh
whole blood (FWB), packed red blood cells in lactated Ringer’s
solution (PRBC in LRS), or washed PRBC in LRS, or 75 mL/kg of
plain LRS (
120). Rats that received either washed PRBC or LRS
Frontiers in Veterinary Science | www.frontiersin.org 5 May 2021 | Volume 8 | Article 661660
Smart and Hughes Fluids and Endothelial Surface Layer
had decreased ESL thi ckness compared to baseline, as measured
by intravital microscopy, whereas those that received either FWB
or PRBC did not show a significant difference to baseline. Results
were similar for change in plasma heparan sulfate concentration.
Therefore, it appeared that the presence of protein within the
FWB and unwashed PRBC products was effective at mitigating
ESL shedding.
Natriuretic Peptid es
Another mechanism of EG shedding during bolus fluid therapy
is the action of natriuretic peptides on the endothelium. Atrial
and brain natriuretic peptides (ANP and BNP, respectively) are
released from the cardiac atria and ventricles during stretch of
the myocardium (
121). These peptides counteract the effects
of hypervolemia by decre asing systemic vascular resistance and
causing a natriuresis, among other mechanisms. Natriuretic
peptides also increase vascular permeability (
122, 123). A more
recently discovered role of these peptides is shedding of the
EG. All three major natriuretic peptides (ANP, BNP, and C-
type NP) can shed the EG, as measured by increased syndecan-
1 and heparan sulfate concentrations in the effluent of guinea
pig coronary arteries (88, 124). These studies also demonstrated
increased vascular permeability and visualized denudation of the
ESL via electron microscopy.
Fast intravenous administration of isotonic crystalloid fluid
may stimulate natriuretic peptide release during the bolus phase.
The dose of crysta lloid administered for treatment of shock far
exceeds the volume remaining in circulation after redistribution.
This rapid rise and fall of blood volume during a crystalloid
bolus was demonstrated by Silverstein and others (
7), whereby
the blood volume rapidly rose by 76% during a large bolus
of 0.9% saline before dissipating. Therefore, it is possible that
atria may become “over-stretched” during crystalloid bolus
fluid therapy and release natriuretic peptides, which may then
contribute to systemic EG shedding. Two studies in human
surgical patients have shown a rise in A NP in parallel with
EG biomarker concentrations after fluid loading (125, 126).
These studies showed a significant increase in serum syndecan-
1 and hy aluronan concentrations but not heparan sulf ate. One
additional study showed an increase in ANP and all three EG
biomarker concentrations in cardiac surgical patients; in both
those undergoing cardiopulmonary bypass and those undergoing
“off-pump” procedures (
127). In contrast, one study in people
undergoing hysterectomy (n = 26) showed no significant rise
in BNP or biomarker concentrations (syndecan-1 a nd heparan
sulfate) after 25 mL/kg of LRS given over 30 min during surgery
(128). An additional study showed no significant rise in BNP or
any EG biomarker concentrations (syndecan-1, heparan sulfate,
hyaluronan) in human surgical patients or healthy volunteers
during a modest fluid load of 3 mL/kg of 20% albumin, though
sample size was small (n = 15 per group) (129). A recent canine
hemorrhagic shock study did not observe a significant increase in
plasma ANP concentration after 80 mL/kg of balanced isotonic
crystalloid given over 20 min, however small sample size (n = 6)
and baseline variability may have hampered the ability to detect a
difference (
111).
Other Considerations Related to EG
Biomarker Type
One of the challenges of interpreting studies that measure EG
biomarker concentrations as the primary assessment of EG
shedding is the variability in choice of biomarker. Across multiple
studies investigating the effect of fluid loading on the EG, it
appears that hyaluronan is consistently increased immediately
after a large volume of crystalloid, or synthetic colloid fluid
in th e setting of hypervolemia (
111, 125, 126). Syndecan-1
concentration significantly increases after fluid in humans and
rats (
110, 125, 126, 130132), but not in all studies (128, 133).
Heparan sulfate has not been shown to significantly increase in
humans (12 5, 126, 128) but has been shown to increase in rats
(120, 130, 133). Unpicking t h ese consistencies is difficult due
to differences across species, study designs, fluid doses, t iming
of intervention and blood s a mpling, and choice of comparator
(other fluid vs. no fl uid vs. sham). Studies may also vary
in whether or not they adjust the biomarker concentration
for the effects of hemodilution, such as indexing to albumin,
hemoglobin, or other tracer concentration in the blood. Although
this may help to account for the different pharmacodynamics
of each fluid type, and their variable dilution of blood tracer
components, indexation of biomarker concentrations can create
a margin of error. On closer inspection of individual studies,
inconsistency has been shown within the studies themselves,
showing a significant change in one biomarker but not the other
(
125, 126, 133). As mentioned above, the source of circulating
EG biomarkers may not be restricted to th e endothelium
and other sources, such as the interstitium or surface of
other cells, may be contributing. Therefore, a healthy sceptism
should be applied to any conclusion drawn from biomarker
concentrations alone.
COMPARATIVE EFFECTS OF DIFFERENT
FLUID TYPES
Much of t he evidence concerning EG or ESL shedding
after fluid therapy concerns the use of large volumes
of crystalloid fluid. Administering other fluid types
that have less redistribution to the interstitium may
theoretically be associated with less EG shedding. This
section assesses the evidence for the comparative effect of
crystalloid, synthetic colloid, and hy pertonic fluids on the
EG or ESL.
Isotonic Crystalloid Fluid
Multiple rodent studies have shown that administration of large
volumes of cryst a lloid fluid for hemorrhagic shock is associated
with more EG shedding, compared to fluids containing protein
(
109, 110, 120, 130133). However, it is difficult to compare
interventions in regards to effects on the EG in many of
these studies, as cardiovascular parameters are either not
closely monitored or are different between t reatment groups.
Shock itself, without fluid resuscitation, can cause EG shedding
(
109, 110, 131, 134136), which be c omes a confounder when
comparing resuscitation strategies that provide inequitable blood
Frontiers in Veterinary Science | www.frontiersin.org 6 May 2021 | Volume 8 | Article 661660
Smart and Hughes Fluids and Endothelial Surface Layer
volume expansion. However, several key studies allow some
assessment of this issue. A murine hemorrhagic shock model
showed that administration of protein-poor fluids, either washed
PRBCs or LRS, was associated with a thinner ESL, whereas
protein-containing fluids (FWB and PRBCs) were not (120).
Cardiovascular parameters after resuscitation appeared similar
across these groups, based on the limited data available. Another
murine hemorrhagic shock model compared seven different
fluid strategies: 15 mL/kg of FWB, PRBC, fresh frozen plasma
(FFP) or 5% albumin, 8 mL/kg of 3% saline, 45 mL/kg of
0.9% saline, or 75 mL/k g of LRS (13 0). Shock index, usually
defined by heart rate divided by systolic blood pressure,
was compared before hemorrhage and immediately after fluid
resuscitation within each fluid group. Rats that received either
5% albumin, LRS, 3% saline, or 0.9% saline had a significantly
increased shock index after fluid resuscitation, whereas rats
that received FWB, PRBC, and FFP showed no change in
shock index. When comparing only the fluid types that did
not resolve shock, based on shock index, rats that received 5%
albumin had higher ESL thickness and lower EG biomarker
concentrations, compared to the crystalloid groups. Finally,
a murine hemorrhagic shock model used a blood pressure-
targeted resuscitation method for comparing crystalloid fluid
with plasma (
109). Administration of LRS was associated
with a thinner ESL, visualized on electron microscopy, and
decreased endothelial expression of syndecan-1, compared to
administration of plasma.
There is a scarcity of “large animal” models, or those
including pigs, sheep, and dogs, that have assessed ESL shedding
after fluid resuscitation. This may be parti ally due to a
limitation on validated assays available for measuring glycocalyx
biomarkers across these species. Current commercially available
validated options for assessi ng EG shedding are restricted to
circulating hyaluronan concentration. A canine hemorrhagic
shock model did not detect a significant difference in plasma
hyaluronan concentrations when comparing dogs that received
20 mL/kg of FWB (n = 6) with those that received 80
mL/kg of Plasmalyte-148
R
(n = 6) (
111). Other differences
in parameters were seen between the crystalloid and colloid
fluid groups in this study, which is discussed further below. In
an ovine endotoxemia model, a pressure-targeted resuscitation
method using vasopressor th erapy was used to compare
either norepinephrine alone or 0. 9% sali ne (40 mL/kg) in
combination with norepinephrine (
137). Although there was
no comparison to resuscitation with a protein-containing fluid,
the comparison of crystalloid fluid to no fluid in the setting
of sepsis provided interesting results. There was no significant
difference in serum hyaluronan concentrations between groups
over time, though sheep that received 0.9% saline showed
a gre at er rate of increase in this biomarker. This greater
increase was not observed directly after the fluid bolus, like
in the aforementioned canine study, but beyond 6 h, which
may have been related to worsening septic shock in this
group. Atrial natriuretic peptide was significantly higher at
the end of the fluid bolus in the sheep that received fluid
(see previous discussion on natriuretic peptides). Detection of
between-group differences was also likely affected by small
sample size in th is study (n = 8 per group), similar t o the
study in dogs.
