Skip to main content
Lippincott Open Access logoLink to Lippincott Open Access
. 2018 Apr 5;38(7):1427–1439. doi: 10.1161/ATVBAHA.118.310839

Endothelial Glycocalyx as a Shield Against Diabetic Vascular Complications

Involvement of Hyaluronan and Hyaluronidases

Sophie Dogné 1,, Bruno Flamion 1, Nathalie Caron 1
PMCID: PMC6039403  PMID: 29880486

Abstract

The endothelial glycocalyx (EG), which covers the apical surface of the endothelial cells and floats into the lumen of the vessels, is a key player in vascular integrity and cardiovascular homeostasis. The EG is composed of PGs (proteoglycans), glycoproteins, glycolipids, and glycosaminoglycans, in particular hyaluronan (HA). HA seems to be implicated in most of the functions described for EG such as creating a space between blood and the endothelium, controlling vessel permeability, restricting leukocyte and platelet adhesion, and allowing an appropriate endothelial response to flow variation through mechanosensing. The amount of HA in the EG may be regulated by HYAL (hyaluronidase) 1, the most active somatic hyaluronidase. HYAL1 seems enriched in endothelial cells through endocytosis from the bloodstream. The role of the other main somatic hyaluronidase, HYAL2, in the EG is uncertain. Damage to the EG, accompanied by shedding of one or more of its components, is an early sign of various pathologies including diabetes mellitus. Shedding increases the blood or plasma concentration of several EG components, such as HA, heparan sulfate, and syndecan. The plasma levels of these molecules can then be used as sensitive markers of EG degradation. This has been shown in type 1 and type 2 diabetic patients. Recent experimental studies suggest that preserving the size and amount of EG HA in the face of diabetic insults could be a useful novel therapeutic strategy to slow diabetic complications. One way to achieve this goal, as suggested by a murine model of HYAL1 deficiency, may be to inhibit the function of HYAL1. The same approach may succeed in other pathological situations involving endothelial dysfunction and EG damage.

Keywords: cardiovascular disease, diabetes mellitus, endothelial cells, glycocalyx, hyaluronic acid, permeability

Overview of the Endothelial Glycocalyx

Endothelial Glycocalyx: A Unique Extracellular Matrix

Eukaryotic cells are endowed with a complex external layer of PGs (proteoglycans), glycoproteins, and glycolipids called extracellular matrix (ECM), which regulates outside-in signaling, protects the cell from exterior aggression, and maintains tissue integrity. The composition and thickness of this ECM vary according to cell types. In the vascular system, a polymeric sugar-rich network covers the apical surface of the endothelial cells (ECs), in a structure that floats into the lumen of the vessels. This particular ECM, commonly called endothelial glycocalyx (EG), was observed for the first time in 1966 in capillaries of rat intestinal mucosa using electron microscopy combined with ruthenium red staining.1 Other more recent methods have shown this structure in several types of vessels and estimated its structural or functional thickness to anything between 0.5 and 5 μm, depending on the method and the vessel caliber. The methods most often used probe the EG accessibility to macromolecules, red blood cells, or white blood cells; analyze the velocity of microparticles near the vessel wall; or directly observe the structure through labeling.28

Despite uncertainty about its size and access properties, the importance of the EG in vascular integrity and cardiovascular homeostasis has progressively been recognized.7,913 Some of its essential roles are creating a space between blood and the endothelium; controlling vessel permeability through withholding or slowing down protein and macromolecule passage, which leads to regulation of water efflux; restricting leukocyte and platelet adhesion to the endothelium, thus moderating inflammation and thrombosis; and allowing an appropriate EC response to flow variation through mechanosensing. These functions are described in more detail in this review before turning to the damaging effects of diabetes mellitus on the EG.

EG Composition and Homeostasis

The term EG is often used to describe a dense—although nonuniform—layer adjacent to ECs, particularly when observed in vitro. In vivo, the loose luminal layer covering the endothelium is sometimes called endothelial surface layer.14 In this review, the term EG will be used to describe the structure of this coat, without distinguishing the different layers and without stressing any difference between the in vitro and in vivo situations.

The EG is a porous, hair-like, regularly organized layer composed of PGs, glycoproteins, glycolipids, and glycosaminoglycans (GAGs).14,15 The main PGs in the EG are sdc (syndecan)-1, -2 and -4 and gpc1 (glypican-1), all firmly bound to the cell plasma membrane. Syndecans are transmembrane proteins, whereas glypicans stick to lipid rafts via a glycosylphosphatidylinositol anchor.16 Other endothelial PGs such as mimecan and perlecan are soluble, secreted proteins present in both EG and blood.17

Various GAGs fit in the EG: heparan sulfate (HS), chondroitin sulfate, hyaluronan (HA), and to a lesser extent dermatan sulfate and keratan sulfate. At physiological pH, the EG is negatively charged. HS, covalently bound to proteins, is the most abundant GAG in the EG, with an average content of 50% to 90% of all GAGs. Chondroitin sulfate and dermatan sulfate, also covalently linked to proteins, are the second GAGs in abundance. HA, the only nonsulfated GAG in the EG, is less abundant and does not covalently bind to proteins. However, contrary to other GAGs, HA has a very large degree of polymerization so it can reach a molecular weight >1 million Dalton. HA incorporates deeply inside the EG, in the denser layer adjacent to the ECs.9,14 Various investigations using enzymatic treatments have confirmed the nonuniform distribution of GAGs within the EG. Actually, the impact of GAG-degrading enzymes on EG thickness is highly variable because of the nature of the vessel, the method used to analyze EG thickness, and the treatment itself (amount and duration of exposure). For example, bacterial heparinase (which degrades HS) injected into rat mesenteric postcapillary venules, and analyzed using intravital microscopy, induces a 43% decrease in EG thickness, whereas chondroitinase and hyaluronidase can decrease EG thickness by 34% and 26%, respectively.14 The results variability is particularly high for testicular hyaluronidase, which induces a decrease of EG thickness of 58% in rat myocardial capillaries, when observed through electronic microscopy3 and of 25% in mouse carotid arteries when observed using 2 photon laser scanning microscopy.18 In vitro, hyaluronidase produces a mere 15% decrease in EG thickness of bovine lung microvascular ECs measured by atomic force microscopy.19 The in vitro versus in vivo compositions of the EG may differ.

Plasma proteins such as albumin, orosomucoid, antithrombin III, extracellular superoxide dismutase, lipases, growth factors (vascular endothelial growth factor, fibroblast growth factor), and chemokines integrate and loosely associate with this network of GAGs and PGs.7 Altogether, they form the looser layer called endothelial surface layer.

EG Dynamics Under Physiological and Pathological Conditions: The Role of Sheddases

During vessel development, the EG is present on ECs as soon as a flow is initiated.20 In adult life, or on cultured cells, the EG remains as long as ECs are subjected to flow. The EG is often absent from cultivated ECs under static conditions,21 although recent studies were able to observe this structure on glomerular ECs.22

Under physiological conditions, the EG is a stable structure resulting from a balance (Figure 1) between (1) shedding of its components, following continuous passage of the blood flow (shear stress); (2) adsorption of components from circulating blood; and (3) synthesis of new components, including HA, by ECs.7,9,10,23,24 The type of flow imposed on ECs also influences the thickness and composition of the EG. A thinner EG is observed in vascular tree areas that are predisposed to develop atherosclerosis,25 for example, at arterial bifurcations where flow is disturbed. In these areas, the amount of HA and HS decreases and LDLs (low-density lipoproteins) rapidly accumulate in the intima.26 On the other hand, a pulsatile laminar flow imposed on cultured ECs mimics healthy intravascular conditions and raises HA synthesis through the HA synthase HAS2 (HA synthase 2).27 Shear stress also increases HA incorporation into the EG.28 Therefore, HA seems to be systematically associated with favorable conditions for the endothelium.

Figure 1.

Figure 1.

Endothelial glycocalyx structure dynamics. Endothelial glycocalyx (EG) structure (A) under physiological conditions and (B) in pathological conditions. In light green: hyaluronan (HA), in red: heparan sulfate (HS), in dark: chondroitin sulfate, in purple: plasma proteins, in dark green: glycoproteins and proteoglycans such as sdc (syndecan)-1. A, EG structure and composition is the result of a balance between shedding of its components after shear stress, the adsorption of components from circulating blood, and synthesis of new components by endothelial cells (ECs). HA synthesis increases under protective flow (pulsatile laminar flow). B, Under pathological conditions, EG integrity is damaged through the shedding of one or more of its components (eg, sdc1, HS, or HA) into the blood. Under disturbed flow (such as in vascular tree bifurcations areas), HA EG content decreases.

Several pathological conditions and various molecules are known to induce shedding of ≥1 components of the EG (eg, sdc1, HS, or HA) in the blood and eventually the urine as follows: inflammation,29 hyperglycemia,30 septicemia, ischemia-reperfusion,24 cardiopulmonary bypass surgery and ensuing lack of oxygen,31 abnormal shear stress,28 hypertension, viral infections,12 oxidized LDLs,32 and TNF-α (tumor necrosis factor α).33 The size of the EG also decreases in collateral arteries after vascular occlusion of the main branch.34

The cellular mechanisms involved in this EG degradation are not well known but key enzymes, such as heparanase, released by mast cells and podocytes, and HYAL1 (hyaluronidase 1), a hyaluronidase that is concentrated in, and perhaps released by, ECs,35,36 seem to be the main candidates of a group that can be called sheddases. For instance, heparanase activity increases in blood and tissues during respiratory failure associated with septicemia37 or following angiopoietin-2 stimulation during sepsis.38 Similarly, in the atherosclerosis-prone ApoE (apolipoprotein E) deficient mice made diabetic, atrasentan, an endothelin receptor antagonist, or inhibition of monocyte chemotactic protein-1 protects against glomerular EG alteration via a decrease of heparanase expression.39,40 Recently, we showed that HYAL1-deficient mice display a thicker EG and are protected from EG alterations and endothelial dysfunction in early diabetes mellitus.41

Other sheddases have also been suggested. (1) MMPs (matrix metalloproteases) seem to be responsible for the degradation of EG in viral infections such as dengue or hantavirus.4244 In cultured ECs, TNF-α activates MMP-9, which in turn cleaves sdc4 and HS.45 (2) Thrombin and plasmin, involved in the thrombotic and fibrinolytic processes, respectively, also cleave sdc4 expressed on the surface of human umbilical vascular ECs.46 (3) Cathepsin B and tryptase levels increase in coronary blood after cardiac ischemia, in parallel with EG degradation.47,48 (4) Serine proteases, such as elastase and proteinase 3, released by endothelium-adherent leukocytes, can also participate in EG degradation.37 (5) In cultured ECs, HYAL2 attached to the external side of the plasma membrane is implicated in glycocalyx impairment under low shear stress.49

Functions of the EG

In all arterial or venous blood vessels of any caliber, the negatively charged EG participates in the selective permeability of the endothelial barrier, acting as a charge and size barrier against protein and macromolecule diffusion from blood to the interstitial space. Early, in-depth EG studies have reported that its enzymatic degradation leads to increased vascular permeability,50 a fact that is not disputed.

As it delimits a space between circulating blood and ECs, the EG also participates in endothelial protection and homeostasis. Under physiological conditions, the GAG chains form a screen in front of adhesion molecules and sterically prevent binding of leukocytes or platelets to adhesion receptors such as ICAM1 (intercellular adhesion molecule 1), VCAM1 (vascular cell adhesion molecule 1), and von Willebrand factor. Therefore, EG degradation impairs a major protection against endothelial activation and leads to increased leukocyte adhesion and possibly thrombosis.51

The EG functions go far beyond steric protection of the endothelium. The EG has also been identified as a vascular protector through regulation of the vessel microenvironment, according to the following mechanisms. (1) The EG catches several enzymes, inhibitors, or agonists within its structure. For example, the EG may interact with antithrombin III and thrombomodulin, thereby contributing to the antithrombotic properties of a healthy endothelium.7,52 (2) The EG also binds cytokines and modulates the inflammatory response. For example, sdc1, wearing HS chains, creates a chemotactic interleukin-8 gradient which guides neutrophil recruitment to the appropriate places.53 (3) The EG globally reduces oxidative stress through superoxide dismutase retention.7 (4) Finally, one of the main functions of the EG is mechanosensing (aka mechanotransduction), together with shear stress transduction.

