Next Article in Journal
Sex Specificity in the Mixed Effects of Blood Heavy Metals and Cognitive Function on Elderly: Evidence from NHANES
Previous Article in Journal
Metabolomic Profiling in Children with Celiac Disease: Beyond the Gluten-Free Diet
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Glycocalyx–Sodium Interaction in Vascular Endothelium

1
Department of Medical Physiology, Makerere University College of Health Sciences, Kampala P.O. Box 7072, Uganda
2
Department of Obstetrics and Gynecology, School of Medicine, Makerere University College of Health Sciences, Kampala P.O. Box 7072, Uganda
3
Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
4
Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
5
Department of Physiological Sciences, School of Medicine and Health Sciences, Mulungushi University, Kabwe P.O. Box 80415, Zambia
*
Authors to whom correspondence should be addressed.
Nutrients 2023, 15(13), 2873; https://doi.org/10.3390/nu15132873
Submission received: 20 May 2023 / Revised: 19 June 2023 / Accepted: 22 June 2023 / Published: 25 June 2023
(This article belongs to the Special Issue Cardiovascular Disease, Immune System and Nutrition)

Abstract

:
The glycocalyx generally covers almost all cellular surfaces, where it participates in mediating cell-surface interactions with the extracellular matrix as well as with intracellular signaling molecules. The endothelial glycocalyx that covers the luminal surface mediates the interactions of endothelial cells with materials flowing in the circulating blood, including blood cells. Cardiovascular diseases (CVD) remain a major cause of morbidity and mortality around the world. The cardiovascular risk factors start by causing endothelial cell dysfunction associated with destruction or irregular maintenance of the glycocalyx, which may culminate into a full-blown cardiovascular disease. The endothelial glycocalyx plays a crucial role in shielding the cell from excessive exposure and absorption of excessive salt, which can potentially cause damage to the endothelial cells and underlying tissues of the blood vessels. So, in this mini review/commentary, we delineate and provide a concise summary of the various components of the glycocalyx, their interaction with salt, and subsequent involvement in the cardiovascular disease process. We also highlight the major components of the glycocalyx that could be used as disease biomarkers or as drug targets in the management of cardiovascular diseases.

1. Background

The blood vascular system consists of a network of vessels ranging from as large as the aorta, arteries, and veins to the smaller units including arterioles, venules, and capillaries [1]. With the exception of the capillaries whose wall is made up of a single layer of endothelial cells, the other vessels have a wall with several tissue layers called tunics: tunica interna, tunica media, and tunica adventitia/externa [2]. The innermost tissue layer in the wall of the blood vessels is the luminal surface layer, consisting of continuously arranged endothelial cells [3]. The single layer of endothelial cells making up the capillaries is interrupted with some fenestrations or pores in some tissues such as in the gastrointestinal tract to enable the transportation of molecules across the capillary wall [3,4,5]. The luminal surface of endothelial cells is lined with a meshwork consisting of glycoproteins, proteoglycans, and hyaluronic acid/hyaluronan (HA), forming a gel-like material known as the endothelial glycocalyx [6,7]. The main components of the endothelial glycocalyx therefore include cell surface heparan sulfate proteoglycans (HSPGs) with associated chondroitin sulfate (CS) polysaccharide chains in certain syndecans (SDCs) and hyaluronic acid/hyaluronan (HA) [8]. The HA in the glycocalyx helps to reinforce its thickness and provides it with a gel-like appearance on the surface of the endothelial cells [9]. The endothelial-associated HSPGs consist of syndecans (SDCs) and glypicans (GPCs) that are located on the cell membranes [10] and perlecan secreted into the extracellular matrix with a pericellular localization near the basement membrane [10,11].
The glycocalyx plays an important protective role on the tissues that it covers, especially the epithelial and endothelial surfaces [12]. For instance, the endothelial glycocalyx has been reported to act as a salt-buffer on the endothelial surfaces, preventing the rapid absorption of sodium chloride (NaCl) from plasma and, thus, protecting the endothelium and the underlying tissues from the damaging effects of direct exposure to excessive amounts of salt [13]. An intact glycocalyx is also essential for the normal structure or function of endothelial cells, and endothelial dysfunction is mainly associated with old age, as well as pathological conditions including inflammatory conditions, atherosclerosis, hypertension, and heart failure [14]. The extensive sulfation of the glycosaminoglycan (GAG) chains among the proteoglycans within the endothelial glycocalyx increases its negativity that allows it to bind plasma proteins such as albumin [15]. The major components of the endothelial glycocalyx that project into the vascular lumen are also involved in flow-mediated “mechanosensation-mechanotranduction” that affects nitric oxide synthesis and vasodilation [16]. In addition, the glycocalyx is involved in endothelial cell calcium dynamics as the mechanotransduction processes are mediated by the glycocalyx components are believed to either increase the calcium permeability into the cells or its release from the endoplasmic reticulum reservoirs [17]. The mechanotransduction of the luminal shear stress due to the increased blood flow into various chemical processes in the endothelial cells, involves the interaction of the glycocalyx with both membrane and cytoskeletal proteins [18].
Glycocalyx homeostasis is very important in a sense that there is constant biosynthesis and degradation of its components to maintain a balanced cell surface layer [9]. Deviations from this balance either through excessive degradation or biosynthesis of the glycocalyx has been associated with various disease states [19,20]. The excessive degradation of the glycocalyx has been linked to disease conditions including cardiovascular diseases, hypertension, diabetes, and some cancers [21]. On the other hand, increased biosynthesis of the glycocalyx components has been observed and associated with various types of cancers [21,22,23,24]. There are certain enzymes that are generally implicated in the degradation of components of the glycocalyx including: matrix metalloproteinases (MMPs), ADAM-17 (a disintegrin and metalloproteinase), and ‘sheddases’ such as secretases, heparanases, and hyaluronidases (HYAL 1to 3) [25,26,27]. Circulating levels of the glycocalyx degradation enzymes and/or levels of these components of the glycocalyx such as syndecans or HA in plasma may be used to gauge the status of the endothelial glycocalyx in various disease states [28]. Thus, prolonged cardio-pulmonary bipass in individuals undergoing cardiac surgery has been associated with increased glycocalyx degradation based on the increased levels of syndecan-1 in circulation [29].
This mini-review is intended to provide a concise summary of the literature showing the interactions between the endothelial and glycocalyx or its components with salt, strategies to reduce the damaging effects of excessive sodium chloride (NaCl) on the endothelium, and the potential use of the glycocalyx as a biomarker of the endothelial status in cardiovascular-related conditions.