Synthetic Colloid Fluids
Given t hat crystalloid fluids generally require a larger volume
to expand circulating blood volume to the same extent as
colloid fluids (
69), crystalloids cause greater hemodilution
during the fluid bolus and, potentially, greater natriuretic
peptide release. This begs the question if administration
of colloid fluids causes less EG shedding than crystalloid
fluid. Two main types of synthetic colloid are currently in
use: HES and gelatin products. The aforementioned canine
hemorrhagic shock study included both of these fluid types at
20 mL/kg, as comparator fluids to 80 mL/kg of Plasmalyte-
148
R
(
111). Dogs administered HES had significantly lower
plasma hyaluronan concentration immediately after the fluid
bolus, compared to both FWB and crystalloid groups. In
contrast, the group that received 4% succinylated gelatin had
significantly increased plasma hyaluronan concentration 40
and 100 min after the end of the fluid bolus, compared to
FWB. Given that inflammation may cause EG shedding, these
differential effects observed between these two colloids may
be due to release of inflammatory mediators; HES has been
associated with mitigation of inflammation (
138141) whereas
gelatin has been associated with pro-inflammatory effects (142).
However, there were no differences in plasma concentrations
of inflammatory mediators between the two colloid groups in
this study (111). Therefore, the effect of gelatin on hyaluronan
shedding may be a direct effect of the fluid, rather than
pro-inflammatory effects, such as washout of the interstitium.
Another hemorrhagic shock study in rats replaced the shed
volume with crystalloid, at either shed volume, two ti mes shed
volume, or three times shed volume, or shed volume with
HES (n = 6 per group) (
141). This study did not identify
any significant differences between groups in response of blood
pressure to fluid resuscitation, organ syndecan-1 expression,
or circulating syndecan-1 concentrati on. A hemorrhagic shock,
stroke volume-targeted resuscitation model in pigs c ompared
balanced crystalloid to HES and found no differences in post-
fluid serum syndecan-1 or glypican concentrations (6). Blood was
sampled 120 min after commencement of fluids, therefore any
peaks in EG biomarker shedding may have been missed. Given
the limited evidence for different effects of colloid fluids on the
EG or ESL, compared to crystalloids, a conclusion cannot be
currently drawn.
Hypertonic Crystalloid Fluids
Hypertonic solutions exert immunomodulatory effects both
in in vitro and animal model studies, including decreased
leucocyte response to pro-inflammatory stimuli or circulatory
shock states, when compared to other fluid types (
143156).
Administration of hypertonic saline to human trauma patients
also reduced inflammatory biomarker concentration, compared
to isotonic saline (
157, 158). Several studies have also shown a
reduction in vascular leakage of macromolecules (
147, 151, 155).
Given these effects, it is possible that hypertonic solutions may
have a beneficial effect in regards to reducing ESL shedding.
Frontiers in Veterinary Science | www.frontiersin.org 7 May 2021 | Volume 8 | Article 661660
Smart and Hughes Fluids and Endothelial Surface Layer
In the rodent hemorrhagic shock model previously discussed
comparing many different type of fluids, rats that received 8
mL/kg of 3% saline had a lower increase from baseline in
plasma heparan sulfate concentration, compared to both isotonic
crystalloid groups (45 mL/kg of 0.9% saline or 75 mL/kg of
LRS), and a lower loss of ESL thickness (
130). This difference
was despite a persistence of an elevated shock index and
hyperlactatemia after resuscitation with 3% saline. Therefore,
although hypertonic saline may have had less impact on the
EG, it may have also provided inadequate fluid resuscitation. A
preliminary randomized clinical trial in people with suspected
sepsis could not demonstrate a difference between groups in
hyaluronan concentrations after treatment allocation of either
5 mL/kg of 3% saline or 10 mL/kg of 0.9% saline (86).
Serum syndecan-1 concentration decreased after 0.9% saline,
compared to b aseline, whereas serum syndecan-1 concentration
did not change after 3% saline. Though this significant difference
between groups may reflect hemodilution, as twice the volume of
0.9% saline was administered, this was not reflected in differences
in hemoglobin concentration. It is unclear what the clinical
significance is of this subtle difference in change in serum
syndecan-1 concentration, whether it reflects more syndecan-1
shedding in the hypertonic saline group or not. Limitations of
this study included a low severity of illness and small sample size,
and further studies are needed to clarify the effects of hypertonic
solutions on the EG.
GLYCOPROTECTIVE THERAPIES
The clinical relevance of ESL shedding due to fast or large-
volume fluid administration is yet to be determined. This type
of fluid administration can cause shedding of the ESL, which
may propagate microcirculatory dysfunction, inflammation,
and procoagulation. Therefore, there has been much interest
developing in “glycoprotective fluid strategies or adjunctive
therapies during the resuscitation phase.
Restricting or Avoiding Fluid
Blood volume expansion is th e cornerstone of managing
circulatory shock, with some types of shock an exception
such as cardiogenic shock. Conventionally, this is achieved by
administering large (more than 20 mL/kg) volumes of crystalloid
fluid intravenously as rapidly as possible, or as a bolus (
159, 160).
This serves to increase cardiac preload and improve cardiac
output. The need for this intervention is intuitive for shock
caused by hypovolemia; however, some types of shock have a
component of vasodilation and microcirculatory dysfunction.
This includes states of shock due to a systemic inflammatory
response, such as sepsis and blunt trauma. For these types
of shock, improvement of macrohemodynamic variables, such
as cardiac output and blood pressure, may not improve
microcirculatory blood flow in a linear way. This is particularly
pertinent to septic shock, where the role of fluid bolus th erapy
has been called into question, with a movement toward early
vasopressor therapy instead (
161, 162). Further to the lack of
improvement in microcirculatory flow, it has been suggested
that fluid bolus therapy in sepsis can propagate vasodilation,
due to endothelial shear stress, release of nitric oxide, and
direct vasodilatory effects of natriuretic peptides. This may
contribute to some patients becoming refractory to vasopressor
therapy. In an ovine endotoxemia model, administration of 40
mL/kg of 0.9% saline was associated with a significantly higher
dose of noradrenaline subsequently administered in order to
maintain blood pressure, compared to sheep that received only
noradrenaline (
137). Given the concern that bolus fluids may
cause harm in sepsis, a preliminary randomized clinical trial
was completed demonstrating feasibility of restricting crystalloid
fluid early in the treatment of sepsis in people (163). Following on
from this, a large multi-center human randomized clinical trial
is underway to compare a restrictive fluid strategy with a more
liberal one in people with early sepsis (164).
It is much less known if restricting crystalloid fluid in
other types of shock may be beneficial. In the setting of
human trauma, discussion concerning restrictive crystalloid
fluid strategies focuses on delayed resuscitation or permissive
hypotension in order to avoid clot disruption and dilutional
coagulopathy before definitive hemostasis (
4, 165). Although
animal models have shown that fluid resuscitation is likely
beneficial for severe uncontrolled hemorrhage, it increases the
risk of mortality in lower grades of severity of bleeding, compared
to not administering any fluid (166). Hypotensive resuscitation,
in parallel with lower volumes of administered fluids, is also
associated with a reduced risk of mort a lity in human trauma
patients (167, 168), th ough the evidence is mixed (169). This
benefit is likely related to improved coagulation and clot stability;
it is unknown if there are also benefits associated with reducing
endothelial dysfunction. Currently, the recommendation in
human trauma medicine is for limitation of “clear” fluid and
early, yet judicious, administration of blood products. This
includes packed red blood cells, plasma, and platelets in parallel
during resuscitation. The early administration of plasma may
not only serve to limit dilutional coagulopathy but also assist
with repairing the ESL. It is difficult to translate this practice
to veterinary medicine, given the cost of transfusion, limited
availability, and variability in blood banking practices. Given
there are many factors entering into the cost to benefit assessment
of administering plasma or albumin products to individuals,
theoretical benefits of plasma for the ESL are of lesser import ance,
until more is known. Also, caution should be a pplied when
restricting crystalloid fluid to any patient with shock, as the
benefits of a restrictive strategy may be only relevant to certain
patient populations, such as sepsis. This concept was highlighted
by a recent randomized clinical trial in human surgical patients
whereby participants randomized to receive a restrictive fluid
strategy had a significantly higher rate of acute kidney injury and
surgical site infection, compared to those randomized to a liberal
fluid strategy (
170).
Slowing Fluid Administration
Fast fluid administration serves to rapidly improve clinical
signs and macrohemodynamic variables, and has been the
bread-and-butter of emergency and critical care medicine
for decades. However, normalization of macrohemodynamic
variables in people does not always translat e to improved
Frontiers in Veterinary Science | www.frontiersin.org 8 May 2021 | Volume 8 | Article 661660
Smart and Hughes Fluids and Endothelial Surface Layer
microcirculatory flow (4749, 51). This is termed a lack of
hemodynamic coherence (171). Intravenous fluid delivered
rapidly during the stabilization phase may be contributing to
persistence of distur bed microcirculatory flow. When v iewing
the microcirculation using sidestream dark field microscopy
during anesthesia in pigs, fast fluid administration (20 mL/kg/hr)
for 3 h was associated with development of heterogeneity in
capillary blood flow, compared to “standard” anesthesia fluid
rates (5 mL/kg/hr) (
172). Although a direct relationship between
estimated ESL thickness and microcirculatory flow has not
been established in the clinical setting (46), it is possible that
amplification of ESL shedding caused by fast fluid administration
may be contributing to microcirculatory dysfunction. It is
possible that slower fluid administration may achieve the same
resuscitation end-points while avoiding some of these deleterious
effects on the endothelium. This concept has been explored
in a handful of studies. In a rodent model where 40% of the
blood volume was removed by atraumati c hemorrhage, rats were
assigned to receive no fluid, rapid fluid (0.9% saline at three times
shed volume over 30 min), or slow fluid (0.9% s aline at three
times shed volume over 12 h) (173). Rats in the fast fluid group
showed higher inflammatory cytokine concentrations early in
the study and some markers of worse outcome over the latter
part of the study, including lower mean arterial blood pressure,
higher blood glucose and lactate concentrations, and an increase
in markers of lung injury, compared to rats in the slow fl uid
group. As the rats were euthanased after 24 h, i t is unknown how
their recovery progressed. In a human open randomized clinical
trial, including 50 surgical and 16 septic patients, no significant
difference was found in estimated ESL thickness between patients
that received fast crystalloid administration (5–10 min) and slow
crystalloid administration (20–30 min) (
48). However, an overall
decrease in estimated ESL thickness after fluid administration
was observed. Given that there is some evidence t hat rapid fluid
administration (i.e., within 10 min) may not provide any clinical
benefit over slower fluid administration (i.e., within 20 to 60 min)
(8, 174), larger clinic al trials are justified to explore effects of fluid
rate on the microcirculation and clinical outcomes.