EG and Mechanotransduction

Mechanotransduction is a complex process, which is not completely understood. It means that extracellular sensory stimuli such as shear stress, compression, or cell tension induce ≥1 intracellular signals via activation of specific receptor types, such as tyrosine kinase and G protein–coupled receptors.54 Mechanotransduction is initiated on the cell surface and transmitted to the cytoskeleton, in the direction of intercellular junction sites and cell-matrix adhesion sites, as well as toward the nucleus for transcription regulation. Being in close contact with blood flow in the vessel lumen and directly attached to the cytoskeleton actin on the other side, EG extensions are ideally located to detect shear stress and transmit it to the surrounding intracellular structures.55,56 In fact, the EG has been proposed for many years as a key actor in flow-induced vasorelaxation.57 However, to this date, the actual intermediate signals of flow-induced, shear stress–related mechanotransduction have not been elucidated, and the role of EG in this regard is unresolved. What is known is that enzymatic degradation of the EG by neuraminidase, heparinase, or hyaluronidase decreases flow-induced nitric oxide (NO) production,55,58 confirming that sialic acids, HA, and HS-containing GAG chains play an important role in this signal transmission. One of the main roles of the EG in mechanotransduction may thus be initiation of NO-dependent vasorelaxation by shear forces acting directly on the EC surface.11 NO-dependent relaxation can also be induced by various agonists such as acetylcholine acting on their respective EC receptors. EC alignment with the direction of flow is also inhibited after EG destruction by heparinase treatment,59 bringing further evidence to the role of the EG in EC response to blood flow.

Core proteins of EC PGs, such as sdc1, sdc2, sdc4, and gpc1, also take part in the process of mechanotransduction. The cytoplasmic tails of syndecans associate with the cytoskeleton, although the secondary structure of their ectodomains predicts a flexible molecule.60 Fluid shear stress transmitted to the core proteins of the EG and the connection to the actin cytoskeleton mediate specific cell signaling such as NO production and cytoskeletal reorganization. Sdc1 or sdc 4 depletion alter flow sensing and the cell alignment that should follow.61,62 In the absence of sdc1, ECs exposed to an atheroprotective flow abolish key signaling events such as Akt phosphorylation and exhibit an inflammatory and atherosclerotic phenotype.63 Silencing of the gpc1 gene inhibits shear stress–induced activation of the endothelial NO synthase.61

Various elements other than the EG also participate in mechanotransduction. However, most of them are directly associated with the EG.64 These elements comprise the following. (1) Integrin-type cell adhesion receptors: these transmembrane proteins interact on one side with the ECM components, whereas their cytoplasmic domain links to the cytoskeleton. Some integrins present in the EG are involved in flow-induced vasodilation and arterial pressure regulation.65,66 (2) Mechanosensor microdomains called caveolae: these 50- to 100-nm deep plasma membrane invaginations are enriched in cholesterol, sphingolipids, and different signaling molecules such as receptors with tyrosine kinase activity, G protein–coupled receptors, kinases, phosphatases, and ion channels.67 Clustering of these molecules in caveolae optimizes signal transduction. Caveolae are closely related to the EG because they contain the HA receptor CD44 and essential EG PGs such as gpc1. (3) The primary cilium on ECs: this 1- to 5-μm-long protrusion into the vessel lumen contains cytoskeleton microtubules connected to the cytoplasm and may thus play a critical role in mechanotransduction. For instance, primary cilia on embryonic heart ECs induce a shear response through Kruppel-like factor-2 activation, which positively regulates the endothelial NO synthase and downregulates endothelin production.68 These sensory primary cilia are exclusively present in areas of low and disturbed blood69,70 and appear as a self-regulatory mechanism for ECs to modulate their response to shear stress. The cilia may take over mechanotransduction in poor EG areas.68

Several mechanisms have been proposed to explain the mechanosensor function of the EG, although none of them is well demonstrated. The 2 main proposals are as follows: (1) the EG may reorganize under shear stress, thanks to the mobility of gpc1, attached to the plasma membrane through a glycosylphosphatidylinositol anchor. This would lead to a clustering of HS and activation of intracellular signals16,71; and (2) the HS and HA molecules may deform (torque), allowing them to interact with some elements of the cytoskeleton and activate NO synthesis.72,73

Significance of HA for EG Functions

Despite the importance of HA in the EG, the way HA attaches to the plasma membrane and integrates the EG remains unclear. HA is either bound to its receptor CD447476 or remains attached to the extracellular part of the HA synthases, which are located at the inner face of the EC plasma membrane and extrude their reaction product extracellularly through a pore.77,78 HA can also interact with chondroitin sulfate on sdc154 or can link to the EG through HA-binding proteins.79 Finally, HA may self-assemble to form a fibrous network.80

From the earliest studies of EG enzymatic degradation, it appeared that both HA and HS are involved in EG integrity and functions.81,82 Optical and fluorescence microscopy studies have suggested that HS, concentrated in the upper layer of the EG, is responsible for maintaining its structure, whereas HA and chondroitin sulfate, present in the denser layer adjacent to the ECs, would ensure the selective permeability of the EG.14 This hypothesis remains untested.

EG degradation through hyaluronidase treatment revealed an involvement of HA in most of the functions described for EG (Figure 2; Table 1).

Figure 2.

Figure 2.

Hyaluronan (HA) involvement in endothelial glycocalyx (EG) functions. A, Protection and homeostasis of the endothelium: EG as a screen in front of adhesion molecules, preventing binding of leukocytes to adhesion receptors and reducing endothelial activation by limiting the direct effect of shear on EC plasma membrane. B, Mechanotransduction: signal transduction from the blood to the cytoskeleton of the EC: NO production induced in response to shear stress, and vasodilation during reactive hyperemia. C, Selective permeability of the endothelial barrier with regard to protein and macromolecule diffusion leading to a regulation of water efflux. Protection against proteinuria and fluid leakage.

Table 1.

Evidence of HA Involvement in Many Key Functions of the Endothelial Glycocalyx

graphic file with name atv-38-1427-g002.jpg

EG in Diabetic Pathology

EG Destruction During Diabetes Mellitus

Chronic hyperglycemia induces both micro- and macrovascular complications that affect various organs like the skin, muscles, heart, brain, eyes, and kidneys. Microvascular complications comprise retinopathy, nephropathy, and neuropathy. The main macrovascular complications result from atherosclerotic plaques and include ischemic heart disease, stroke, and peripheral artery disease. All these macrovascular events may also occur independently of diabetes mellitus, so it may be difficult to disentangle the effects of diabetes mellitus and atherosclerosis per se. In fact, diabetes mellitus–induced atherosclerosis seems to follow the same histological course as in nondiabetic conditions, including endothelial injury, smooth muscle cell proliferation, foam cell development and infiltration, platelet activation, and increased inflammation. In diabetes mellitus, all these pathogenic steps occur in an accelerated way, in more diffuse localizations, with higher vascular remodeling and more plaque ruptures.8386 Some key factors in the biochemical pathways involved in the progression of atherosclerosis in diabetic patients have been identified. They comprise overproduction of reactive oxygen species (ROS), increased formation of advanced glycation end products (AGEs) and activation of AGE receptors, enhanced polyol and hexosamine fluxes, PKC activation, and chronic vascular inflammation.85

A large body of evidence supports the link between the development of atheromatous lesions and a thin EG overlying local thickening of the intima in bifurcation and branching areas.87,88 Although this vascular phenomenon has not been specifically studied in the case of diabetes mellitus, it is clear that most diabetic micro- and macrovascular complications, if not all, are also coupled with damaged EG. We will focus our discussion on diabetes mellitus–related EG damage.

The involvement of EG damage in diabetic pathophysiology, which had been suspected for a long time, was actually demonstrated only recently. Intravital microscopy revealed a decrease in EG systemic volume both in type 1 diabetes mellitus (T1DM) patients and during induced acute hyperglycemia.89,90 A decrease of EG volume in sublingual and retinal vessels was also observed in type 2 diabetes mellitus (T2DM) patients.91 Damage to the EG has also been reported in animal models. In rat and mouse T1DM models of retinopathy, the size of the EG of retinal vessels is significantly decreased.92,93 In db/db mice, simulating T2DM, the EG of brain microvessels is altered.94 Mouse models of diabetic nephropathy, because of either streptozotocin-induced or congenital (Akita mice) diabetes mellitus, also show a damaged glomerular EG.9597

In terms of functional consequences, EG deterioration during diabetes mellitus is clearly associated with many typical phenomena of micro- and macrovascular complications, such as decreased arterial vasodilation98 (a typical step in endothelial dysfunction), increased endothelial permeability to macromolecules,30 and, in the retinal vessels, fluid leakage, loss of pericytes, and neovascularization.93 EG damage during diabetes mellitus is also associated with activation of coagulation,90 increased leukocyte adhesion,92 development of albuminuria,9597 and deterioration of the blood-brain barrier.94

Endothelial Dysfunction and Vascular Complications of Diabetes Mellitus

Endothelial dysfunction is generally defined as a decrease in the capacity of the vessel to dilate in response to an endothelium-dependent vasodilator (eg, acetylcholine or bradykinin) or in response to blood flow.99,100 Other signs of endothelial dysfunction are changes in adhesion molecules (selectin, ICAM1) and proinflammatory molecules expression, decrease in platelet aggregation inhibition (procoagulation status), and modification in the regulation of smooth muscle cell proliferation (proliferative status).

Endothelial dysfunction is universally present in T1DM and T2DM patients and in experimental diabetes mellitus models.101 It is an early predictor of micro- and macrovascular complications related to diabetes mellitus. However, the link between diabetes mellitus and the pathogenesis of vascular complications cannot be summarized by a single mechanism because in diabetic patients, especially T2DM, hyperglycemia can also be coupled with hypertension, lipid metabolism deregulation, or low systemic inflammation. However, hyperglycemia remains primordial in the cause of diabetes mellitus–related endothelial dysfunction.102

Mechanisms of EG Alteration During Hyperglycemia

The mechanisms of EG degradation in diabetes mellitus, or in other diseases for that matter, are not well known.103 It was suggested that hyperglycemia modifies the sulfation of HS in the Golgi apparatus, but investigations of this hypothesis yielded conflicting results.104,105 Other authors suggested a decreased amount of GAGs on PGs, which could then modify the barrier properties of the EG.106 The relevance of these in vitro observations remains uncertain. More convincingly, the involvement of 3 factors in EG deterioration during diabetes mellitus has received experimental support. These factors are ROS, AGEs, and activation of the sheddases heparanase and HYAL1.

The involvement of ROS in diabetic complications is well established. An overproduction of O2 (superoxide anion) and its resulting increased oxidative stress systematically accompany hyperglycemia. This pathway has been implicated in all 4 main mechanisms pointed out to explain the impact of hyperglycemia on ECs: activation of the polyol and hexosamine pathways, protein kinase C activation, and AGE formation.107 Antioxidant treatment can partially avoid EG damage.90,108 Finally, ROS are able to depolymerize GAGs and more particularly HA.109111 AGEs, another systematic consequence of hyperglycemia, are also responsible for high–molecular weight HA degradation.112

Besides ROS and AGEs, usual suspects in all diabetic complications, the roles of heparanase and the hyaluronidase HYAL1 have recently been exposed. Heparanase is secreted by macrophages and endothelin-activated podocytes and seems directly implicated in the process of EG degradation leading to diabetic nephropathy.39,113,114 For instance, inhibition of monocyte chemoattractant protein-1 restores the EG in a mouse model of combined diabetic/atherosclerotic nephropathy through reduction of heparanase expression by macrophages.40

Evidence on the implication of HYAL1 in EG degradation is also substantial. The amount of HYAL1 in the plasma, measured by ex vivo enzymatic activity, increases during diabetes mellitus in mice, rats, and humans.41,89,91,115,116 Furthermore, our laboratory has shown that streptozotocin-induced diabetic mice lacking HYAL1 display a thicker EG and are protected from HA shedding, endothelial dysfunction, and microalbuminuria in early diabetes mellitus, when compared with HYAL1 competent mice.41 Together, these observations point to HYAL1 as a major actor of early diabetic vascular (and possibly renal) complications.