2. Major Components of the Glycocalyx in CVD Pathophysiology

The components of the glycocalyx involved in CVD pathophysiology include membrane-bound heparan sulfate proteoglycans (such as syndecans and glypicans), CD44, and a membrane-associated polysaccharide called hyaluronic acid (or hyaluronan) (Figure 1).

2.1. Heparan Sulfate Proteoglycans (Syndecans and Glypicans)

The cellular biosynthesis of the HSPGs was extensively well reviewed previously [30], so we will not provide any additional details here.

2.1.1. Syndecans

Syndecans belong to a family of single-pass transmembrane proteoglycans with three major domains: an ectodomain on the outside, a transmembrane domain, and a cytoplasmic domain on the inner side of the plasma membrane [31]. The ectodomain is modified with conjugated glycosaminoglycan (GAG) chains in form of either heparan sulfate (HS) or chondroitin sulfate (CS) chains or both, which enable syndecans to act as cell surface co-receptors [32]. Heparan sulfate chains are the major GAG in syndencans, but CS chains are also found on syndecans 1 and 3 [33]. The GAG chains are conjugated to the ectodomain of a core protein, expressed by any of the four mammalian genes that encode for syndecans 1 to 4 (SDCs 1 to 4) [34]. The GAG-attachment site on the ectodomain is at both ends in the case of SDC-1 and SDC-3, but it is near the terminal end for SDC-2 and SDC-4 [35]. The presence of cleavage sites on the ectodomains of syndecans renders them liable to being ‘slashed’ from the plasma membrane by ‘sheddases’ (proteolytic enzymes such as matrix metalloproteinases) and released into plasma (becoming ‘solubilized’ as soluble syndecans), which may be potentially utilized as biomarkers for various diseases [35,36,37]. The HS and CS chains are modified with unique sulfation patterns along their lengths, making them highly negatively charged and enabling them to bind with a wide range of positively charged particles including positively charged peptides (e.g., growth factors) and electrolytes including sodium ions. This configuration renders the entire cell surface glycocalyx with an overall negative charge, which serves a protective function for the epithelial and endothelial cells by binding the sodium ions and preventing the cells from damage due to excessive exposure to sodium ions.
Although no major mutations in syndecans have been reported that are associated with specific diseases, single nucleotide polymorphisms in the syndecans SDC-3 and SDC-4 are linked with lipid metabolism dysregulation [38]. The single nucleotide polymorphisms (SNPs) in the SDC-3 core-protein ectodomain (i.e., rs2282440 and rs2491132) were positively associated with obesity among Koreans [39] and with metabolic syndrome among Taiwanese people [40,41], suggesting that SDC-3 could be an important genetic modifier in the cardiovascular disease risk factors; whereas, in a cardiovascular risk study conducted among Finnish adults, the SNPs in SDC-4 were associated with obesity, hypertension, and an increased prevalence of coronary artery disease [42]. Moreover, the endothelial cells that were subjected to abnormal physical forces such as enhanced shear stress due to rapid blood flow during hypertension can cause an increased expression of SDC-4 alongside other extracellular matrix molecules [43]. Syndecans are believed to be involved in cardiac fibrosis, which is a key step in heart disease [44]. In addition, it has been demonstrated in mice that the overexpression of syndecan-4 can lead to an increased activation of Calcineurin-NFAT-dependent signaling, which exacerbates hypertrophy in an overloaded heart model [45]. Increased shedding of the endothelial cell surface proteoglycans occurs during the cardiovascular disease process, and elevated levels of some of the components of the glycocalyx such as syndecan-1 have been observed [46,47]. This has led to the suggestion that these altered levels of syndecan-1 may be potential prognostic biomarkers for patients suffering from ischemic heart disease and heart failure [48,49].