Choice of Clear Fluid Type
Intense ongoing debate surrounds the choice of fluid for critically
ill patients, including crystalloid, synthetic colloid, and protein-
containing solutions. These conversations surround relative fluid
effectiveness and adverse effects, many of which are still yet to
be fully elucidated for clinical relevance in certain spe ci es or
disease subsets. Adding in the potential benefit or detriment of
certain fluid types to preservation of the ESL and optimizing
microcirculation is still premature. Bearing t hat in mind, the
current evidence supports mitigating hemodilution and avoiding
hypervolemia in order to optimize ESL recovery. There is
theoretical benefit for the ESL to the use of protein-containing
solutions, such as plasma and albumin, rather than “clear” fluids,
but this must be carefully considered against any detrimental
effects of blood products, including financial cost. At this stage,
no clear recommendation can be made for veterinary medicine,
and more clinical research is required.
Adjunctive Therapies
Many drugs have been explored as to their protective or
resurrecting actions for the ESL and have been reviewed in detail
elsewhere (
175, 176). These therapies may become viable in the
future for use during the optimization and stabilization phase
of fluid resuscitation (177). Therapies that are in current use
in veterinary medicine that have been shown to have beneficial
effects on t he EG or ESL include albumin solution, plasma,
hydrocortisone, heparin, and N-acetylcysteine.
CONCLUSIONS
The coating of endothelium by the EG and its associated
molecules serves many functions in the body, both int act
and when denuded. Infusion of large volumes of fluid causes
disruption of this layer, which may propagate interstitial
edema and inflammation. There is some evidence that slow
fluid administration or restricting the volume of crystalloid
fluid for shock resuscitation may benefit the patient. E xactly
how to do this and for which patient subset this strate gy
benefits in veterinary medicine remains to be identified. Clinical
veterinary research assessing the effect of glycoprotective therapy
or strategies is currently limited by a lack of validated
EG biomarker assays and afford able, reliable technology for
estimating ESL thickness in vivo. Further work is required
on developing validated, reliable EG biomarker assays and
determining the relationship between these biomarkers and
shedding of the ESL in vivo, especially for dogs and cats.
Despite these current limitations, future research directions
should also focus on strategies limiting crystalloid fluid
volumes, especially in the setting of sepsis and early
vasopressor drug therapy.
AUTHOR CONTRIBUTIONS
LS: preparation of manuscript and final approval.
DH: revision of manuscript and final approval. Both
authors contributed to the article and approved the
submitted version.
REFERENCES
1. Angus DC, Barnato AE, Bell D, Bellomo R, Chong CR, Coats TJ, et al.
A systematic review and meta-analysis of early goal-directed therapy
for septic shock: the ARISE, ProCESS and ProMISe Investigators.
Intensive Care Med. (2015) 41:15 49 –6 0. doi: 10.1007/s00134-01
5-3822-1
2. Maitland K, Babiker A, Kiguli S, Molyneux E. The FEAST trial of
fluid bolus in African children with severe infection. Lancet. (2012)
379:613. doi: 10.1016/S014 0- 6 73 6(1 2)60 2 60 -8
3. Wiedemann HP, Wheeler AP, Bernard GR, Thompson BT,
Hayden D, deBoisblanc B, et al. Comparison of two fluid-
management strategies in acute lung injury. New Eng J Med. (2006)
354:2564–75. doi: 10.1056/NEJMoa062 2 00
Frontiers in Veterinary Science | www.frontiersin.org 9 May 2021 | Volume 8 | Article 661660
Smart and Hughes Fluids and Endothelial Surface Layer
4. Albreiki M, Voegeli D. Permissive hypotensive resuscitation in adult patients
with traumatic haemorrhagic shock: a systematic review. Eur J Trauma
Emerg Surg. (2018) 44:19 1– 20 2. doi: 10.1007/s00068-017-0862-y
5. Yozova ID, Howard J, Sigrist NE, Adamik KN. Current
trends in volume replacement therapy and the use of
synthetic colloids in small animals-an internet-based survey
2016. Front Vet Sci. (2017) 4:140. doi: 10.3389/fvets.2017.
00140
6. Laszlo I, Demeter G, Oveges N, Erces D, Kaszaki J, Tanczos K, et al.
Volume-replacement ratio for crystalloids and colloids during bleeding
and resuscitation: an animal experiment. Intensive Care Med Exp. (2017)
5:52. doi: 10.1186/s4063 5- 01 7 -0 16 5- y
7. Silverstein DC, Aldrich J, Haskins SC, Drobatz KJ, Cowgill LD.
Assessment of changes in blood volume in response to resuscitative
fluid administration in dogs. J Vet Emerg Crit Care. (2005)
15:185–92. doi: 10.1111/j.1476 -4 4 31 .20 05 .00 13 8.x
8. Ho L, Lau L, Churilov L, Riedel B, McNicol L, Hahn RG, et al.
Comparative evaluation of crystalloid resuscitation rate in a human
model of compensated haemorrhagic shock. Shock. (2016) 46:149–
57. doi: 10.1097/SHK.0000 00 00 0 00 00 61 0
9. Orbegozo Cortes D, Gamarano Barros T, Njimi H, Vincent JL.
Crystalloids versus colloids: exploring differences in fluid requirements
by systematic review and meta-regression. Anesth Analg. (2015)
120:389–402. doi: 10.1213/ANE.000 00 0 00 00 00 0 56 4
10. Tarbell JM, Pahakis MY. Mechanotransduction and the glycocalyx. J Intern
Med. (2006) 259:339–50. doi: 10.1111/j.1365-2796.2006.01620.x
11. Rosenberg RD, Shworak NW, Liu J, Schwartz JJ, Zhang L. Heparan sulfate
proteoglycans of the cardiovascular system. Specific structures emerge but
how is synthesis regulated? J Clin Invest. (1997) 100(Suppl. 11):S67–75.
12. Couchman JR. Syndecans: proteoglycan regulators of cell-sur f ace
microdomains? Nat Rev Mol Cell Biol. (2003) 4:926–37. doi: 10.1038/nrm
1257
13. Bame KJ. Heparanases: endoglycosidases that degrade heparan sulfate
proteoglycans. Glycobiology. (2001) 11:91–8r. doi: 10.1093/glycob/11.6.91R
14. Fraser JR, Laurent TC, Laurent UB. Hyaluronan: its nature,
distribution, functions and turnover. J Intern Med. (1997)
242:27–33. doi: 10.1046/j.1365 -2 7 96 .19 97 .00 17 0.x
15. Reitsma S, Slaaf DW, Vink H, van Zandvoort MA, oude Egbrink MG. The
endothelial glycocalyx: composition, functions, and visualization. Pflugers
Arch. (2007) 454:345–59. doi: 10.1007/s00424-007-0212-8
16. Henry CB, Duling BR. Permeation of the luminal capillary
glycocalyx is determined by hyaluronan. Am J Physiol. (1999)
277:H508–14. doi: 10.1152/ajpheart.1999.277.2.H508
17. Kolarova H, Ambruzova B, Svihalkova Sindlerova L, Klinke
A, Kubala L. Modulation of endothelial glycocalyx structure
under inflammatory conditions. Mediators Inflamm. (2014)
2014:694312. doi: 10.1155/2014 /6 94 31 2
18. Paulus P, Jennewein C, Zacharowski K. Biomarkers of endothelial
dysfunction: can they help us deciphering systemic inflammation and sepsis?
Biomarker. (2011) 16(Suppl. 1):S11–21. doi: 10.3109/1354750X.2011.587893
19. Bashandy GM. Implications of recent accumulating knowledge about
endothelial glycocalyx on anesthetic management. J Anesth. (2015) 29:269–
78. doi: 10.1007/s00540 - 01 4- 18 8 7- 6
20. Becker BF, Chappell D, Jacob M. Endothelial glycocalyx and coronary
vascular permeability: the fringe benefit. Basic Res Cardiol. (2010) 105:68 7–
701. doi: 10.1007/s0039 5 -0 10 -0 1 18 -z
21. Schnitzer JE, Carley WW, Palade GE. Specific albumin binding to
microvascular endothelium in culture. Am J Physiol. (1988) 254(Pt. 2):H425–
37. doi: 10.1152/ajpheart.1988.254.3.H425
22. Chang SW, Westcott JY, Henson JE, Voelkel NF. Pulmonary vascular
injury by polycations in perfused rat lungs. J Appl Physiol. (1987) 62:1932–
43. doi: 10.1152/j appl.1987 .62 .5.19 3 2
23. Antunes E, Mariano M, Cirino G, Levi S, de Nucci G. Pharmacological
characterization of polycation-induced rat hind-paw oedema. Br J
Pharmacol. (1990) 101:986–90. doi: 10.1111/j.1476-5381.1990.tb14193.x
24. Vehaskari VM, Chang CT, Stevens JK, Robson AM. The effects of polycations
on vascular permeability in the rat. A proposed role for charge sites. J Clin
Invest. (1984) 73:1053–61. doi: 10.1172/JCI111290
25. Levick JR, Michel CC. Microvascular fluid exchange and the revised Starling
principle. Cardiovasc Res. (2010) 87:198–210. doi: 10.1093/cvr/cvq062
26. Woodcock TE, Woodcock TM. Revised Starling equation and the
glycocalyx model of transvascular fluid exchange: an improved paradigm
for prescribing intravenous fluid therapy. Br J Anaesth. (2012) 108:384–
94. doi: 10.1093/bja/aer515
27. Adamson RH, Michel CC. Pathways through the intercellular clefts of frog
mesenteric capillaries. J Physiol. (1993) 466:303–27.