Role of Hyaluronidases

Like all vertebrate hyaluronidases, HYAL1 is an endo-β-N-acetyl-hexosaminidase that acts through hydrolysis of its substrate.117 It is the most active enzyme in HA degradation and homeostasis, as shown by comparative HA-degrading activities of various hyaluronidases (HYAL2 is much less active118) and by 40- to 100-fold elevations of plasma HA levels in HYAL1-deficient humans.119 A theoretical model for HA catabolism in somatic tissue proposes internalization of HA through the coupled action of the HA receptor CD44 and HYAL2 on the cell surface, in an acidic microenvironment created by the Na+/H+ exchanger-1 proton pump. This allows cleavage of HA into 20-kDa fragments that are then routed to the lysosomes in which HYAL1 continues the degradation of HA into tetrasaccharides.120,121

Despite this model, the way HYAL1 could cleave HA in the EG is not known. One possibility is that ECs could endocytose HYAL1 from the plasma, where it circulates without being active (the pH being too high). HYAL1 could then reach its optimal pH of activity (ca. 3.7) in the EC lysosomes after endocytosis. In support of this scheme, uptake of intravenously injected HYAL1 into liver sinusoidal ECs has been demonstrated.35 In addition, several cell types including RAW264.7 macrophages,122 Hu22RV1 from prostate adenocarcinoma,123 and EA.hy926 ECs (S. Dogné unpublished data, 2014) display an active endocytosis of HYAL1. The supply of HYAL1 from the blood to ECs could, therefore, increase during diabetes mellitus. This hypothesis is attractive but several gaps in knowledge remain. For example, what is the source of increased plasma HYAL1 in diabetes mellitus? Is increased plasma HYAL1 a cause or a consequence of EG degradation? Is there, in addition, an increased acidic microenvironment in the diabetic EG that could allow increased HA endocytosis into ECs (perhaps through HYAL2) and thus increased HA degradation? All these elements remain to be explored.

HYAL2, the other main somatic hyaluronidase implicated in HA metabolism, may also play a role in the shedding of EG HA during diabetes mellitus because HYAL2 is a glycosylphosphatidylinositol-anchored enzyme located at the surface of the ECs as in many other cell types.124 Indeed, HYAL2 has been suggested as an actor of EG damage in cultured ECs under low shear stress.49 Furthermore, HYAL2-deficient mice raised in our laboratory have a thicker EG than their wild-type counterparts.125 However, to our knowledge, no evidence of increased HYAL2 activity in diabetes mellitus or implication of HYAL2 in EG degradation besides cultured ECs has been provided. The role of HYAL2 in diabetic complications remains speculative.

Plasma Markers of EG Shedding

Signs of EG degradation in clinical conditions can be evaluated by measuring the elevation of some EG components in plasma, usually sdc1, HS, or HA. Which of these components is altered depends on the pathology.

During ischemia/reperfusion procedures in vascular surgery (global or regional ischemia), drastic increases in sdc1 (ca. 40×) and HS (10×) can be measured in the blood.31 In immediate survivors of cardiac arrest, significant increases in sdc1, HS, and HA plasma levels are observed.126 Ischemia-reperfusion performed in vivo on rat liver or on isolated guinea pig heart increases the release of HS and sdc1. This elevation is associated with a decrease of EG thickness, as observed by electron microscopy.47,127

In sepsis, sdc1 and HS plasma levels serve as blood markers of EG disruption128 and strongly correlate with the disease severity.129 Recently, HA plasma levels have also proved to be elevated in sepsis and to represent a prognostic marker for morbidity and survival.130 Association of EG degradation with inflammation is observed in a dog septic shock model where sdc1 and HS plasma levels correlate with those of IL (interleukin)-6 and TNF-α.131

Sdc1 and HS plasma levels are also indirect markers of EG degradation in hemorrhaged rats after resuscitation fluids.132 After prolonged moderate-intensity endurance training, the same markers have been proposed to evaluate EG integrity.133

In chronic kidney disease, sdc1 and HA plasma levels increase continuously with the severity stages of the disease134; they are also increased in dialysis patients.135 A rat model of chronic kidney disease shows increased sdc1 plasma levels coupled to a significant decrease in EG thickness, as measured by atomic force microscopy.134

Finally, in diabetes mellitus, an alteration in blood GAG concentration (increased HA, decreased HS, and chondroitin sulfate) was reported as early as 1973.136,137 A slightly different alteration in serum GAG concentration (increased chondroitin sulfate and dermatan sulfate, decreased HS) was later reported in T2DM.138 More recently, HA and sdc1 were proposed as blood or plasma markers of EG shedding in diabetes mellitus.89,90,139,140 HA is discussed below. Sdc1 plasma levels increase in T2DM patients139 and in T1DM patients with diabetic nephropathy, in parallel with microalbuminuria.140,141

In summary, plasma markers of EG shedding such as sdc1 and HA are available and have been validated in several cardiovascular diseases and in diabetes mellitus but to date have not made their way into the clinic, perhaps because the importance of EG degradation besides other markers of severity of diabetes mellitus is not yet fully recognized.

Shedding of EG HA During Diabetes Mellitus

As previously described, hyperglycemia is responsible for EG deterioration through several mechanisms, 3 of which (ROS, AGEs, and hyaluronidase activity) are capable of depolymerizing HA. Shrinkage, alteration, or degradation of the EG is associated with development of cardiovascular diseases.142 During acute hyperglycemia or in patients with T1DM, the size of the EG is reduced by 50% to 80% and plasma HA concentration increases by 30% to 80%.89,90,116 This HA shedding contributes to increasing vascular permeability, overactivated coagulation, and decreasing flux-induced vasodilation. A degraded EG coupled with an increased plasma HA concentration is also observed in T2DM patients91 and in rodent models of T1DM.41,115 Furthermore, in HYAL1-deficient mice, an unusual increase in EG thickness coupled with a decrease in HA shedding results in a protection of endothelial function and glomerular barrier during early diabetes mellitus.41 Shedding of HA further to alteration of the EG can therefore be considered as a solid observation in the pathogenesis of diabetic vascular and renal complications.

Dual and Opposing Roles of HA in Vascular Complications

Because HA localizes in the ECM of all tissues, it is also implicated in diabetic tissue remodeling, notably in vessel walls and kidneys, leading to micro- and macrovascular complications. In diabetic rat glomeruli, abnormal deposits of HA combine with infiltration of macrophages and monocytes.143,144 In addition, neosynthesized HA accumulates within mesangial cells and induces the release of HA aggregates in the extracellular space, forming cables which are adhesive for monocytes.145 This HA accumulation within the mesangium could participate in the development of diabetic glomerulosclerosis. Information on HA accumulation in diabetic vessels is limited. In the aortic intima of diabetic pigs, for instance, HA accumulates only in the presence of hyperlipidemia.146 On the other hand, abnormal HA deposition, especially in vessel walls and plaques, has been implicated in several models of atherosclerosis in the absence of diabetes mellitus.147151

There is, however, a striking example in an atheromatic disease model, the ApoE−/− mice, where the suppression of HA deposits in the tissues (including vessel walls) during atherogenesis does not produce beneficial effects, on the contrary, because of simultaneous disappearance of EG HA. Indeed the inhibition of HA synthesis using 4-methylumbelliferone in these ApoE−/− mice led to altered relaxation in response to acetylcholine and to a rise in plaque areas.152 The thickness of EG was reduced by the HA synthesis inhibitor. Whether a true protection of vessel integrity would be afforded by maintaining EG HA in the face of diabetic insults remains to be demonstrated.

Prevention of EG Degradation: A New Therapeutic Target

Several recent studies and reviews have concluded to the therapeutic necessity to avoid EG degradation or to promote its synthesis and recovery in both atherosclerotic disease and diabetes mellitus.12,13,153 EG recovery after experimental enzymatic degradation, using hyaluronidase or heparinase injections, was the subject of several early studies showing that recovery was relatively fast and could be accelerated by infusion of a mixture of HA and chondroitin sulfate or by supplementation with exogenous HS, respectively.51,82 Prevention of EG degradation during pathologies is undoubtedly more complex, although some successes have been observed in animal models. Studies in humans are rare.

The main examples can be summarized as follows. (1) In rats, using plasma instead of saline for resuscitation after hemorrhagic shock increases sdc1 expression and restores EG thickness.154 (2) In guinea pigs, albumin prevents EG degradation (shedding) during cardiac transplantation, decreasing fluid leakage and preventing leukocyte adhesion to the endothelium.155 This EG stabilization seems to be mediated by sphingosine-1 phosphate naturally linked to albumin.156 (3) The antioxidants superoxide dismutase and catalase preserve EG HS amount and barrier property in cultured glomerular ECs submitted to ROS.157 These antioxidants also maintain EG thickness and inhibit leukocyte adhesion in hamster cremaster muscle capillaries after oxidized LDL challenge.158 (4) Doxycycline decreases EG shedding and resulting leukocyte adhesion on ECs during ischemia-reperfusion in rats.159 (5) On the isolated guinea pig heart, hydrocortisone and antithrombin also preserve EG thickness, maintaining its antiadhesive and barrier properties, alleviating vascular leakage (edema) and inflammation.160162 (6) Intravenous injection of exogenous HA during ischemia-reperfusion in mice avoids thinning of EG and damage to the permselectivity barrier of the EG.5 (7) Intravenous injection of wheat germ agglutinin lectin, which adsorbs to the EG, in rats with chronic kidney disease reduces albuminuria.163 (8) Finally, in patients with hypercholesterolemia, rosuvastatin also preserves EG thickness and prevents HA shedding through normalization of the LDL levels.164

On diabetic pathology, the main approaches tested to date are listed in Table 2 and illustrated in Figure 3. Only one of the potential therapeutic products (sulodexide) has been tested in humans but found to not provide sufficient clinical efficacy for limiting long-term diabetic renal complications.165 In animal models, inhibitors of heparanase and HYAL1 have been suggested as new therapeutic tools.3941,166

Table 2.

Therapeutic Approaches to Protect or Improve the Endothelial Glycocalyx During Diabetes Mellitus

graphic file with name atv-38-1427-g004.jpg

Figure 3.

Figure 3.

Summary of the main approaches tested to prevent endothelial glycocalyx (EG) degradation or to restore it in diabetes mellitus. Effects of each tested component (various colors) are pointed out with a closed tip arrow, at every stage where benefits have been observed. Only sulodexide has been tested in humans but found to not provide sufficient clinical efficacy for limiting long-term diabetic renal complications. In animal models, inhibitors of heparanase and HYAL1 (hyaluronidase 1) are suggested as new therapeutic tools. AGE indicates advanced glycation end product; ETA, endothelin receptor A; ETB, endothelin receptor B; HA, hyaluronan; HS, heparan sulfate; and ROS, reactive oxygen species.

Conclusions

Damage to the EG during hyperglycemia seems to be an initial and critical step in provoking endothelial dysfunction. Although the mechanisms of this degradation are not well understood, preserving some key components of the EG seems a good proposal to try and prevent diabetic micro- and macrovascular complications, most of which are initiated by endothelial dysfunction. This dysfunction may result in large part from EG damage. HA is a major component of the EG and was shown to be required in various functions of this protective layer. HA may also buffer ROS and AGE attacks during hyperglycemic episodes. Although several therapeutic molecules have been tested for EG protection and restoration in animal models of vascular diseases and even diabetes mellitus, none of them has been shown to have clinical effectiveness. Preserving the size and amount of EG HA in the face of diabetic insults could be a useful novel therapeutic strategy to slow diabetic complications. One way to achieve this goal, as suggested by a murine model of HYAL1 deficiency, may be to inhibit the function of HYAL1, the main circulating hyaluronidase that seems enriched in the endothelial layer and may participate in HA removal from, and collapse of, the EG during diabetes mellitus.

Acknowledgments

The assistance of M. Savels (University of Namur) is greatly appreciated.

Disclosures

None.

Highlights

  • The endothelial glycocalyx (EG) covers the apical surface of the endothelial cells and is essential to vascular integrity and cardiovascular homeostasis.

  • Hyaluronan, a key glycosaminoglycan of the EG, is implicated in most of the functions described for EG. The amount of hyaluronan in the EG may be regulated by the HYAL (hyaluronidase) 1.

  • Diabetes mellitus induces damage to the EG, including shedding of hyaluronan into the plasma and increases in plasma hyaluronidase. This is an early sign of vascular dysfunction.

  • Preserving the size and amount of EG hyaluronan, for example, through HYAL1 inhibition, is an attractive novel therapeutic strategy to slow diabetic vascular complications.

References

  • 1.Luft JH. Fine structures of capillary and endocapillary layer as revealed by ruthenium red. Fed Proc. 1966;25:1773–1783. [PubMed] [Google Scholar]
  • 2.Vink H, Duling BR. Identification of distinct luminal domains for macromolecules, erythrocytes, and leukocytes within mammalian capillaries. Circ Res. 1996;79:581–589. doi: 10.1161/01.res.79.3.581. [DOI] [PubMed] [Google Scholar]
  • 3.van den Berg BM, Vink H, Spaan JA. The endothelial glycocalyx protects against myocardial edema. Circ Res. 2003;92:592–594. doi: 10.1161/01.RES.0000065917.53950.75. doi: 10.1161/01.RES.0000065917.53950.75. [DOI] [PubMed] [Google Scholar]
  • 4.Smith ML, Long DS, Damiano ER, Ley K. Near-wall micro-PIV reveals a hydrodynamically relevant endothelial surface layer in venules in vivo. Biophys J. 2003;85:637–645. doi: 10.1016/s0006-3495(03)74507-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Rubio-Gayosso I, Platts SH, Duling BR. Reactive oxygen species mediate modification of glycocalyx during ischemia-reperfusion injury. Am J Physiol Heart Circ Physiol. 2006;290:H2247–H2256. doi: 10.1152/ajpheart.00796.2005. doi: 10.1152/ajpheart.00796.2005. [DOI] [PubMed] [Google Scholar]
  • 6.Megens RT, Reitsma S, Schiffers PH, Hilgers RH, De Mey JG, Slaaf DW, oude Egbrink MG, van Zandvoort MA. Two-photon microscopy of vital murine elastic and muscular arteries. Combined structural and functional imaging with subcellular resolution. J Vasc Res. 2007;44:87–98. doi: 10.1159/000098259. doi: 10.1159/000098259. [DOI] [PubMed] [Google Scholar]
  • 7.Reitsma S, Slaaf DW, Vink H, van Zandvoort MA, oude Egbrink MG. The endothelial glycocalyx: composition, functions, and visualization. Pflugers Arch. 2007;454:345–359. doi: 10.1007/s00424-007-0212-8. doi: 10.1007/s00424-007-0212-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Nieuwdorp M, Meuwese MC, Mooij HL, Ince C, Broekhuizen LN, Kastelein JJ, Stroes ES, Vink H. Measuring endothelial glycocalyx dimensions in humans: a potential novel tool to monitor vascular vulnerability. J Appl Physiol (1985) 2008;104:845–852. doi: 10.1152/japplphysiol.00440.2007. doi: 10.1152/japplphysiol.00440.2007. [DOI] [PubMed] [Google Scholar]
  • 9.Pries AR, Secomb TW, Gaehtgens P. The endothelial surface layer. Pflugers Arch. 2000;440:653–666. doi: 10.1007/s004240000307. doi: 10.1007/s004240000307. [DOI] [PubMed] [Google Scholar]
  • 10.Weinbaum S, Tarbell JM, Damiano ER. The structure and function of the endothelial glycocalyx layer. Annu Rev Biomed Eng. 2007;9:121–167. doi: 10.1146/annurev.bioeng.9.060906.151959. doi: 10.1146/annurev.bioeng.9.060906.151959. [DOI] [PubMed] [Google Scholar]
  • 11.Drake-Holland AJ, Noble MI. Update on the important new drug target in cardiovascular medicine - the vascular glycocalyx. Cardiovasc Hematol Disord Drug Targets. 2012;12:76–81. doi: 10.2174/187152912801823183. [DOI] [PubMed] [Google Scholar]
  • 12.Becker BF, Jacob M, Leipert S, Salmon AH, Chappell D. Degradation of the endothelial glycocalyx in clinical settings: searching for the sheddases. Br J Clin Pharmacol. 2015;80:389–402. doi: 10.1111/bcp.12629. doi: 10.1111/bcp.12629. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Tarbell JM, Cancel LM. The glycocalyx and its significance in human medicine. J Intern Med. 2016;280:97–113. doi: 10.1111/joim.12465. doi: 10.1111/joim.12465. [DOI] [PubMed] [Google Scholar]
  • 14.Gao L, Lipowsky HH. Composition of the endothelial glycocalyx and its relation to its thickness and diffusion of small solutes. Microvasc Res. 2010;80:394–401. doi: 10.1016/j.mvr.2010.06.005. doi: 10.1016/j.mvr.2010.06.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Squire JM, Chew M, Nneji G, Neal C, Barry J, Michel C. Quasi-periodic substructure in the microvessel endothelial glycocalyx: a possible explanation for molecular filtering? J Struct Biol. 2001;136:239–255. doi: 10.1006/jsbi.2002.4441. doi: 10.1006/jsbi.2002.4441. [DOI] [PubMed] [Google Scholar]
  • 16.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.0086249. doi: 10.1371/journal.pone.0086249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Kinsella MG, Bressler SL, Wight TN. The regulated synthesis of versican, decorin, and biglycan: extracellular matrix proteoglycans that influence cellular phenotype. Crit Rev Eukaryot Gene Expr. 2004;14:203–234. doi: 10.1615/critreveukaryotgeneexpr.v14.i3.40. [DOI] [PubMed] [Google Scholar]
  • 18.Reitsma S, oude Egbrink MG, Vink H, van den Berg BM, Passos VL, Engels W, Slaaf DW, van Zandvoort MA. Endothelial glycocalyx structure in the intact carotid artery: a two-photon laser scanning microscopy study. J Vasc Res. 2011;48:297–306. doi: 10.1159/000322176. doi: 10.1159/000322176. [DOI] [PubMed] [Google Scholar]
  • 19.Job KM, O’Callaghan R, Hlady V, Barabanova A, Dull RO. The biomechanical effects of resuscitation colloids on the compromised lung endothelial glycocalyx. Anesth Analg. 2016;123:382–393. doi: 10.1213/ANE.0000000000001284. doi: 10.1213/ANE.0000000000001284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Henderson-Toth CE, Jahnsen ED, Jamarani R, Al-Roubaie S, Jones EA. The glycocalyx is present as soon as blood flow is initiated and is required for normal vascular development. Dev biol. 2012;369:330–339. doi: 10.1016/j.ydbio.2012.07.009. doi: 10.1016/j.ydbio.2012.07.009. [DOI] [PubMed] [Google Scholar]
  • 21.Potter DR, Damiano ER. The hydrodynamically relevant endothelial cell glycocalyx observed in vivo is absent in vitro. Circ Res. 2008;102:770–776. doi: 10.1161/CIRCRESAHA.107.160226. doi: 10.1161/CIRCRESAHA.107.160226. [DOI] [PubMed] [Google Scholar]
  • 22.Singh A, Fridén V, Dasgupta I, Foster RR, Welsh GI, Tooke JE, Haraldsson B, Mathieson PW, Satchell SC. High glucose causes dysfunction of the human glomerular endothelial glycocalyx. Am J Physiol Renal Physiol. 2011;300:F40–F48. doi: 10.1152/ajprenal.00103.2010. doi: 10.1152/ajprenal.00103.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Laurent TC, Fraser JR. Hyaluronan. FASEB J. 1992;6:2397–2404. [PubMed] [Google Scholar]
  • 24.Mulivor AW, Lipowsky HH. Inflammation- and ischemia-induced shedding of venular glycocalyx. Am J Physiol Heart Circ Physiol. 2004;286:H1672–H1680. doi: 10.1152/ajpheart.00832.2003. doi: 10.1152/ajpheart.00832.2003. [DOI] [PubMed] [Google Scholar]
  • 25.Lewis JC, Taylor RG, Jones ND, St Clair RW, Cornhill JF. Endothelial surface characteristics in pigeon coronary artery atherosclerosis. I. Cellular alterations during the initial stages of dietary cholesterol challenge. Lab Invest. 1982;46:123–138. [PubMed] [Google Scholar]
  • 26.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 Arch. 2009;457:1199–1206. doi: 10.1007/s00424-008-0590-6. doi: 10.1007/s00424-008-0590-6. [DOI] [PubMed] [Google Scholar]
  • 27.Maroski J, Vorderwülbecke BJ, Fiedorowicz K, Da Silva-Azevedo L, Siegel G, Marki A, Pries AR, Zakrzewicz A. Shear stress increases endothelial hyaluronan synthase 2 and hyaluronan synthesis especially in regard to an atheroprotective flow profile. Exp Physiol. 2011;96:977–986. doi: 10.1113/expphysiol.2010.056051. doi: 10.1113/expphysiol.2010.056051. [DOI] [PubMed] [Google Scholar]
  • 28.Gouverneur M, Berg B, Nieuwdorp M, Stroes E, Vink H. Vasculoprotective properties of the endothelial glycocalyx: effects of fluid shear stress. J Intern Med. 2006;259:393–400. doi: 10.1111/j.1365-2796.2006.01625.x. doi: 10.1111/j.1365-2796.2006.01625.x. [DOI] [PubMed] [Google Scholar]
  • 29.Henry CB, Duling BR. TNF-alpha increases entry of macromolecules into luminal endothelial cell glycocalyx. Am J Physiol Heart Circ Physiol. 2000;279:H2815–H2823. doi: 10.1152/ajpheart.2000.279.6.H2815. doi: 10.1152/ajpheart.2000.279.6.H2815. [DOI] [PubMed] [Google Scholar]
  • 30.Zuurbier CJ, Demirci C, Koeman A, Vink H, Ince C. Short-term hyperglycemia increases endothelial glycocalyx permeability and acutely decreases lineal density of capillaries with flowing red blood cells. J Appl Physiol (1985) 2005;99:1471–1476. doi: 10.1152/japplphysiol.00436.2005. doi: 10.1152/japplphysiol.00436.2005. [DOI] [PubMed] [Google Scholar]
  • 31.Rehm M, Bruegger D, Christ F, Conzen P, Thiel M, Jacob M, Chappell D, Stoeckelhuber M, Welsch U, Reichart B, Peter K, Becker BF. Shedding of the endothelial glycocalyx in patients undergoing major vascular surgery with global and regional ischemia. Circulation. 2007;116:1896–1906. doi: 10.1161/CIRCULATIONAHA.106.684852. doi: 10.1161/CIRCULATIONAHA.106.684852. [DOI] [PubMed] [Google Scholar]
  • 32.Constantinescu AA, Vink H, Spaan JA. Elevated capillary tube hematocrit reflects degradation of endothelial cell glycocalyx by oxidized LDL. Am J Physiol Heart Circ Physiol. 2001;280:H1051–H1057. doi: 10.1152/ajpheart.2001.280.3.H1051. doi: 10.1152/ajpheart.2001.280.3.H1051. [DOI] [PubMed] [Google Scholar]
  • 33.Chappell D, Hofmann-Kiefer K, Jacob M, Rehm M, Briegel J, Welsch U, Conzen P, Becker BF. TNF-alpha induced shedding of the endothelial glycocalyx is prevented by hydrocortisone and antithrombin. Basic Res Cardiol. 2009;104:78–89. doi: 10.1007/s00395-008-0749-5. doi: 10.1007/s00395-008-0749-5. [DOI] [PubMed] [Google Scholar]
  • 34.Grundmann S, Schirmer SH, Hekking LH, Post JA, Ionita MG, de Groot D, van Royen N, van den Berg B, Vink H, Moser M, Bode C, de Kleijn D, Pasterkamp G, Piek JJ, Hoefer IE. Endothelial glycocalyx dimensions are reduced in growing collateral arteries and modulate leucocyte adhesion in arteriogenesis. J Cell Mol Med. 2009;13(9B):3463–3474. doi: 10.1111/j.1582-4934.2009.00735.x. doi: 10.1111/j.1582-4934.2009.00735.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Gasingirwa MC, Thirion J, Mertens-Strijthagen J, Wattiaux-De Coninck S, Flamion B, Wattiaux R, Jadot M. Endocytosis of hyaluronidase-1 by the liver. Biochem J. 2010;430:305–313. doi: 10.1042/BJ20100711. doi: 10.1042/BJ20100711. [DOI] [PubMed] [Google Scholar]
  • 36.Bourguignon V, Flamion B. Respective roles of hyaluronidases 1 and 2 in endogenous hyaluronan turnover. FASEB J. 2016;30:2108–2114. doi: 10.1096/fj.201500178R. doi: 10.1096/fj.201500178R. [DOI] [PubMed] [Google Scholar]
  • 37.Schmidt EP, Yang Y, Janssen WJ, et al. The pulmonary endothelial glycocalyx regulates neutrophil adhesion and lung injury during experimental sepsis. Nat Med. 2012;18:1217–1223. doi: 10.1038/nm.2843. doi: 10.1038/nm.2843. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Lukasz A, Hillgruber C, Oberleithner H, Kusche-Vihrog K, Pavenstädt H, Rovas A, Hesse B, Goerge T, Kümpers P. Endothelial glycocalyx breakdown is mediated by angiopoietin-2. Cardiovasc Res. 2017;113:671–680. doi: 10.1093/cvr/cvx023. doi: 10.1093/cvr/cvx023. [DOI] [PubMed] [Google Scholar]