28. Jacob M, Bruegger D, Rehm M, S toeckelhuber M, Welsch U, Conzen P, et
al. The endothelial glycocalyx affords compatibility of Starling’s principle
and high cardiac interstitial albumin levels. Cardiovasc Res. (2007) 73:575–
86. doi: 10.1016/j.c ardiores.2006.11.021
29. Hu X, Adamson RH, Liu B, Curry FE, Weinbaum S. Starling forces that
oppose filtration after tissue oncotic pressure is increased. J Physiol Heart
Circ Physiol. (2000) 279:H1724–36. doi: 10.1152/ajpheart.2000.279.4.H1724
30. Cruz-Chu ER, Malafeev A, Pajarskas T, Pivkin IV, Koumoutsakos P.
Structure and response to flow of the glycocalyx layer. Biophys J. (2014)
106:232–43. doi: 10.1016/j.bpj.20 13 .09 .06 0
31. Dull RO, Mecham I, McJames S. Heparan sulfates mediate pressure-induced
increase in lung endothelial hydraulic conductivity via nitric oxide/reactive
oxygen species. Am J Physiol Lung Cell Mol Physiol. (2007) 292:L1452–
8. doi: 10.1152/ajplung.00376.2006
32. McDonald KK, Cooper S, Danielzak L, Leask RL. Glycocalyx degradation
induces a proinflammatory phenotype and increased leukocyte
adhesion in cultured endothelial cells under flow. PLoS ONE. (2016)
11:e0167576. doi: 10.1371/journal.pone.01675 76
33. Yao Y, Rabodzey A, Dewey CF Jr. Glycocalyx modulates the
motility and proliferative response of vascular endothelium
to fluid shear stress. Am J Physiol Heart Circ Physiol. (2007)
293:H1023–30. doi: 10.1152/ajpheart.00162.2007
34. Florian JA, Kosky JR, Ainslie K, Pang Z, Dull RO, Tarbell JM. Heparan
sulfate proteoglycan is a mechanosensor on endothelial cells. Circ Res. (2003)
93:e136–42. doi: 10.1161/01 .RES .00 00 1 01 74 4 .478 6 6.D5
35. Ebong EE, Lopez-Quintero SV, Rizzo V, Spray DC, Tarbell JM. Shear-induced
endothelial NOS activation and remodeling via heparan sulfate, glypican-1,
and syndecan-1. Integr Biol. (2014) 6:338–47. doi: 10.1039/C3IB40199E
36. Zeng Y, Waters M, Andrews A, Honarmandi P, Ebong EE, Rizzo V, et al.
Fluid shear stress induces the clustering of heparan sulfate via mobility of
glypican-1 in lipid rafts. Am J Physiol Heart Circ Physiol. (2013) 305:H811–
20. doi: 10.1152/ajpheart.00764.2012
37. Zeng Y, Tarbell JM. The adaptive remodeling of endothelial
glycocalyx in response to fluid shear stress. PLoS ONE. (2014)
9:e86249. doi: 10.1371/journal.pone.008624 9
38. Liu JX, Yan ZP, Zhang Y Y, Wu J, Liu XH, Zeng Y. Hemodynamic shear stress
regulates the transcriptional expression of heparan sulfate proteoglycans
in human umbilical vein endothelial cell. Cell Mol Biol. (2016) 62:28–34.
doi: 10.14715/cmb/2016.62.8.5
39. Lipowsky HH. The endothelial glycocalyx as a barrier to leukocyte adhesion
and its mediation by extracellular proteases. Ann Biomed Eng. (2012) 40:840–
8. doi: 10.1007/s10 4 39 -0 11 - 04 27 - x
40. Lygizos MI, Yang Y, Altmann CJ, Okamura K, Hernando AA, Perez MJ, et al.
Heparanase mediates renal dysfunction during early sepsis in mice. Physiol
Rep. (2013) 1:e00153. doi: 10.1002/phy2.153
41. Bitan M, Weiss L, Zeira M, Zcharia E, Slavin S, Nagler A, et al. Heparanase
promotes engraftment and prevents graft versus host disease in stem cell
transplantation. PLoS ONE. (2010) 5:e10135. doi: 10.1371/journal.pone.00
10135
42. Constantinescu AA, Vink H, Spaan JA. Endothelial cell glycocalyx modulates
immobilization of leukocytes at the endothelial surface. Arterioscler Thromb
Vasc Biol. (2003) 23:1541–7. doi: 10.1161/01.ATV.0000085630.24353.3D
43. Mulivor AW, Lipowsky HH. Inflammation- and ischemia-induced shedding
of venular glycocalyx. Am J Physiol Heart Circ Physiol. (2004) 286:H1672–
80. doi: 10.1152/ajpheart.00832.2003
44. Cabrales P, Vazquez BY, Tsai AG, Intaglietta M. Microvascular and capillary
perfusion following glycocalyx degradation. J Appl Physiol. (2007) 102:2251–
9. doi: 10.1152/japplphysiol.01155.2006
45. Jacob M, Bruegger D, Rehm M, Welsch U, Conzen P, Becker
BF. Contrasting effects of colloid and crystalloid resuscitation
Frontiers in Veterinary Science | www.frontiersin.org 10 May 2021 | Volume 8 | Article 661660
Smart and Hughes Fluids and Endothelial Surface Layer
fluids on cardiac vascular permeability. Anesthesiol. (2006)
104:1223–31. doi: 10.1097/0000 05 42 -2 0 06 06 0 00 -0 0 01 8
46. Rovas A, Seidel LM, Vink H, Pohlkötter T, Pavenstädt H, Ertmer
C, et al. Association of sublingual microcirculation parameters and
endothelial glycocalyx dimensions in resuscitated s epsis. Crit Care. (2019)
23:260. doi: 10.1186/s1305 4- 01 9 -2 54 2- 2
47. De Backer D, Donadello K, Sakr Y, Ospina-Tascon G, Salgado D, Scolletta
S, et al. Microcirculatory alterations in patients with severe sepsis: impact of
time of assessment and relationship with outcome. Crit Care Med. (2013)
41:791–9. doi: 10.1097/CCM.0b013e3182742e8b
48. Pouska J, Tegl V, Astapenko D, Cerny V, Lehmann C, Benes J. Impact
of intravenous fluid challenge infusion time on macrocirculation and
endothelial glycocalyx in surgical and critically ill patients. Biomed Res Int.
(2018) 2018:1–11. doi: 10.1155/2018/8925345
49. Jhanji S, Vivian-Smith A, Lucena-Amaro S, Watson D, Hinds CJ, Pearse
RM. Haemodynamic optimisation improves tissue microvascular fl ow and
oxygenation after major s urgery: a randomised controlled trial. Crit Care.
(2010) 14:R151. doi: 10.1186/cc9220
50. Trieu M, van Meurs M, van Leeuwen ALI, Van Slyke P, Hoang V,
Geeraedts LMG, et al. Vasculotide, an angiopoietin-1 mimetic, restores
microcirculatory perfus ion and microvascular leakage and decreases fluid
resuscitation requirements in hemorrhagic shock. Anesthesiology. (2018)
128:361–74. doi: 10.1097/ALN.0000 00 00 0 00 01 9 07
51. Tachon G, Harrois A, Tanaka S, Kato H, Huet O, Pottecher J, et al.
Microcirculatory alterations in traumatic hemorrhagic shock. Crit Care Med.
(2014) 42:1433–41. doi: 10.1097/CCM.0000000000000223
52. van Genderen ME, Lima A, Akkerhuis M, Bakker J, van Bommel J. Persistent
peripheral and microcirculatory perfusion alterations after out-of-hospital
cardiac arrest are associated with poor survival. Crit Care Med. (2012)
40:2287–94. doi: 10.1097/CCM.0b013e31825333b2
53. den Uil CA, L agrand WK, van der Ent M, Jewbali LS, Cheng JM, Spronk PE,
et al. Impaired microcirculation predicts poor outcome of patients with acute
myocardial infarction complicated by cardiogenic shock. Eur Heart J. (2010)
31:3032–9. doi: 10.1093/eurheartj/ehq324
54. Wrenshall LE, Stevens RB, Cerra FB, Platt JL. Modulation of macrophage
and B cell function by glycosaminoglycans. J Leukoc Biol. (1999) 66:391–
400. doi: 10.1002/jlb.66.3.39 1
55. Akbarshahi H, Axelsson JB, Said K, Malmstrom A, Fischer H,
Andersson R. TLR4 dependent heparan sulphate-induced pancreatic
inflammatory response is IRF3-mediated. J Transl Med. (2011)
9:219. doi: 10.1186/1479 -5 8 76 -9 -2 1 9
56. Scheibner KA, Lutz MA, Boodoo S, Fenton MJ, Powell JD, Horton MR.
Hyaluronan fragments act as an endogenous danger signal by engaging
TLR2. J Immunol. (2006) 177:1272–81. doi: 10.4049/jimmunol.177.2.1272
57. McKee CM, Penno MB, Cowman M, Burdick MD, Strieter RM, Bao C,
et al. Hyaluronan (HA) fragments induce chemokine gene expression in
alveolar macrophages. The role of HA size and CD44. J Clin Invest. (1996)
98:2403–13. doi: 10.1172/JCI119054
58. Lenart M, Rutkowska-Zapala M, Baj-Krzyworzeka M, Szatanek R,
Weglarczyk K, Smallie T, et al. Hyaluronan carried by tumor-derived
microvesicles induces IL-10 production in classical (CD14++CD16-)
monocytes via PI3K/Akt/mTOR-dependent signalling pathway.