  • 39.Boels MG, Avramut MC, Koudijs A, Dane MJ, Lee DH, van der Vlag J, Koster AJ, van Zonneveld AJ, van Faassen E, Gröne HJ, van den Berg BM, Rabelink TJ. Atrasentan reduces albuminuria by restoring the glomerular endothelial glycocalyx barrier in diabetic nephropathy. Diabetes. 2016;65:2429–2439. doi: 10.2337/db15-1413. doi: 10.2337/db15-1413. [DOI] [PubMed] [Google Scholar]
  • 40.Boels MGS, Koudijs A, Avramut MC, Sol WMPJ, Wang G, van Oeveren-Rietdijk AM, van Zonneveld AJ, de Boer HC, van der Vlag J, van Kooten C, Eulberg D, van den Berg BM, IJpelaar DHT, Rabelink TJ. Systemic monocyte chemotactic protein-1 inhibition modifies renal macrophages and restores glomerular endothelial glycocalyx and barrier function in diabetic nephropathy. Am J Pathol. 2017;187:2430–2440. doi: 10.1016/j.ajpath.2017.07.020. doi: 10.1016/j.ajpath.2017.07.020. [DOI] [PubMed] [Google Scholar]
  • 41.Dogné S, Rath G, Jouret F, Caron N, Dessy C, Flamion B. Hyaluronidase 1 deficiency preserves endothelial function and glycocalyx integrity in early streptozotocin-induced diabetes. Diabetes. 2016;65:2742–2753. doi: 10.2337/db15-1662. doi: 10.2337/db15-1662. [DOI] [PubMed] [Google Scholar]
  • 42.Mulivor AW, Lipowsky HH. Inhibition of glycan shedding and leukocyte-endothelial adhesion in postcapillary venules by suppression of matrixmetalloprotease activity with doxycycline. Microcirculation. 2009;16:657–666. doi: 10.3109/10739680903133714. doi: 10.3109/10739680903133714. [DOI] [PubMed] [Google Scholar]
  • 43.Puerta-Guardo H, Glasner DR, Harris E. Dengue virus NS1 disrupts the endothelial glycocalyx, leading to hyperpermeability. PLoS Pathog. 2016;12:e1005738. doi: 10.1371/journal.ppat.1005738. doi: 10.1371/journal.ppat.1005738. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Connolly-Andersen AM, Thunberg T, Ahlm C. Endothelial activation and repair during hantavirus infection: association with disease outcome. Open Forum Infect Dis. 2014;1:ofu027. doi: 10.1093/ofid/ofu027. doi: 10.1093/ofid/ofu027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Ramnath R, Foster RR, Qiu Y, Cope G, Butler MJ, Salmon AH, Mathieson PW, Coward RJ, Welsh GI, Satchell SC. Matrix metalloproteinase 9-mediated shedding of syndecan 4 in response to tumor necrosis factor α: a contributor to endothelial cell glycocalyx dysfunction. FASEB J. 2014;28:4686–4699. doi: 10.1096/fj.14-252221. doi: 10.1096/fj.14-252221. [DOI] [PubMed] [Google Scholar]
  • 46.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;280:34441–34446. doi: 10.1074/jbc.M501903200. doi: 10.1074/jbc.M501903200. [DOI] [PubMed] [Google Scholar]
  • 47.Annecke T, Chappell D, Chen C, Jacob M, Welsch U, Sommerhoff CP, Rehm M, Conzen PF, Becker BF. Sevoflurane preserves the endothelial glycocalyx against ischaemia-reperfusion injury. Br J Anaesth. 2010;104:414–421. doi: 10.1093/bja/aeq019. doi: 10.1093/bja/aeq019. [DOI] [PubMed] [Google Scholar]
  • 48.Annecke T, Fischer J, Hartmann H, Tschoep J, Rehm M, Conzen P, Sommerhoff CP, Becker BF. Shedding of the coronary endothelial glycocalyx: effects of hypoxia/reoxygenation vs ischaemia/reperfusion. Br J Anaesth. 2011;107:679–686. doi: 10.1093/bja/aer269. doi: 10.1093/bja/aer269. [DOI] [PubMed] [Google Scholar]
  • 49.Kong X, Chen L, Ye P, Wang Z, Zhang J, Ye F, Chen S. The role of HYAL2 in LSS-induced glycocalyx impairment and the PKA-mediated decrease in eNOS-Ser-633 phosphorylation and nitric oxide production. Mol Biol Cell. 2016;27:3972–3979. doi: 10.1091/mbc.E16-04-0241. doi: 10.1091/mbc.E16-04-0241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Henry CB, Durán WN, DeFouw DO. Permselectivity of angiogenic microvessels following alteration of the endothelial fiber matrix by oligosaccharides. Microvasc Res. 1997;53:150–155. doi: 10.1006/mvre.1996.1996. doi: 10.1006/mvre.1996.1996. [DOI] [PubMed] [Google Scholar]
  • 51.Constantinescu AA, Vink H, Spaan JA. Endothelial cell glycocalyx modulates immobilization of leukocytes at the endothelial surface. Arterioscler Thromb Vasc Biol. 2003;23:1541–1547. doi: 10.1161/01.ATV.0000085630.24353.3D. doi: 10.1161/01.ATV.0000085630.24353.3D. [DOI] [PubMed] [Google Scholar]
  • 52.Shimada K, Kobayashi M, Kimura S, Nishinaga M, Takeuchi K, Ozawa T. Anticoagulant heparin-like glycosaminoglycans on endothelial cell surface. Jpn Circ J. 1991;55:1016–1021. doi: 10.1253/jcj.55.1016. [DOI] [PubMed] [Google Scholar]
  • 53.Götte M, Echtermeyer F. Syndecan-1 as a regulator of chemokine function. ScientificWorldJournal. 2003;3:1327–1331. doi: 10.1100/tsw.2003.118. doi: 10.1100/tsw.2003.118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Tarbell JM, Ebong EE. The endothelial glycocalyx: a mechano-sensor and -transducer. Sci Signal. 2008;1:pt8. doi: 10.1126/scisignal.140pt8. doi: 10.1126/scisignal.140pt8. [DOI] [PubMed] [Google Scholar]
  • 55.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–e142. doi: 10.1161/01.RES.0000101744.47866.D5. doi: 10.1161/01.RES.0000101744.47866.D5. [DOI] [PubMed] [Google Scholar]
  • 56.Pahakis MY, Kosky JR, Dull RO, Tarbell JM. The role of endothelial glycocalyx components in mechanotransduction of fluid shear stress. Biochem Biophys Res Commun. 2007;355:228–233. doi: 10.1016/j.bbrc.2007.01.137. doi: 10.1016/j.bbrc.2007.01.137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Pohl U, Herlan K, Huang A, Bassenge E. EDRF-mediated shear-induced dilation opposes myogenic vasoconstriction in small rabbit arteries. Am J Physiol. 1991;261(6)(pt 2):H2016–H2023. doi: 10.1152/ajpheart.1991.261.6.H2016. doi: 10.1152/ajpheart.1991.261.6.H2016. [DOI] [PubMed] [Google Scholar]
  • 58.Mochizuki S, Vink H, Hiramatsu O, Kajita T, Shigeto F, Spaan JA, Kajiya F. Role of hyaluronic acid glycosaminoglycans in shear-induced endothelium-derived nitric oxide release. Am J Physiol Heart Circ Physiol. 2003;285:H722–H726. doi: 10.1152/ajpheart.00691.2002. doi: 10.1152/ajpheart.00691.2002. [DOI] [PubMed] [Google Scholar]
  • 59.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–H1030. doi: 10.1152/ajpheart.00162.2007. doi: 10.1152/ajpheart.00162.2007. [DOI] [PubMed] [Google Scholar]
  • 60.Tarbell JM, Pahakis MY. Mechanotransduction and the glycocalyx. J Intern Med. 2006;259:339–350. doi: 10.1111/j.1365-2796.2006.01620.x. doi: 10.1111/j.1365-2796.2006.01620.x. [DOI] [PubMed] [Google Scholar]
  • 61.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 (Camb) 2014;6:338–347. doi: 10.1039/c3ib40199e. doi: 10.1039/c3ib40199e. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Baeyens N, Mulligan-Kehoe M, Corti F, Simon D, Ross T, Rhodes J, Wang T, Mejean C, Somins M, Humphrey J, Schwartz M. Syndecan 4 is required for endothelial alignment in flow and atheroprotective signaling. Proc Natl Acad Sci USA. 2014;111:17308–17313. doi: 10.1073/pnas.1413725111. doi: 10.1073/pnas.1413725111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Voyvodic PL, Min D, Liu R, Williams E, Chitalia V, Dunn AK, Baker AB. Loss of syndecan-1 induces a pro-inflammatory phenotype in endothelial cells with a dysregulated response to atheroprotective flow. J Biol Chem. 2014;289:9547–9559. doi: 10.1074/jbc.M113.541573. doi: 10.1074/jbc.M113.541573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Gasparski AN, Beningo KA. Mechanoreception at the cell membrane: more than the integrins. Arch Biochem Biophys. 2015;586:20–26. doi: 10.1016/j.abb.2015.07.017. doi: 10.1016/j.abb.2015.07.017. [DOI] [PubMed] [Google Scholar]
  • 65.Muller JM, Chilian WM, Davis MJ. Integrin signaling transduces shear stress–dependent vasodilation of coronary arterioles. Circ Res. 1997;80:320–326. doi: 10.1161/01.res.80.3.320. [DOI] [PubMed] [Google Scholar]
  • 66.Shyy JY, Chien S. Role of integrins in endothelial mechanosensing of shear stress. Circ Res. 2002;91:769–775. doi: 10.1161/01.res.0000038487.19924.18. [DOI] [PubMed] [Google Scholar]
  • 67.Patel HH, Murray F, Insel PA. Caveolae as organizers of pharmacologically relevant signal transduction molecules. Annu Rev Pharmacol Toxicol. 2008;48:359–391. doi: 10.1146/annurev.pharmtox.48.121506.124841. doi: 10.1146/annurev.pharmtox.48.121506.124841. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Hierck BP, Van der Heiden K, Alkemade FE, Van de Pas S, Van Thienen JV, Groenendijk BC, Bax WH, Van der Laarse A, Deruiter MC, Horrevoets AJ, Poelmann RE. Primary cilia sensitize endothelial cells for fluid shear stress. Dev Dyn. 2008;237:725–735. doi: 10.1002/dvdy.21472. doi: 10.1002/dvdy.21472. [DOI] [PubMed] [Google Scholar]
  • 69.Van der Heiden K, Groenendijk BC, Hierck BP, Hogers B, Koerten HK, Mommaas AM, Gittenberger-de Groot AC, Poelmann RE. Monocilia on chicken embryonic endocardium in low shear stress areas. Dev Dyn. 2006;235:19–28. doi: 10.1002/dvdy.20557. doi: 10.1002/dvdy.20557. [DOI] [PubMed] [Google Scholar]
  • 70.Van der Heiden K, Hierck BP, Krams R, de Crom R, Cheng C, Baiker M, Pourquie MJ, Alkemade FE, DeRuiter MC, Gittenberger-de Groot AC, Poelmann RE. Endothelial primary cilia in areas of disturbed flow are at the base of atherosclerosis. Atherosclerosis. 2008;196:542–550. doi: 10.1016/j.atherosclerosis.2007.05.030. doi: 10.1016/j.atherosclerosis.2007.05.030. [DOI] [PubMed] [Google Scholar]
  • 71.Zeng Y, Waters M, Andrews A, Honarmandi P, Ebong EE, Rizzo V, Tarbell JM. 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–H820. doi: 10.1152/ajpheart.00764.2012. doi: 10.1152/ajpheart.00764.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Liu X, Fan Y, Deng X. Mechanotransduction of flow-induced shear stress by endothelial glycocalyx fibers is torque determined. ASAIO J. 2011;57:487–494. doi: 10.1097/MAT.0b013e318233b5ed. doi: 10.1097/MAT.0b013e318233b5ed. [DOI] [PubMed] [Google Scholar]
  • 73.Fu BM, Tarbell JM. Mechano-sensing and transduction by endothelial surface glycocalyx: composition, structure, and function. Wiley Interdiscip Rev Syst Biol Med. 2013;5:381–390. doi: 10.1002/wsbm.1211. doi: 10.1002/wsbm.1211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Aruffo A, Stamenkovic I, Melnick M, Underhill CB, Seed B. CD44 is the principal cell surface receptor for hyaluronate. Cell. 1990;61:1303–1313. doi: 10.1016/0092-8674(90)90694-a. [DOI] [PubMed] [Google Scholar]
  • 75.Nandi A, Estess P, Siegelman MH. Hyaluronan anchoring and regulation on the surface of vascular endothelial cells is mediated through the functionally active form of CD44. J Biol Chem. 2000;275:14939–14948. doi: 10.1074/jbc.275.20.14939. [DOI] [PubMed] [Google Scholar]
  • 76.Singleton PA, Bourguignon LY. CD44 interaction with ankyrin and IP3 receptor in lipid rafts promotes hyaluronan-mediated Ca2+ signaling leading to nitric oxide production and endothelial cell adhesion and proliferation. Exp Cell Res. 2004;295:102–118. doi: 10.1016/j.yexcr.2003.12.025. doi: 10.1016/j.yexcr.2003.12.025. [DOI] [PubMed] [Google Scholar]
  • 77.Prehm P. Identification and regulation of the eukaryotic hyaluronate synthase. Ciba Found Symp. 1989;143:21–30; discussion 30. doi: 10.1002/9780470513774.ch3. [DOI] [PubMed] [Google Scholar]
  • 78.Weigel PH. Hyaluronan synthase: the mechanism of initiation at the reducing end and a pendulum model for polysaccharide translocation to the cell exterior. Int J Cell Biol. 2015;2015:367579. doi: 10.1155/2015/367579. doi: 10.1155/2015/367579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Toole BP. Hyaluronan and its binding proteins, the hyaladherins. Curr Opin Cell Biol. 1990;2:839–844. doi: 10.1016/0955-0674(90)90081-o. [DOI] [PubMed] [Google Scholar]
  • 80.Scott JE, Heatley F. Hyaluronan forms specific stable tertiary structures in aqueous solution: a 13c NMR Study. Proc Natl Acad Sci USA. 1999;96:4850–4855. doi: 10.1073/pnas.96.9.4850. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Desjardins C, Duling BR. Heparinase treatment suggests a role for the endothelial cell glycocalyx in regulation of capillary hematocrit. Am J Physiol. 1990;258(3)(pt 2):H647–H654. doi: 10.1152/ajpheart.1990.258.3.H647. doi: 10.1152/ajpheart.1990.258.3.H647. [DOI] [PubMed] [Google Scholar]
  • 82.Henry CB, Duling BR. Permeation of the luminal capillary glycocalyx is determined by hyaluronan. Am J Physiol. 1999;277(2)(pt 2):H508–H514. doi: 10.1152/ajpheart.1999.277.2.H508. [DOI] [PubMed] [Google Scholar]
  • 83.Chait A, Bornfeldt KE. Diabetes and atherosclerosis: is there a role for hyperglycemia? J Lipid Res. 2009;50 Suppl:S335–S339. doi: 10.1194/jlr.R800059-JLR200. doi: 10.1194/jlr.R800059-JLR200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Siracuse JJ, Chaikof EL. The pathogenesis of diabetic atherosclerosis. In: Shrikhande G, McKinsey J, editors. Diabetes and Peripheral Vascular Disease. Contemporary Diabetes. Totowa, NJ: Humana Press; 2012. pp. 13–26. [Google Scholar]
  • 85.Katakami N. Mechanism of development of atherosclerosis and cardiovascular disease in diabetes mellitus. J Atheroscler Thromb. 2018;25:27–39. doi: 10.5551/jat.RV17014. doi: 10.5551/jat.RV17014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Lopez-Quintero SV, Cancel LM, Pierides A, Antonetti D, Spray DC, Tarbell JM. High glucose attenuates shear-induced changes in endothelial hydraulic conductivity by degrading the glycocalyx. PLoS One. 2013;8:e78954. doi: 10.1371/journal.pone.0078954. doi: 10.1371/journal.pone.0078954. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.van den Berg BM, Spaan JA, Rolf TM, Vink H. Atherogenic region and diet diminish glycocalyx dimension and increase intima-to-media ratios at murine carotid artery bifurcation. Am J Physiol Heart Circ Physiol. 2006;290:H915–H920. doi: 10.1152/ajpheart.00051.2005. doi: 10.1152/ajpheart.00051.2005. [DOI] [PubMed] [Google Scholar]
  • 88.Chiu JJ, Chien S. Effects of disturbed flow on vascular endothelium: pathophysiological basis and clinical perspectives. Physiol Rev. 2011;91:327–387. doi: 10.1152/physrev.00047.2009. doi: 10.1152/physrev.00047.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Nieuwdorp M, Mooij HL, Kroon J, Atasever B, Spaan JA, Ince C, Holleman F, Diamant M, Heine RJ, Hoekstra JB, Kastelein JJ, Stroes ES, Vink H. Endothelial glycocalyx damage coincides with microalbuminuria in type 1 diabetes. Diabetes. 2006;55:1127–1132. doi: 10.2337/diabetes.55.04.06.db05-1619. [DOI] [PubMed] [Google Scholar]
  • 90.Nieuwdorp M, van Haeften TW, Gouverneur MC, Mooij HL, van Lieshout MH, Levi M, Meijers JC, Holleman F, Hoekstra JB, Vink H, Kastelein JJ, Stroes ES. Loss of endothelial glycocalyx during acute hyperglycemia coincides with endothelial dysfunction and coagulation activation in vivo. Diabetes. 2006;55:480–486. doi: 10.2337/diabetes.55.02.06.db05-1103. [DOI] [PubMed] [Google Scholar]
  • 91.Broekhuizen LN, Lemkes BA, Mooij HL, Meuwese MC, Verberne H, Holleman F, Schlingemann RO, Nieuwdorp M, Stroes ES, Vink H. Effect of sulodexide on endothelial glycocalyx and vascular permeability in patients with type 2 diabetes mellitus. Diabetologia. 2010;53:2646–2655. doi: 10.1007/s00125-010-1910-x. doi: 10.1007/s00125-010-1910-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Kumase F, Morizane Y, Mohri S, Takasu I, Ohtsuka A, Ohtsuki H. Glycocalyx degradation in retinal and choroidal capillary endothelium in rats with diabetes and hypertension. Acta Med Okayama. 2010;64:277–283. doi: 10.18926/AMO/40502. doi: 10.18926/AMO/40502. [DOI] [PubMed] [Google Scholar]
  • 93.Kim YH, Kim YS, Roh GS, Choi WS, Cho GJ. Resveratrol blocks diabetes-induced early vascular lesions and vascular endothelial growth factor induction in mouse retinas. Acta Ophthalmol. 2012;90:e31–e37. doi: 10.1111/j.1755-3768.2011.02243.x. doi: 10.1111/j.1755-3768.2011.02243.x. [DOI] [PubMed] [Google Scholar]
  • 94.Liao YJ, Ueno M, Nakagawa T, Huang C, Kanenishi K, Onodera M, Sakamoto H. Oxidative damage in cerebral vessels of diabetic db/db mice. Diabetes Metab Res Rev. 2005;21:554–559. doi: 10.1002/dmrr.579. doi: 10.1002/dmrr.579. [DOI] [PubMed] [Google Scholar]
  • 95.Nagasu H, Satoh M, Kiyokage E, Kidokoro K, Toida K, Channon KM, Kanwar YS, Sasaki T, Kashihara N. Activation of endothelial NAD(P)H oxidase accelerates early glomerular injury in diabetic mice. Lab Invest. 2016;96:25–36. doi: 10.1038/labinvest.2015.128. doi: 10.1038/labinvest.2015.128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Kadoya H, Satoh M, Haruna Y, Sasaki T, Kashihara N. Klotho attenuates renal hypertrophy and glomerular injury in Ins2Akita diabetic mice. Clin Exp Nephrol. 2016;20:671–678. doi: 10.1007/s10157-015-1202-3. doi: 10.1007/s10157-015-1202-3. [DOI] [PubMed] [Google Scholar]
  • 97.Oltean S, Qiu Y, Ferguson JK, et al. Vascular endothelial growth factor-A165b is protective and restores endothelial glycocalyx in diabetic nephropathy. J Am Soc Nephrol. 2015;26:1889–1904. doi: 10.1681/ASN.2014040350. doi: 10.1681/ASN.2014040350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Kelly R, Ruane-O’Hora T, Noble MI, Drake-Holland AJ, Snow HM. Differential inhibition by hyperglycaemia of shear stress- but not acetylcholine-mediated dilatation in the iliac artery of the anaesthetized pig. J Physiol. 2006;573(pt 1):133–145. doi: 10.1113/jphysiol.2006.106500. doi: 10.1113/jphysiol.2006.106500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Furchgott RF, Zawadzki JV. The obligatory role of endothelial cells in the relaxation of arterial smooth muscle by acetylcholine. Nature. 1980;288:373–376. doi: 10.1038/288373a0. [DOI] [PubMed] [Google Scholar]
  • 100.Paniagua OA, Bryant MB, Panza JA. Role of endothelial nitric oxide in shear stress-induced vasodilation of human microvasculature: diminished activity in hypertensive and hypercholesterolemic patients. Circulation. 2001;103:1752–1758. doi: 10.1161/01.cir.103.13.1752. [DOI] [PubMed] [Google Scholar]
  • 101.De Vriese AS, Verbeuren TJ, Van de Voorde J, Lameire NH, Vanhoutte PM. Endothelial dysfunction in diabetes. Br J Pharmacol. 2000;130:963–974. doi: 10.1038/sj.bjp.0703393. doi: 10.1038/sj.bjp.0703393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Brownlee M. The pathobiology of diabetic complications: a unifying mechanism. Diabetes. 2005;54:1615–1625. doi: 10.2337/diabetes.54.6.1615. [DOI] [PubMed] [Google Scholar]
  • 103.Lemkes BA, Nieuwdorp M, Hoekstra JB, Holleman F. The glycocalyx and cardiovascular disease in diabetes: should we judge the endothelium by its cover? Diabetes Technol Ther. 2012;14(suppl 1):S3–S10. doi: 10.1089/dia.2012.0011. doi: 10.1089/dia.2012.0011. [DOI] [PubMed] [Google Scholar]
  • 104.Lauer ME, Hascall VC, Wang A. Heparan sulfate analysis from diabetic rat glomeruli. J Biol Chem. 2007;282:843–852. doi: 10.1074/jbc.M608823200. doi: 10.1074/jbc.M608823200. [DOI] [PubMed] [Google Scholar]
  • 105.van den Born J, Pisa B, Bakker MA, Celie JW, Straatman C, Thomas S, Viberti GC, Kjellen L, Berden JH. No change in glomerular heparan sulfate structure in early human and experimental diabetic nephropathy. J Biol Chem. 2006;281:29606–29613. doi: 10.1074/jbc.M601552200. doi: 10.1074/jbc.M601552200. [DOI] [PubMed] [Google Scholar]
  • 106.Vogl-Willis CA, Edwards IJ. High-glucose-induced structural changes in the heparan sulfate proteoglycan, perlecan, of cultured human aortic endothelial cells. Biochim Biophys Acta. 2004;1672:36–45. doi: 10.1016/j.bbagen.2004.02.005. doi: 10.1016/j.bbagen.2004.02.005. [DOI] [PubMed] [Google Scholar]
  • 107.Brownlee M. Biochemistry and molecular cell biology of diabetic complications. Nature. 2001;414:813–820. doi: 10.1038/414813a. doi: 10.1038/414813a. [DOI] [PubMed] [Google Scholar]
  • 108.Satchell SC, Tooke JE. What is the mechanism of microalbuminuria in diabetes: a role for the glomerular endothelium? Diabetologia. 2008;51:714–725. doi: 10.1007/s00125-008-0961-8. doi: 10.1007/s00125-008-0961-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Sato H, Takahashi T, Ide H, Fukushima T, Tabata M, Sekine F, Kobayashi K, Negishi M, Niwa Y. Antioxidant activity of synovial fluid, hyaluronic acid, and two subcomponents of hyaluronic acid. Synovial fluid scavenging effect is enhanced in rheumatoid arthritis patients. Arthritis Rheum. 1988;31:63–71. doi: 10.1002/art.1780310110. [DOI] [PubMed] [Google Scholar]
  • 110.Kennett EC, Davies MJ. Degradation of matrix glycosaminoglycans by peroxynitrite/peroxynitrous acid: evidence for a hydroxyl-radical-like mechanism. Free Radic Biol Med. 2007;42:1278–1289. doi: 10.1016/j.freeradbiomed.2007.01.030. doi: 10.1016/j.freeradbiomed.2007.01.030. [DOI] [PubMed] [Google Scholar]
  • 111.Kennett EC, Davies MJ. Glycosaminoglycans are fragmented by hydroxyl, carbonate, and nitrogen dioxide radicals in a site-selective manner: implications for peroxynitrite-mediated damage at sites of inflammation. Free Radic Biol Med. 2009;47:389–400. doi: 10.1016/j.freeradbiomed.2009.05.002. doi: 10.1016/j.freeradbiomed.2009.05.002. [DOI] [PubMed] [Google Scholar]