Immunobiology. (2017) 222:1–10. doi: 10.1016/j.imbio.2015.06.019
59. Termeer C, Benedix F, Sleeman J, Fieber C, Voith U, Ahrens T, et al.
Oligosaccharides of Hyaluronan activate dendritic cells via toll-like receptor
4. The J Exp Med. (2002) 195:99–111. doi: 10.1084/jem.20001858
60. Webb LM, Ehrengruber MU, Clark-Lewis I, Baggiolini M, Rot A. Binding to
heparan sulfate or heparin enhances neutrophil responses to interleukin 8.
Proc Natl Acad Sci USA. (1993) 90:7158–62. doi: 10.1073/pnas.90.15.7158
61. Kainulainen V, Wang H, Schick C, Bernfield M. Syndecans, heparan sulfate
proteoglycans, maintain the proteolytic balance of acute wound fluids. J Biol
Chem. (1998) 273:11563–9. doi: 10.1074/jbc.273.19.11563
62. Itano N, Sawai T, Yoshida M, Lenas P, Yamada Y, Imagawa M, et al.
Three isoforms of mammalian hyaluronan synthases have distinct enzymatic
properties. J Biol Chem. (1999) 274:25085–92. doi: 10.1074/jbc.274.35.25085
63. Potter DR, Jiang J, Damiano ER. The recovery time course of the endothelial
cell glycocalyx in vivo and its implications in vitro. Circ Res. (2009) 104:1318–
25. doi: 10.1161/CIRCRESAHA.108.191585
64. Wang JB, Zhang YJ, Guan J, Zhou L, Sheng Y, Zhang Y, et al. Enhanced
syndecan-1 expression on neutrophils in patients with type 2 diabetes
mellitus. Acta Diabetol. (2012) 49:41–6. doi: 10.1007/s00592-011-0265-1
65. Dhodapkar MV, Sanderson R D. Syndecan-1 (CD 138) in
myeloma and lymphoid malignancies: a multifunctional
regulator of cell behavior within the tumor microenvironment.
Leuk Lymph. (1999) 34:35–43. doi: 10.3109/1042819990908
3378
66. Yeaman C, Rapraeger AC. Post-transcriptional regulation of syndecan-1
expression by cAMP in peritoneal macrophages. J Cell Biol. (1993) 122:941–
50. doi: 10.1083/jcb.122.4.941
67. Bartlett AH, Hayashida K, Park PW. Molecular and cellular mechanisms of
syndecans in tissue injury and inflammation. Mol Cells. (2007) 24:153–66.
68. Tkachenko E, Rhodes JM, Simons M. S yndecans: new
kids on the signaling block. Circ Res. (2005) 96:488–
500. doi: 10.1161/01 .RES .00 00 15 9 70 8.7 11 42 .c8
69. Townsley MI, Reed RK, Ishibashi M, Parker JC, Laurent TC, Taylor AE.
Hyaluronan efflux from canine lung with increased hydrostatic pressure
and saline loading. Am J Respir Cr i t Care Med. (1994) 150(Pt. 1):1605–
11. doi: 10.1164/ajrccm.150.6.7952622
70. Reed RK, Townsley MI, Zhao Z, Ishibashi M, Laurent TC, Taylor AE.
Lymphatic hyaluronan flux from skin increases during increased lymph
flow induced by intravenous saline loading. Int J Microcirc Clin Exp. (1994)
14:56–61. doi: 10.1159/000 17 82 0 7
71. Ostgaard G, Reed RK. Intravenous saline infusion in rat increases
hyaluronan efflux in intestinal lymph by increasing lymph flow. Acta Physiol
Scand. (1993) 147:329–35. doi: 10.1111/j.1748-1716.1993.tb09506.x
72. Gallo R, Kim C, Kokenyesi R, Scott Adzick N, Bernfield M. Syndec an-1
and4 are induced during wound repair of neonatal but not fetal skin. J
Invest Dermatol. (1996) 107:676–83.
73. Rops AL, van den Hoven MJ, Baselmans MM, Lensen JF, Wijnhoven
TJ, van den Heuvel LP, et al. Heparan sulfate domains on
cultured activated glomerular endothelial cells mediate leukocyte
trafficking. Kidney Int. (2008) 73:52–62. doi: 10.1038/sj.ki.500
2573
74. Subramanian SV, Fitzgerald ML, Bernfield M. Regulated shedding of
syndecan-1 and4 ectodomains by thrombin and growth factor receptor
activation. J Biol Chem. (1997) 272:14713–20. doi: 10.1074/jbc.272.23.14713
75. Okuyama E, Suzuki A, Murata M, Ando Y, Kato I, Takagi Y, et al. Molecular
mechanisms of syndecan-4 upregulation by TNF-alpha in the endothelium-
like EAhy926 cells. J Biochem. (2013) 154:41–50. doi: 10.1093/jb/mvt024
76. Ramnath R, Foster RR, Qiu Y, Cope G, Butler MJ, Salmon AH, et al.
Matrix metalloproteinase 9-mediated shedding of syndecan 4 in response
to tumor necrosis factor alpha: a contributor to endothelial cell glycocalyx
dysfunction. FASEB J. (2014) 28:4686–99. doi: 10.10 96 /fj.14-252221
77. Strand ME, Herum KM, Rana ZA, Skrbic B, Askevold ET, Dahl CP,
et al. Innate immune signaling induces expression and shedding of the
heparan sulfate proteoglycan syndecan-4 in cardiac fibroblasts and myocytes,
affecting inflammation in the pressure-overloaded heart. FEBS J. (2013)
280:2228–47. doi: 10.1111/febs.12161
78. Echtermeyer F, Streit M, Wilcox-Adelman S, Saoncella S, Denhez F, Detmar
M, et al. Delayed wound repair and impaired angiogenesis in mice lacking
syndecan-4. J Clin Invest. (2001) 107:R9–14. doi: 10.1172/JCI10559
79. Fitzgerald ML, Wang Z, Park PW, Murphy G, Bernfield M. Shedding of
syndecan-1 and4 ectodomains is regulated by multiple signaling pathways
and mediated by a TIMP-3-sensitive metalloproteinase. J Cell Biol. (2000)
148:811–24. doi: 10.1083/jcb.148.4.811
80. Zhang Y, Pasparakis M, Kollias G, Simons M. Myocyte-dependent regulation
of endothelial cell synde can-4 expression. Role of TNF-alpha. J Biol Chem.
(1999) 274:14786–90. doi: 10.1074/jbc.274.21.14786
81. Klein NJ, Shennan GI, Heyderman RS, Levin M. Alteration in
glycosaminoglycan metabolism and surface charge on human umbilical vein
endothelial cells induced by cytokines, endotoxin and neutrophils. J Cell Sci.
(1992) 102 (Pt. 4):821–32.
82. Mohamadzadeh M, DeGrendele H, A rizpe H, Estess P, Siegelman M.
Proinflammatory stimuli regulate endothelial hyaluronan expression and
CD44/HA-dependent primary adhesion. J Clin Invest. (1998) 101:97–
108. doi: 10.1172/JCI1604
Frontiers in Veterinary Science | www.frontiersin.org 11 May 2021 | Volume 8 | Article 661660
Smart and Hughes Fluids and Endothelial Surface Layer
83. Rouschop KM, Roelofs JJ, Claessen N, da Costa Martins P,
Zwaginga JJ, Pals ST, et al. Protection against renal ischemia
reperfusion injury by CD44 disruption. J Am Soc Nephrol. (2005)
16:2034–43. doi: 10.1681/ASN.2005010054
84. Smart L, Macdonald SPJ, Burrows S, Bosio E, Arendts G, Fatovich DM.
Endothelial g lcyoc alyx biomarkers increase in patients with infection
during Emergency Department treatment. J Crit Care. (2017) 42:304–
9. doi: 10.1016/j.jcrc.2017.07.001
85. Smart L, Bosio E, Macdonald SPJ, Dull R, Fatovich DM, Neil C, et al.
Glycocalyx biomarker syndec an-1 is a stronger predictor of respiratory
failure in patients with sepsis due to pneumonia, compared to endocan. J
Crit Care. (2018) 47:93–8. doi: 10.1016/j.jcrc.2018.06.015
86. Smart L, Macdonald SPJ, Bosio E, Fatovich D, Neil C, Arendts G. Bolus
therapy with 3% hypertonic saline or 0.9% saline in emergency department
patients with suspected sepsis: a pilot randomised controlled trial. J Crit Care.
(2019) 52:33–9. doi: 10.1016/j.jcrc.2019.03.009
87. Ebong EE, Macaluso FP, Spray DC, Tarbell JM. Imaging the endothelial
glycocalyx in vitro by rapid freezing/freeze substitution transmission
electron microscopy. Arterioscler Thromb Vasc Biol. (2011) 31:1908–
15. doi: 10.1161/ATVBAHA.111.225268
88. Bruegger D, Jacob M, Rehm M, Loetsch M, Welsch U, Conzen P, et al. Atrial
natriuretic peptide induces shedding of endothelial glycocalyx in coronary
vascular bed of guinea pig hearts. Am J Physiol Heart Circ Physiol. (2005)
289:H1993–9. doi: 10.1152/ajpheart.00218.2005
89. Chappell D, Jacob M, Paul O, Rehm M, Welsch U, Stoeckelhuber M,
et al. The glycocalyx of the human umbilical vein endothelial cell: an
impressive structure ex vivo but not in culture. Circ Res. (2009) 104:1313–
7. doi: 10.1161/CIRCRESAHA.108.187831
90. Chappell D, Hofmann-Kiefer K, Jacob M, Rehm M, Briegel J, Welsch U, et
al. TNF-alpha induced shedding of the endothelial glycocalyx is prevented
by hydrocortisone and antithrombin. Basic Res Cardiol. (2009) 104:78–
89. doi: 10.1007/s00395 - 00 8- 07 4 9- 5
91. Xia Y, Fu BM. Investigation of endothelial surface glycocalyx components
and ultrastructure by single molecule localization microscopy: stochastic
optical reconstruction microscopy (STORM). Yale J Biol Med. (2018)
91:257–66.
92. Mukai S, Takaki T, Nagumo T, Sano M, Kang D, Takimoto M, et
al. Three-dimensional electron microscopy for endothelial glycocalyx
observation using Alcian blue with silver enhancement. Med Mol Morphol.
(2020). doi: 10.1007/s0079 5 -0 20 -0 0 26 7- 1 . [Epub ahead of print].
93. van den Berg BM, Spaan JA, Vink H. Impaired glycocalyx barrier properties
contribute to enhanced intimal low-density lipoprotein accumulation at
the carotid artery bifurcation in mice. Pflugers Archiv. (2009) 457:1199–
206. doi: 10.1007/s0042 4 -0 08 -0 5 90 -6
94. Long DS, Smith ML, Pries AR, Ley K, Damiano ER. Microviscometry reveals
reduced blood viscosity and altered shear rate and shear stress profiles in
microvessels after hemodilution. Proc Natl Acad Sci USA. (2004) 101:10060–
5. doi: 10.1073/pnas.04029371 0 1
95. Vink H, Duling BR. Identification of distinct luminal domains for
macromolecules, erythrocytes, and leukocytes within mammalian capillaries.
Circ Res. (1996) 79:581–9. doi: 10.1161/01.RES.79.3.581
96. Kataoka H, Ushiyama A, Kawakami H, Akimoto Y, Matsubara S, Iijima
T. Fluorescent imaging of endothelial glycocalyx layer with wheat germ
agglutinin using intravital microscopy. Microsc Res Tech. (2016) 79:31–
7. doi: 10.1002/jemt.2260 2
97. Donati A, Damiani E, Domizi R, Romano R, Adrario E, Pelaia P, et al.
Alteration of the sublingual microvascular glycocalyx in critically ill patients.
Microvasc Res. (2013) 90:86–9. doi: 10.1016/j.mvr.2013.08.007
98. Rovas A, Lukasz AH, Vink H, Urban M, Sackarnd J, Pavenstädt H, et al.
Bedside analysis of the sublingual microvascular glycocalyx in the emergency
room and intensive care unit - the GlycoNurse study. Scand J Trauma Resusc
Emerg Med. (2018) 26:16. doi: 10.1186/s13049-018-0483-4
99. Cerny V, Astapenko D, Burkovskiy I, Hyspler R, Ticha A, Trevors MA,
et al. Glycocalyx in vivo measurement. Clin Hemorheol Microcirc. (2017)
67:499–503. doi: 10.3233/CH-17923 5
100. Gommeren K, Allerton FJ, Morin E, Reynaud A, Peeters D, Silverstein
DC. Evaluation of a rapid bedside scoring system for microcirculation
videos acquired from dogs. J Vet Emerg Crit Care. (201 4) 24:554–
61. doi: 10.1111/vec.12212
101. Henry CB, Duling BR. TNF-alpha increases entry of macromolecules into
luminal endothelial cell glycocalyx. Am J Physiol Heart Circ Physiol. (2000)
279:H2815–23. doi: 10.1152/ajpheart.2000.279.6.H2815
102. Kurzelewski M, Czarnowska E, Beresewicz A. Superoxide- and nitric oxide-
derived species mediate endothelial dysfunction, endothelial glycocalyx
disruption, and enhanced neutrophil adhesion in the post-ischemic guinea-
pig heart. J Physiol Pharmacol. (2005) 56:163–78.
103. Chappell D, Jacob M, Hofmann-Kiefer K, Rehm M, Welsch U,
Conzen P, et al. Antit hrombin reduces shedding of the endothelial
glycocalyx following ischaemia/reperfusion. Cardiovasc Res. (2009)
83:388–96. doi: 10.1093/cvr/cvp097
104. Singh A, Ramnath RD, Foster RR, Wylie EC, Friden V,
Dasgupta I, et al. Reactive oxygen species modulate the barrier
function of the human glomerular endothelial glycocalyx.
PLoS ONE. (2013) 8:e55852. doi: 10.1371/journal.pone.00
55852
105. Schmidt A, Echtermeyer F, Alozie A, Brands K, Buddecke E . Plasmin-
and thrombin-accelerated shedding of syndecan-4 ectodomain
generates cleavage sites at Lys(114)-Arg(115) and Lys(129)-Val(130)
bonds. J Biol Chem. (2005) 280:34441–6. doi: 10.1074/jbc.M5019
03200
106. Strand ME, Aronsen JM, Braathen B, Sjaastad I, Kvaloy H, Tonnessen T, et
al. Shedding of syndecan-4 promotes immune cell recruitment and mitigates
cardiac dysfunction after lipopolysaccharide challenge in mice. J Mol Cell
Cardiol. (2015) 88:133–44. doi: 10.1016/j.yjmcc.2015.10.003
107. Colburn P, Kobayashi E, Buonassisi V . Depleted level of heparan sulfate
proteoglycan in the extracellular matrix of endothelial cell cultures exposed
to endotoxin. J Cell Physiol. (1994) 159:121–30. doi: 10 .100 2 /jcp.1041590116
108. Mulivor AW, Lipowsky HH. Inhibition of glycan shedding and
leukocyte-endothelial adhesion in postcapillary venules by suppression
of matrixmetalloprotease activity with doxycycline. Microcirculation. (2009)
16:657–66. doi: 10.3109/10 73 96 8 09 03 1 33 71 4
109. Kozar RA, Peng Z, Zhang R, Holcomb JB, Pati S, Park P, et
al. Plasma restoration of endothelial glycocalyx in a rodent
model of hemorrhagic shock. Anesth Analg. (2011) 112:1289–
95. doi: 10.1213/ANE.0b0 1 3e3 1 82 10 3 85 c
110. Peng Z, Pati S, Potter D, Brown R, Holcomb JB, Grill R, et al. Fresh frozen
plasma lessens pulmonary endothelial inflammation and hyperperme ability
after hemorrhagic shock and is associated with loss of syndecan 1. Shock.
(2013) 40:195–202. doi: 10.1097/SHK.0b013e31829f91fc
111. Smart L, Boyd CJ, Claus MA, Bosio E, Hosgood G, Raisis A. Large-volume
crystalloid fluid is associated with increased hyaluronan shedding and
inflammation in a canine hemorrhagic shock model. Inflammation. (2018)
41:1515–23. doi: 10.1007/s107 5 3- 01 8- 0 79 7- 4
112. Marechal X, Favory R, Joulin O, Montaigne D, Hassoun S, Decoster
B, et al. Endothelial glycocalyx damage during endotoxemia
coincides with microcirculatory dysfunction and vas cular oxid ative
stress. Shock. (2008) 29:572–6. doi: 10.1097/SHK.0b013 e3 18 1 5
7e926
113. Naumann DN, Hazeldine J, Davies DJ, Bishop J, Midwinter MJ, Belli
A, et al. Endotheliopathy of trauma is an on-scene phenomenon, and
is associated with multiple organ dysfunction syndrome: a prospective
observational study. Shock. (2018) 49:420–8. doi: 10.1097/SHK.00000000000
00999
114. Tapking C, Hernekamp JF, Horter J, Kneser U, Haug V, Vogelpohl
J, et al. Influence of burn severity on endothelial glycocalyx shedding
following thermal trauma: a prospective observational study. Burns.
(2020). doi: 10.1016/j.burns.2 02 0.0 7.02 1 . [Epub ahead of print].
115. Hippensteel JA, Uchimido R, Tyler PD, Burke RC, Han X,
Zhang F, et al. Intravenous fluid resuscitation is associated
with septic endothelial glycocalyx degradation. Crit Care. (2019)
23:259. doi: 10.1186/s13 05 4- 0 19 -2 53 4 -2
116. Pries AR, Se comb TW, Sperandio M, Gaehtgens P. Blood flow resistance
during hemodilution: effect of plasma composition. Cardiovasc Res. (1998)
37:225–35. doi: 10.1016/S0 00 8- 63 6 3(9 7)00 2 26 -5
Frontiers in Veterinary Science | www.frontiersin.org 12 May 2021 | Volume 8 | Article 661660
Smart and Hughes Fluids and Endothelial Surface Layer
117. Chappell D, Jacob M, Rehm M, Stoeckelhuber M, Welsch U, Conzen P,
et al. Heparinase selectively sheds heparan sulphate from the endothelial
glycocalyx. Biol Chem. (2008) 389:79–82. doi: 10.1515/BC.2008.005
118. Huxley VH, Curry FE. Albumin modulation of capillary permeability:
test of an adsorption mechanism. Am J Physiol. (1985) 248(Pt. 2):H264–
73. doi: 10.1152/ajpheart.1985.248.2.H264
119. Huxley VH, Curry FE. Differential actions of albumin and plasma on
capillary solute permeability. Am J Physiol. (1991) 2 60 (Pt. 2):H1645–
54. doi: 10.1152/ajpheart.1991.260.5.H1645
120. Torres LN, Sondeen JL, Dubick MA, Filho IT. Systemic and microvascular
effects of resuscitation with blood products after severe hemorrhage in rats.