  • 112.Katsumura C, Sugiyama T, Nakamura K, Obayashi H, Hasegawa G, Oku H, Ikeda T. Effects of advanced glycation end products on hyaluronan photolysis: a new mechanism of diabetic vitreopathy. Ophthalmic Res. 2004;36:327–331. doi: 10.1159/000081635. doi: 10.1159/000081635. [DOI] [PubMed] [Google Scholar]
  • 113.Hiebert LM, Han J, Mandal AK. Glycosaminoglycans, hyperglycemia, and disease. Antioxid Redox Signal. 2014;21:1032–1043. doi: 10.1089/ars.2013.5695. doi: 10.1089/ars.2013.5695. [DOI] [PubMed] [Google Scholar]
  • 114.Garsen M, Rops AL, Li J, van Beneden K, van den Branden C, Berden JH, Rabelink TJ, van der Vlag J. Endothelial nitric oxide synthase prevents heparanase induction and the development of proteinuria. PLoS One. 2016;11:e0160894. doi: 10.1371/journal.pone.0160894. doi: 10.1371/journal.pone.0160894. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Ikegami-Kawai M, Okuda R, Nemoto T, Inada N, Takahashi T. Enhanced activity of serum and urinary hyaluronidases in streptozotocin-induced diabetic Wistar and GK rats. Glycobiology. 2004;14:65–72. doi: 10.1093/glycob/cwh011. doi: 10.1093/glycob/cwh011. [DOI] [PubMed] [Google Scholar]
  • 116.Nieuwdorp M, Holleman F, de Groot E, Vink H, Gort J, Kontush A, Chapman MJ, Hutten BA, Brouwer CB, Hoekstra JB, Kastelein JJ, Stroes ES. Perturbation of hyaluronan metabolism predisposes patients with type 1 diabetes mellitus to atherosclerosis. Diabetologia. 2007;50:1288–1293. doi: 10.1007/s00125-007-0666-4. doi: 10.1007/s00125-007-0666-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Stern R, Jedrzejas MJ. Hyaluronidases: their genomics, structures, and mechanisms of action. Chem Rev. 2006;106:818–839. doi: 10.1021/cr050247k. doi: 10.1021/cr050247k. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Vigdorovich V, Strong RK, Miller AD. Expression and characterization of a soluble, active form of the jaagsiekte sheep retrovirus receptor, Hyal2. J Virol. 2005;79:79–86. doi: 10.1128/JVI.79.1.79-86.2005. doi: 10.1128/JVI.79.1.79-86.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Natowicz MR, Short MP, Wang Y, Dickersin GR, Gebhardt MC, Rosenthal DI, Sims KB, Rosenberg AE. Clinical and biochemical manifestations of hyaluronidase deficiency. N Engl J Med. 1996;335:1029–1033. doi: 10.1056/NEJM199610033351405. doi: 10.1056/NEJM199610033351405. [DOI] [PubMed] [Google Scholar]
  • 120.Stern R. Devising a pathway for hyaluronan catabolism: are we there yet? Glycobiology. 2003;13:105R–115R. doi: 10.1093/glycob/cwg112. doi: 10.1093/glycob/cwg112. [DOI] [PubMed] [Google Scholar]
  • 121.Stern R. Hyaluronan catabolism: a new metabolic pathway. Eur J Cell Biol. 2004;83:317–325. doi: 10.1078/0171-9335-00392. doi: 10.1078/0171-9335-00392. [DOI] [PubMed] [Google Scholar]
  • 122.Puissant E, Gilis F, Dogné S, Flamion B, Jadot M, Boonen M. Subcellular trafficking and activity of Hyal-1 and its processed forms in murine macrophages. Traffic. 2014;15:500–515. doi: 10.1111/tra.12162. doi: 10.1111/tra.12162. [DOI] [PubMed] [Google Scholar]
  • 123.McAtee CO, Berkebile AR, Elowsky CG, Fangman T, Barycki JJ, Wahl JK, III, Khalimonchuk O, Naslavsky N, Caplan S, Simpson MA. Hyaluronidase Hyal1 increases tumor cell proliferation and motility through accelerated vesicle trafficking. J Biol Chem. 2015;290:13144–13156. doi: 10.1074/jbc.M115.647446. doi: 10.1074/jbc.M115.647446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Andre B, Duterme C, Van Moer K, Mertens-Strijthagen J, Jadot M, Flamion B. Hyal2 is a glycosylphosphatidylinositol-anchored, lipid raft-associated hyaluronidase. Biochem Biophys Res Commun. 2011;411:175–179. doi: 10.1016/j.bbrc.2011.06.125. doi: 10.1016/j.bbrc.2011.06.125. [DOI] [PubMed] [Google Scholar]
  • 125.Onclinx C, Dogne S, Jadin L, Andris F, Grandfils C, Jouret F, Mullier F, Flamion B. Deficiency in mouse hyaluronidase 2: a new mechanism of chronic thrombotic microangiopathy. Haematologica. 2015;100:1023–1030. doi: 10.3324/haematol.2015.123828. doi: 10.3324/haematol.2015.123828. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Grundmann S, Fink K, Rabadzhieva L, Bourgeois N, Schwab T, Moser M, Bode C, Busch HJ. Perturbation of the endothelial glycocalyx in post cardiac arrest syndrome. Resuscitation. 2012;83:715–720. doi: 10.1016/j.resuscitation.2012.01.028. doi: 10.1016/j.resuscitation.2012.01.028. [DOI] [PubMed] [Google Scholar]
  • 127.Li J, Yuan T, Zhao X, Lv GY, Liu HQ. Protective effects of sevoflurane in hepatic ischemia-reperfusion injury. Int J Immunopathol Pharmacol. 2016;29:300–307. doi: 10.1177/0394632016638346. doi: 10.1177/0394632016638346. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Steppan J, Hofer S, Funke B, Brenner T, Henrich M, Martin E, Weitz J, Hofmann U, Weigand MA. Sepsis and major abdominal surgery lead to flaking of the endothelial glycocalix. J Surg Res. 2011;165:136–141. doi: 10.1016/j.jss.2009.04.034. doi: 10.1016/j.jss.2009.04.034. [DOI] [PubMed] [Google Scholar]
  • 129.Henrich M, Gruss M, Weigand MA. Sepsis-induced degradation of endothelial glycocalix. ScientificWorldJournal. 2010;10:917–923. doi: 10.1100/tsw.2010.88. doi: 10.1100/tsw.2010.88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Anand D, Ray S, Srivastava LM, Bhargava S. Evolution of serum hyaluronan and syndecan levels in prognosis of sepsis patients. Clin Biochem. 2016;49:768–776. doi: 10.1016/j.clinbiochem.2016.02.014. doi: 10.1016/j.clinbiochem.2016.02.014. [DOI] [PubMed] [Google Scholar]
  • 131.Yini S, Heng Z, Xin A, Xiaochun M. Effect of unfractionated heparin on endothelial glycocalyx in a septic shock model. Acta Anaesthesiol Scand. 2015;59:160–169. doi: 10.1111/aas.12418. doi: 10.1111/aas.12418. [DOI] [PubMed] [Google Scholar]
  • 132.Torres Filho IP, Torres LN, Salgado C, Dubick MA. Plasma syndecan-1 and heparan sulfate correlate with microvascular glycocalyx degradation in hemorrhaged rats after different resuscitation fluids. Am J Physiol Heart Circ Physiol. 2016;310:H1468–H1478. doi: 10.1152/ajpheart.00006.2016. doi: 10.1152/ajpheart.00006.2016. [DOI] [PubMed] [Google Scholar]
  • 133.Majerczak J, Grandys M, Duda K, Zakrzewska A, Balcerczyk A, Kolodziejski L, Szymoniak-Chochol D, Smolenski RT, Bartosz G, Chlopicki S, Zoladz JA. Moderate-intensity endurance training improves endothelial glycocalyx layer integrity in healthy young men. Exp Physiol. 2017;102:70–85. doi: 10.1113/EP085887. doi: 10.1113/EP085887. [DOI] [PubMed] [Google Scholar]
  • 134.Padberg JS, Wiesinger A, di Marco GS, Reuter S, Grabner A, Kentrup D, Lukasz A, Oberleithner H, Pavenstädt H, Brand M, Kümpers P. Damage of the endothelial glycocalyx in chronic kidney disease. Atherosclerosis. 2014;234:335–343. doi: 10.1016/j.atherosclerosis.2014.03.016. doi: 10.1016/j.atherosclerosis.2014.03.016. [DOI] [PubMed] [Google Scholar]
  • 135.Vlahu CA, Lemkes BA, Struijk DG, Koopman MG, Krediet RT, Vink H. Damage of the endothelial glycocalyx in dialysis patients. J Am Soc Nephrol. 2012;23:1900–1908. doi: 10.1681/ASN.2011121181. doi: 10.1681/ASN.2011121181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Malathy K, Kurup PA. Levels of glycosaminoglycans in the serum of diabetic patients. Indian J Biochem Biophys. 1973;10:132–133. [PubMed] [Google Scholar]
  • 137.Ceriello A, Giugliano D, Dello Russo P, Passariello N, Saccomanno F, Sgambato S. Glycosaminoglycans in human diabetes. Diabete Metab. 1983;9:32–34. [PubMed] [Google Scholar]
  • 138.Olczyk K. Glycosaminoglycans of human serum and their alterations in diabetes mellitus. Acta Biochimica Polonica. 1996;43:567–574. [PubMed] [Google Scholar]
  • 139.Wang JB, Zhang YJ, Zhang Y, Guan J, Chen LY, Fu CH, Du HJ, Sheng Y, Zhou L, Si YF, Zhang Y. Negative correlation between serum syndecan-1 and apolipoprotein A1 in patients with type 2 diabetes mellitus. Acta Diabetol. 2013;50:111–115. doi: 10.1007/s00592-010-0216-2. doi: 10.1007/s00592-010-0216-2. [DOI] [PubMed] [Google Scholar]
  • 140.Kolseth IB, Reine TM, Parker K, Sudworth A, Witczak BJ, Jenssen TG, Kolset SO. Increased levels of inflammatory mediators and proinflammatory monocytes in patients with type I diabetes mellitus and nephropathy. J Diabetes Complications. 2017;31:245–252. doi: 10.1016/j.jdiacomp.2016.06.029. doi: 10.1016/j.jdiacomp.2016.06.029. [DOI] [PubMed] [Google Scholar]
  • 141.Svennevig K, Kolset SO, Bangstad HJ. Increased syndecan-1 in serum is related to early nephropathy in type 1 diabetes mellitus patients. Diabetologia. 2006;49:2214–2216. doi: 10.1007/s00125-006-0330-4. doi: 10.1007/s00125-006-0330-4. [DOI] [PubMed] [Google Scholar]
  • 142.Broekhuizen LN, Mooij HL, Kastelein JJ, Stroes ES, Vink H, Nieuwdorp M. Endothelial glycocalyx as potential diagnostic and therapeutic target in cardiovascular disease. Curr Opin Lipidol. 2009;20:57–62. doi: 10.1097/MOL.0b013e328321b587. doi: 10.1097/MOL.0b013e328321b587. [DOI] [PubMed] [Google Scholar]
  • 143.Dunlop ME, Clark S, Mahadevan P, Muggli E, Larkins RG. Production of hyaluronan by glomerular mesangial cells in response to fibronectin and platelet-derived growth factor. Kidney Int. 1996;50:40–44. doi: 10.1038/ki.1996.284. [DOI] [PubMed] [Google Scholar]
  • 144.Wang A, Hascall VC. Hyaluronan structures synthesized by rat mesangial cells in response to hyperglycemia induce monocyte adhesion. J Biol Chem. 2004;279:10279–10285. doi: 10.1074/jbc.M312045200. doi: 10.1074/jbc.M312045200. [DOI] [PubMed] [Google Scholar]
  • 145.Wang A, Ren J, Wang CP, Hascall VC. Heparin prevents intracellular hyaluronan synthesis and autophagy responses in hyperglycemic dividing mesangial cells and activates synthesis of an extensive extracellular monocyte-adhesive hyaluronan matrix after completing cell division. J Biol Chem. 2014;289:9418–9429. doi: 10.1074/jbc.M113.541441. doi: 10.1074/jbc.M113.541441. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.McDonald TO, Gerrity RG, Jen C, Chen HJ, Wark K, Wight TN, Chait A, O’Brien KD. Diabetes and arterial extracellular matrix changes in a porcine model of atherosclerosis. J Histochem Cytochem. 2007;55:1149–1157. doi: 10.1369/jhc.7A7221.2007. doi: 10.1369/jhc.7A7221.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Evanko SP, Raines EW, Ross R, Gold LI, Wight TN. Proteoglycan distribution in lesions of atherosclerosis depends on lesion severity, structural characteristics, and the proximity of platelet-derived growth factor and transforming growth factor-beta. Am J Pathol. 1998;152:533–546. [PMC free article] [PubMed] [Google Scholar]