J Trauma Acute Care Surg. (2014) 77:716–23. doi: 10.1097/TA.00000000000
00448
121. D’Souza SP, Davis M, Baxter GF. Autocrine and paracrine actions
of natriuretic peptides in the heart. Pharmacol Ther. (2004) 101:113–
29. doi: 10.1016/j.pharmthera.2003.11.00 1
122. Chen W, Gassner B, Borner S, Nikolaev VO, Schlegel N, Waschke J, et al.
Atrial natriuretic peptide enhances microvascular albumin permeability by
the caveolae-mediated transcellular pathway. Cardiovasc Res. (2012) 9 3 :141 –
51. doi: 10.1093/cvr/cvr279
123. Zimmerman RS, Trippodo NC, MacPhee AA, Martinez AJ, Barbee RW.
High-dose atrial natriuretic factor enhances albumin escape from the
systemic but not the pulmonary circulation. Circ Res. (1990) 67:461–
8. doi: 10.1161/01.RES .67 .2.46 1
124. Jacob M, Saller T, Chappell D, Rehm M, Welsch U, Becker BF. Physiological
levels of A-, B- and C-type natriuretic peptide shed the endothelial
glycocalyx and enhance vascular permeability. Basic Res Cardiol. (2013)
108:347. doi: 10.1007/s003 95 -0 1 3- 03 47 - z
125. Chappell D, Bruegge r D, Potzel J, Jacob M, Brettner F, Vogeser M,
et al. Hypervolemia increases release of atrial natriuretic peptide
and shedding of the endothelial glycocalyx. Crit Care. (2014)
18:538. doi: 10.1186/s1305 4- 01 4 -0 53 8- 5
126. Belavi
´
c M, Sotošek TokmadŽi
´
c V, Fiši
´
c E, Brozovi
´
c Krijan A,
Striki
´
c N, Lon
ˇ
cari
´
c Katušin M, et al. The effect of various doses
of infusion solutions on the endothelial glycocalyx layer in
laparoscopic cholecystectomy patients. Minerva Anestesiol. (2018)
84:1032–43. doi: 10.23736/S037 5- 93 93 .18 .12 15 0 -X
127. Bruegger D, Schwartz L, Chappell D, Jacob M, Rehm M, Vogeser M,
et al. Release of atrial natriuretic peptide precedes shedding of the
endothelial glycocalyx equally in patients undergoing on- and off-pump
coronary artery bypass surgery. Basic Res Cardiol. (2011) 106:1111–
21. doi: 10.1007/s00395 - 01 1- 02 0 3- y
128. Nemme J, Krizhanovskii C, Ntika S, Sabelnikovs O, Vanags I, Hahn
RG. Hypervolemia does not cause degradation of the endothelial
glycocalyx layer during open hysterectomy performed under
sevoflurane or propofol anesthesia. Acta Anaesthesiol Scand. (2020)
64:538–45. doi: 10.1111/aas.13511
129. Hasselgren E, Zdolsek M, Zdolsek JH, Björne H, Krizhanovskii C, Ntika
S, et al. Long intravascular persistence of 20% albumin in postoperative
patients. Anesth Analg. (2019) 129:1232–9. doi: 10.1213/ANE.00000000000
04047
130. Torres Filho IP, Torres LN, Salgado C, Dubick MA. Plasma synde can-1
and heparan sulfate correlate with microvascular glycocalyx deg rad ation in
hemorrhaged rats after different resuscitation fluids. Am J Physiol Heart Circ
Physiol. (2016) 310:H1468–78. doi: 10.1152/ajpheart.00006.2016
131. Torres LN, Sondeen JL, Ji L, Dubick MA, Torres Filho I. Evaluation
of resuscitation fluids on endothelial glycocalyx, venular blood flow, and
coagulation function after hemorrhagic shock in rats. J Trauma Acute Care
Surg. (2013) 75:759–66. doi: 10.1097/TA.0b013e3182a92514
132. Torres LN, Chung KK, Salgado CL, Dubick MA , Torres Filho IP.
Low-volume resuscitation with normal saline is associated with
microvascular endothelial dysfunction after hemorrhage in rats,
compared to colloids and balanced crystalloids. Crit Care. (2017)
21:160. doi: 10.1186/s1305 4- 01 7 -1 74 5- 7
133. Nelson A, Statkevicius S, Schott 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) 4:6. doi: 10.1186/s40635-016-0080-7
134. Aksu U, Bezemer R, Yavuz B, Kandil A , Demirci C, Ince C.
Balanced vs unbalanced crystalloid resuscitation in a near-
fatal model of hemorrhagic shock and the effects on renal
oxygenation, oxidative stress, and inflammation. Resuscitation. (2012)
83:767–73. doi: 10.1016/j.resuscitation.2011.11.022
135. Sillesen M, Rasmussen LS, Jin G, Jepsen CH, Imam A, Hwabejire
JO, et al. Assessment of coagulopathy, endothelial injury, and
inflammation after traumatic brain injury and hemorrhage in a
porcine model. J Trauma Acute Care Surg. (2014) 76:12–9; discussion
9–20. doi: 10.1097/TA.0b013e3182aaa675
136. Annecke T, Rehm M, Bruegger D, Kubitz JC, Kemming GI, Stoe ck elhuber
M, et al. Ischemia-reperfusion-induced unmeasured anion generation and
glycocalyx shedding: sevoflurane versus propofol anesthesia. J Invest Surg.
(2012) 25:162–8. doi: 10.3109/08941939.2011.618524
137. Byrne L, Obonyo NG, Diab SD, Dunster KR, Passmore MR, Boon AC,
et al. Unintended consequences: fluid resus citation worsens shock in an
ovine model of endotoxemia. Am J Respir Crit Care Med. (2018) 198:1043–
54. doi: 10.1164/rccm.201801-0064OC
138. Hofbauer R, Moser D, Hornykewycz S, Frass M, Kapiotis
S. Hydroxyethyl starch reduces the chemotaxis of white
cells through endothelial cell monolayers. Transfusion. (1999)
39:289–94. doi: 10.1046/j.15 37 - 29 95 .19 99 .39 39 9 21 92 86 .x
139. Wang P, Li Y, Li J. Hydroxyethyl starch 130/0.4 prevents the early
pulmonary inflammatory response and oxidative stress after hemorrhagic
shock and resuscitation in rats. Int Immunopharmacol. (2009) 9:347–
53. doi: 10.1016/j.intimp.2008.12.0 14
140. Pascual JL, Ferri L E, C haud hury P, S eely AJ, Campisi G, Giannias B, et al.
Hemorrhagic shock resuscitation with a low molecular weight starch reduces
neutrophil-endothelial interactions and vessel leakage in vivo. Surg Infect.
(2001) 2:275–87; discussion 87–8. doi: 10.1089/10962960152 8 13 31 3
141. Seo EH, Park HJ, Piao LY, Lee JY, Oh CS, Kim SH. Immune response in fluid
therapy with crystalloids of different ratios or colloid for rats in haemorrhagic
shock. Sci Rep. (2020) 10:8067. doi: 10.1 03 8/ s4 15 98 - 02 0- 65 0 63 -4
142. Ozturk T, Onur E, Cerrahoglu M, Calgan M, Nizamoglu F, Civi M.
Immune and inflammatory role of hydroxyethyl starch 130/0.4 and fluid
gelatin in patients undergoing coronary surgery. Cytokine. (2015) 74:69–
75. doi: 10.1016/j.cyto.2014.10 .00 2
143. Thiel M, Buessecker F, Eberhardt K, Chouker A, Setzer F, Kreimeier U, et
al. Effects of hypertonic saline on expression of human polymorphonuclear
leukocyte adhesion molecules. J Leuk Biol. (2001) 70:261–73.
doi: 10.1189/jlb.70.2.261
144. Ciesla DJ, Moore EE, Musters RJ, Biffl WL, Silliman CA. Hypertonic
saline alteration of the PMN cytoskeleton: implications for signal
transduction and the cytotoxic response. J Trauma. (2001) 50:206–
12. doi: 10.1097/000 05 37 3 -2 00 10 2 00 0- 0 00 04
145. Banerjee A, Moore EE, McLaughlin NJ, Lee L, Jones WL, Johnson JL,
et al. Hyperosmolarity attenuates TNF-alpha-mediated proinflammatory
activation of human pulmonary microvascular endothelial cells. Shock.
(2013) 39:366–72. doi: 10.1097/SHK.0b013e3182894016
146. Junger WG, Hoyt DB, Davis RE, Herdon-Remelius C, Namiki S, Junger H, et
al. Hypertonicity regulates the function of human neutrophils by modulating
chemoattractant receptor signaling and activating mitogen-activated protein
kinase p38. J Clin Invest. (1998) 101:2768–79. doi: 10.1172/JCI1354
147. Rizoli SB, Kapus A, Fan J, Li YH, Marshall JC, Rotstein OD.
Immunomodulatory effects of hypertonic resuscitation on the development
of lung inflammation following hemorrhagic shock. J Immunol.
(1998) 161:6288–96.
148. Ciesla DJ, Moore EE, Zallen G, Biffl WL, Silliman CC.
Hypertonic saline attenuation of polymorphonuclear neutrophil
cytotoxicity: timing is everything. J Trauma. (20 00 ) 48:388–
95. doi: 10.1097/000 05 37 3 -2 00 00 3 00 0- 0 00 04
149. Angle N, Hoyt DB, Cabello-Passini R, Herdon-Remelius C, Loomis W,
Junger WG. Hypertonic saline resuscitation reduces neutrophil margination
by suppressing neutrophil L selectin expression. J Trauma. (1998) 45:7–12.
150. Gonzalez RJ, Moore EE, Ciesla DJ, Neto JR, Biffl WL,
Silliman CC. Hyperosmolarity abrogates neutrophil cytotoxicity
provoked by post-shock mesenteric lymph. Shock. (2002)
18:29–32. doi: 10.1097/000 24 38 2 -2 00 20 7 00 0- 0 00 06
Frontiers in Veterinary Science | www.frontiersin.org 13 May 2021 | Volume 8 | Article 661660
Smart and Hughes Fluids and Endothelial Surface Layer
151. Pascual JL, Ferri LE, Seely AJ, Campisi G, Chaudhury P, Giannias B, et al.
Hypertonic saline resuscitation of hemorrhagic shock diminishes neutrophil
rolling and adherence to endothelium and reduces in vivo vascular
leakage. Ann Surg. (2002) 236:634–42. doi: 10.1097/0 0 00 06 5 8- 20 02 1 10 0
0-00014
152. Theobaldo MC, Barbeiro HV, Barbeiro DF, Petroni R, Soriano FG.
Hypertonic saline solution reduces the inflammatory response in
endotoxemic rats. Clinics. (2012) 67:1463–8. doi: 10.6061/clinics/20
12(12)18
153. Petroni RC, Biselli PJ, de Lima TM, Theobaldo MC, C aldini ET, Pimentel
RN, et al. Hypertonic saline (NaCl 7.5%) reduces LPS-induced acute lung
injury in rats. Inflammation. (2015) 38:2026–35. doi: 10.1007/s10753-01
5-0183-4
154. Yu G, Chi X, Hei Z, Shen N, Chen J, Zhang W, et al. Small volume
resuscitation with 7.5% hypertonic saline, hydroxyethyl starch 130/0.4
solution and hypertonic sodium chloride hydroxyethyl starch 40 injection
reduced lung injury in endotoxin shock rats: comparison with saline. Pulm
Pharmacol Ther. (2012) 25 :27 –3 2. doi: 10.1016/j.pupt.2011.10.003
155. Tan GK, Ng JK, Tan KW, Angeli V, Moochhala S, Ooi EE, et
al. Hypertonic saline reduces vascular leakage in a mouse model of
severe dengue. PloS ONE. (2013) 8:e61621. doi: 10.1371/journal.pone.00
61621
156. Sun YX, Wu XS, Gao Z, Wang F, Liu S, Chen XL. Effe ct of 200 mEq/L
Na+ hypertonic saline resuscitation on systemic inflammatory response
and oxidative stress in severely burned rats. J Surg Res. (2013) 185:477–
84. doi: 10.1016/j.jss.2013.06.039
157. Junger WG, Rhind SG, Rizoli SB, Cuschieri J, Shiu MY, Baker AJ,
et al. Resuscitation of traumatic hemorrhagic shock patients with
hypertonic saline-without dextran-inhibits neutrophil and endothelial cell
activation. Shock. (2012) 38:341–50. doi: 10.1097/SHK.0b013e3182635
aca
158. Rhind SG, Crnko NT, Baker AJ, Morrison LJ, Shek PN, Scarpelini S,
et al. Prehospital resuscitation with hypertonic saline-dextran modulates
inflammatory, coagulation and endothelial activation marker profiles in
severe traumatic brain injured patients. J Neuroinflammation. (2010)
7:5. doi: 10.1186/1742 -2 0 94 -7 -5
159. de Laforcade A, Silverstein DC. Shock. In: Silverstein DC, Hopper K, editors.
Small Animal Critical Care Medicine. 2nd ed. St. Louis, MI: Elsevier Saunders
(2015). p. 26–30.
160. Boyd CJ, Smart L. Hypovolemic shock. In: Drobatz KJ, Hopper K, Rozanski
E, Silverstein D C, editors. T extbook of Small Animal Emergency Medicine.
2nd ed. Hoboken, NJ: John Wiley & Sons (2018). p. 986–92.
161. Marik PE, Byrne L, van Haren F. Fluid resuscitation in sepsis:
the great 30 mL pe r kg hoax. J Thor Dis. (2020) 12(Suppl.
1):S37–47. doi: 10.21037/jtd.2019 .12 .84
162. Macdonald SPJ, Taylor DM, Keijzers G, Arendts G, Fatovich DM, Kinnear
FB, et al. REstricted Fluid REsuscitation in Sepsis-associated Hypotension
(REFRESH): study protocol for a pilot randomised controlled trial. Trials.
(2017) 18:399. doi: 10.1186/s13063-017-2137-7
163. Macdonald SPJ, Keijzers G, Taylor DM, Kinnear F, Arendts G, Fatovich DM,
et al. Restricted fluid resuscitation in suspe cted sepsis associated hypotension
(REFRESH): a pilot randomised controlled trial. Intensive Care Med. (2018)
44:2070–8. doi: 10.1007/s0013 4 -0 18 -5 4 33 -0
164. Self WH, Semler MW, Bellomo R, Brown SM, deBoisblanc BP, Exline MC,
et al. Liberal versus restrictive intravenous fluid therapy for early septic
shock: rationale for a randomized trial. Ann Emerg Med. (2018) 72:457–
66. doi: 10.1016/j.annemergmed.2018.03.039
165. Bickell WH, Wall MJ Jr., Pepe PE, Martin RR, Ginger VF, et al.
Immediate versus delayed fluid resuscitation for hypotensive patients
with penetrating torso injuries. New Eng J Med. (1994) 331:1105–
9. doi: 10.1056/NEJM199 41 0 27 33 1 17 01
166. Mapstone J, Roberts I, Evans P. Fluid resuscitation strategies:
a systematic review of animal trials. J Trauma. (2003) 55:571–
89. doi: 10.1097/01.TA.000006296 8.6 98 67 .6F
167. Safiejko K, Smereka J, Filipiak KJ, Szarpak A, Dabrowski M, Ladny JR, et
al. Effectiveness and safety of hypotension fluid resuscitation in traumatic
hemorrhagic shock: a systematic review and meta-analysis of randomized
controlled trials. Cardiol J. (2020). doi: 10.5603/CJ.a2020.0096. [Epub ahead
of print].
168. Wang CH, Hsieh WH, Chou HC, Huang YS, Shen JH, Yeo YH, et al. Liberal
versus restricted fluid resuscitation strategies in trauma patients: a systematic
review and meta-analysis of randomized controlled trials and observational
studies
. Crit Care Med. (2014) 42:954–61. doi: 10.1097/CCM.000000000 0 00
0050
169. Kwan I, Bunn F, Chinnock P, Roberts I. Timing and volume of fluid
administration for patients with bleeding. Cochrane Database S yst Rev.
(2014) 2014:Cd002245. doi: 10.1002/14651858.CD002245.pub2
170. Myles PS, Bellomo R, Corcoran T, Forbes A, Peyton P, Story D,
et al. Restrictive versus liberal fluid therapy for major abdominal
surgery. New Eng J Med. (2018) 378:2263–74. doi: 10.1056/NEJMoa18
01601
171. Chow RS. Terms, definitions, nomenclature and routes of fluid
administration. Front Vet Sci. (2021) 7:591218. doi: 10.3389/fvets.2020.59
1218
172. Turek Z, Cerny V, Parizkova R, Samek J, Oberreiter M. Prolonged
hypervolemic hemodilution decreases functional capillary density
of ileal mucosa in pigs revealed by sidestream dark-field imaging.
J Zhejiang Univ Sci B. (2011) 12:867–74. doi: 10.1631/jzus.B10
00385
173. Yu TC, Yang FL, Hsu BG, Wu WT, Chen SC, Lee RP, et al. Deleterious
effects of aggressive rapid crystalloid resuscitation on treatment of
hyperinflammatory response and lung injury induced by hemorrhage
in aging rats. J Surg Res. (2014) 187:587–95. doi: 10.1016/j.jss.2013.
10.061
174. Iro MA, Sell T, Brown N, Maitland K. Rapid intravenous rehydration of
children with acute gastroenteritis and dehydration: a systematic review
and meta-analysis. BMC Pediatr. (2018) 18:44. doi: 10.1186/s12887-018-
1006-1
175. Gaudette S, Hughes D, Boller M. The endothelial glycocalyx:
structure and function in health and critical illness. J
Vet Emerg Cr i t Care. (2020) 30:117–34. doi: 10.1111/ve
c.12925
176. Weinbaum S, Cancel LM, Fu BM, Tarbell JM. The glycocalyx
and its role in vascular physiology and vascular related diseases.
Cardiovasc Eng Technol. (2020) 12:37–71. doi: 10.1007/s13239-020-0
0485-9
177. Malbrain M, Langer T, Annane D, Gattinoni L, Elbers P, Hahn
RG, et al. Intravenous fluid therapy in the perioperative and critical
care setting: executive summary of the International Fluid Academy
(IFA). Ann Intensive Care. (2020) 10:64. doi: 10.1186/s13613-020-0
0679-3
Conflict of Interest: The authors declare that the research was conducted in the
absence of any commercial or financial relationships that could be construed as a
potential conflict of interest.
Copyright © 2021 Smart and Hughes. This is an open-access ar ticle distributed
under the terms of the Creative Commons Attribution License (CC BY). The use,
distribution or reproduction in other forums is permitted, provided the original
author(s) and the copyright owner(s) are credited and that the original publication
in this journal is cited, in accordance with accepted academic practice. No use,
distribution or reproduction is permitted which d oes not comply with these terms.
Frontiers in Veterinary Science | www.frontiersin.org 14 May 2021 | Volume 8 | Article 661660