  • 148.Vendrov AE, Madamanchi NR, Niu XL, Molnar KC, Runge M, Szyndralewiez C, Page P, Runge MS. NADPH oxidases regulate CD44 and hyaluronic acid expression in thrombin-treated vascular smooth muscle cells and in atherosclerosis. J Biol Chem. 2010;285:26545–26557. doi: 10.1074/jbc.M110.143917. doi: 10.1074/jbc.M110.143917. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Lorentzen KA, Chai S, Chen H, Danielsen CC, Simonsen U, Wogensen L. Mechanisms involved in extracellular matrix remodeling and arterial stiffness induced by hyaluronan accumulation. Atherosclerosis. 2016;244:195–203. doi: 10.1016/j.atherosclerosis.2015.11.016. doi: 10.1016/j.atherosclerosis.2015.11.016. [DOI] [PubMed] [Google Scholar]
  • 150.Seike M, Ikeda M, Matsumoto M, Hamada R, Takeya M, Kodama H. Hyaluronan forms complexes with low density lipoprotein while also inducing foam cell infiltration in the dermis. J Dermatol Sci. 2006;41:197–204. doi: 10.1016/j.jdermsci.2005.10.008. doi: 10.1016/j.jdermsci.2005.10.008. [DOI] [PubMed] [Google Scholar]
  • 151.Vigetti D, Viola M, Karousou E, Genasetti A, Rizzi M, Clerici M, Bartolini B, Moretto P, De Luca G, Passi A. Vascular pathology and the role of hyaluronan. ScientificWorldJournal. 2008;8:1116–1118. doi: 10.1100/tsw.2008.145. doi: 10.1100/tsw.2008.145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Nagy N, Freudenberger T, Melchior-Becker A, Röck K, Ter Braak M, Jastrow H, Kinzig M, Lucke S, Suvorava T, Kojda G, Weber AA, Sörgel F, Levkau B, Ergün S, Fischer JW. Inhibition of hyaluronan synthesis accelerates murine atherosclerosis: novel insights into the role of hyaluronan synthesis. Circulation. 2010;122:2313–2322. doi: 10.1161/CIRCULATIONAHA.110.972653. doi: 10.1161/CIRCULATIONAHA.110.972653. [DOI] [PubMed] [Google Scholar]
  • 153.Kolářová H, Ambrůzová B, Svihálková Šindlerová L, Klinke A, Kubala L. Modulation of endothelial glycocalyx structure under inflammatory conditions. Mediators Inflamm. 2014;2014:694312. doi: 10.1155/2014/694312. doi: 10.1155/2014/694312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Kozar RA, Peng Z, Zhang R, Holcomb JB, Pati S, Park P, Ko TC, Paredes A. Plasma restoration of endothelial glycocalyx in a rodent model of hemorrhagic shock. Anesth Analg. 2011;112:1289–1295. doi: 10.1213/ANE.0b013e318210385c. doi: 10.1213/ANE.0b013e318210385c. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Jacob M, Paul O, Mehringer L, Chappell D, Rehm M, Welsch U, Kaczmarek I, Conzen P, Becker BF. Albumin augmentation improves condition of guinea pig hearts after 4 hr of cold ischemia. Transplantation. 2009;87:956–965. doi: 10.1097/TP.0b013e31819c83b5. doi: 10.1097/TP.0b013e31819c83b5. [DOI] [PubMed] [Google Scholar]
  • 156.Zeng Y, Adamson RH, Curry FR, Tarbell JM. Sphingosine-1-phosphate protects endothelial glycocalyx by inhibiting syndecan-1 shedding. Am J Physiol Heart Circ Physiol. 2014;306:H363–H372. doi: 10.1152/ajpheart.00687.2013. doi: 10.1152/ajpheart.00687.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Singh A, Ramnath RD, Foster RR, Wylie EC, Fridén V, Dasgupta I, Haraldsson B, Welsh GI, Mathieson PW, Satchell SC. Reactive oxygen species modulate the barrier function of the human glomerular endothelial glycocalyx. PLoS One. 2013;8:e55852. doi: 10.1371/journal.pone.0055852. doi: 10.1371/journal.pone.0055852. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Vink H, Constantinescu AA, Spaan JA. Oxidized lipoproteins degrade the endothelial surface layer: implications for platelet-endothelial cell adhesion. Circulation. 2000;101:1500–1502. doi: 10.1161/01.cir.101.13.1500. [DOI] [PubMed] [Google Scholar]
  • 159.Lipowsky HH. The endothelial glycocalyx as a barrier to leukocyte adhesion and its mediation by extracellular proteases. Ann Biomed Eng. 2012;40:840–848. doi: 10.1007/s10439-011-0427-x. doi: 10.1007/s10439-011-0427-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Chappell D, Jacob M, Hofmann-Kiefer K, Rehm M, Welsch U, Conzen P, Becker BF. Antithrombin reduces shedding of the endothelial glycocalyx following ischaemia/reperfusion. Cardiovasc Res. 2009;83:388–396. doi: 10.1093/cvr/cvp097. doi: 10.1093/cvr/cvp097. [DOI] [PubMed] [Google Scholar]
  • 161.Chappell D, Jacob M, Hofmann-Kiefer K, Bruegger D, Rehm M, Conzen P, Welsch U, Becker BF. Hydrocortisone preserves the vascular barrier by protecting the endothelial glycocalyx. Anesthesiology. 2007;107:776–784. doi: 10.1097/01.anes.0000286984.39328.96. doi: 10.1097/01.anes.0000286984.39328.96. [DOI] [PubMed] [Google Scholar]
  • 162.Chappell D, Dörfler N, Jacob M, Rehm M, Welsch U, Conzen P, Becker BF. Glycocalyx protection reduces leukocyte adhesion after ischemia/reperfusion. Shock. 2010;34:133–139. doi: 10.1097/SHK.0b013e3181cdc363. doi: 10.1097/SHK.0b013e3181cdc363. [DOI] [PubMed] [Google Scholar]
  • 163.Salmon AH, Ferguson JK, Burford JL, Gevorgyan H, Nakano D, Harper SJ, Bates DO, Peti-Peterdi J. Loss of the endothelial glycocalyx links albuminuria and vascular dysfunction. J Am Soc Nephrol. 2012;23:1339–1350. doi: 10.1681/ASN.2012010017. doi: 10.1681/ASN.2012010017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Meuwese MC, Mooij HL, Nieuwdorp M, van Lith B, Marck R, Vink H, Kastelein JJ, Stroes ES. Partial recovery of the endothelial glycocalyx upon rosuvastatin therapy in patients with heterozygous familial hypercholesterolemia. J Lipid Res. 2009;50:148–153. doi: 10.1194/jlr.P800025-JLR200. doi: 10.1194/jlr.P800025-JLR200. [DOI] [PubMed] [Google Scholar]
  • 165.Packham DK, Wolfe R, Reutens AT, Berl T, Heerspink HL, Rohde R, Ivory S, Lewis J, Raz I, Wiegmann TB, Chan JC, de Zeeuw D, Lewis EJ, Atkins RC Collaborative Study Group. Sulodexide fails to demonstrate renoprotection in overt type 2 diabetic nephropathy. J Am Soc Nephrol. 2012;23:123–130. doi: 10.1681/ASN.2011040378. doi: 10.1681/ASN.2011040378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Garsen M, Lenoir O, Rops AL, Dijkman HB, Willemsen B, van Kuppevelt TH, Rabelink TJ, Berden JH, Tharaux PL, van der Vlag J. Endothelin-1 induces proteinuria by heparanase-mediated disruption of the glomerular glycocalyx. J Am Soc Nephrol. 2016;27:3545–3551. doi: 10.1681/ASN.2015091070. doi: 10.1681/ASN.2015091070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.VanTeeffelen JW, Brands J, Jansen C, Spaan JA, Vink H. Heparin impairs glycocalyx barrier properties and attenuates shear dependent vasodilation in mice. Hypertension. 2007;50:261–267. doi: 10.1161/HYPERTENSIONAHA.107.089250. doi: 10.1161/HYPERTENSIONAHA.107.089250. [DOI] [PubMed] [Google Scholar]
  • 168.VanTeeffelen JW, Brands J, Janssen BJ, Vink H. Effect of acute hyaluronidase treatment of the glycocalyx on tracer-based whole body vascular volume estimates in mice. J Appl Physiol (1985) 2013;114:1132–1140. doi: 10.1152/japplphysiol.00842.2012. doi: 10.1152/japplphysiol.00842.2012. [DOI] [PubMed] [Google Scholar]
  • 169.Meuwese MC, Broekhuizen LN, Kuikhoven M, Heeneman S, Lutgens E, Gijbels MJ, Nieuwdorp M, Peutz CJ, Stroes ES, Vink H, van den Berg BM. Endothelial surface layer degradation by chronic hyaluronidase infusion induces proteinuria in apolipoprotein E-deficient mice. PLoS One. 2010;5:e14262. doi: 10.1371/journal.pone.0014262. doi: 10.1371/journal.pone.0014262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Galie PA, van Oosten A, Chen CS, Janmey PA. Application of multiple levels of fluid shear stress to endothelial cells plated on polyacrylamide gels. Lab Chip. 2015;15:1205–1212. doi: 10.1039/c4lc01236d. doi: 10.1039/c4lc01236d. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Foster RR, Armstrong L, Baker S, Wong DW, Wylie EC, Ramnath R, Jenkins R, Singh A, Steadman R, Welsh GI, Mathieson PW, Satchell SC. Glycosaminoglycan regulation by VEGFA and VEGFC of the glomerular microvascular endothelial cell glycocalyx in vitro. Am J Pathol. 2013;183:604–616. doi: 10.1016/j.ajpath.2013.04.019. doi: 10.1016/j.ajpath.2013.04.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Kumagai R, Lu X, Kassab GS. Role of glycocalyx in flow-induced production of nitric oxide and reactive oxygen species. Free Radic Biol Med. 2009;47:600–607. doi: 10.1016/j.freeradbiomed.2009.05.034. doi: 10.1016/j.freeradbiomed.2009.05.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Dragovich MA, Chester D, Fu BM, Wu C, Xu Y, Goligorsky MS, Zhang XF. Mechanotransduction of the endothelial glycocalyx mediates nitric oxide production through activation of trp channels. Am J Physiol Cell Physiol. 2016;311:C846–C853. doi: 10.1152/ajpcell.00288.2015. doi: 10.1152/ajpcell.00288.2015. [DOI] [PubMed] [Google Scholar]
  • 174.Ramiro-Diaz J, Barajas-Espinosa A, Chi-Ahumada E, Perez-Aguilar S, Torres-Tirado D, Castillo-Hernandez J, Knabb M, de Rosa AB, Rubio R. Luminal endothelial lectins with affinity for N-acetylglucosamine determine flow-induced cardiac and vascular paracrine-dependent responses. Am J Physiol Heart Circ Physiol. 2010;299:H743–H751. doi: 10.1152/ajpheart.00790.2009. doi: 10.1152/ajpheart.00790.2009. [DOI] [PubMed] [Google Scholar]
  • 175.VanTeeffelen JW, Constantinescu AA, Vink H, Spaan JA. Hypercholesterolemia impairs reactive hyperemic vasodilation of 2A but not 3A arterioles in mouse cremaster muscle. Am J Physiol Heart Circ Physiol. 2005;289:H447–H454. doi: 10.1152/ajpheart.01298.2004. doi: 10.1152/ajpheart.01298.2004. [DOI] [PubMed] [Google Scholar]
  • 176.Yung S, Chau MK, Zhang Q, Zhang CZ, Chan TM. Sulodexide decreases albuminuria and regulates matrix protein accumulation in C57BL/6 mice with streptozotocin-induced type I diabetic nephropathy. PLoS One. 2013;8:e54501. doi: 10.1371/journal.pone.0054501. doi: 10.1371/journal.pone.0054501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Achour A, Kacem M, Dibej K, Skhiri H, Bouraoui S, El May M. One year course of oral sulodexide in the management of diabetic nephropathy. J Nephrol. 2005;18:568–574. [PubMed] [Google Scholar]
  • 178.Eskens BJ, Zuurbier CJ, van Haare J, Vink H, van Teeffelen JW. Effects of two weeks of metformin treatment on whole-body glycocalyx barrier properties in db/db mice. Cardiovasc Diabetol. 2013;12:175. doi: 10.1186/1475-2840-12-175. doi: 10.1186/1475-2840-12-175. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Arteriosclerosis, Thrombosis, and Vascular Biology are provided here courtesy of Wolters Kluwer Health

RESOURCES