2.1.2. Glypicans

Glypicans are membrane-bound or cell surface heparan sulfate proteoglycans with one or two heparan sulfate chains and a glycosylphosphatidylinositol anchor that attaches the entire molecule to the plasma membrane [50]. This group of molecules consists of six members, from glypican-1 to -6, which are expressed predominantly during embryonic development [51]. They are involved in facilitating Wnt, hedgehog, fibroblast growth factor, and bone-morphogenic proteins/peptide signaling [51,52,53]. Their GPI anchor is cleaved by the expression of an enzyme known as Notum that releases not only glypicans but also other GPI-anchored proteins from the cell surface [54]. Glypicans are a major component of the glycocalyx alongside syndecans, which undergo significant remodeling by sheer stress on the endothelial surface during atherosclerosis [55]. Glypican- 1 is the main member among the glypican family that has been confirmed to be associated with the endothelial glycocalyx, lining the luminal surface of blood vessels [30]. Glypican-1 is believed to be involved in the mechanosensation leading to shear-induced endothelial nitric oxide synthesis, which is required for improving the compliance of blood vessels through vasodilation [56].

2.2. Hyaluronan

Hyaluronan is a long polysaccharide of alternating glucuronic acid (GlcA) and N-acetylglucosamine (GlcNAc) sugar residues, which are un-sulfated but result in a highly negatively charged molecule [57]. It is pericellularly synthesized by a family of three pericellular enzymes known as hyaluronan synthases: from HAS-1 to -3 [58,59]. It is then degraded by a family of six hyaluronidase (HYALs) members, of which HYAL-1 and HYAL-2 are the most potent enzymes [60,61]. HYAL-2 is believed to have a role in promoting pulmonary vascular remolding and pulmonary hypertension [61]. The deficiency of HYAL-3 increases collagen deposition, promoting post-myocardial infarction fibrosis [61]. Hyaluronan is most known as an extracellular matrix molecule, but, due to its close association with various cell surface structures including its cognate receptor CD44, integrins, and proteoglycans, it contributes to the surface gel-like glycocalyx of the synthesizing cell [62]. The hyaluronan receptor-CD44 can also be cleaved from the cell surface by proteases such as ADAM-10, ADAM-17, and MMP-14 [63,64]. A delicate balance is maintained normally between hyaluronan bio-synthesis and degradation depending on the activity of HASs and HYL enzymes, which varies in different physiological and pathological conditions [62]. The hyaluronan synthesis and degradation processes are indeed crucial during cardiovascular development or angiogenesis [65,66]. Native hyaluronan is believed to inhibit angiogenesis, whereas oligosaccharides formed following hyaluronan degradation seem to promote angiogenesis by increasing endothelial cell proliferation and migration [67,68]. The interactions of hyaluronan with reactive oxygen or nitrogen species (ROS/RNS) and their consequences on tissue homeostasis are well summarized by Berdiaki et al., whereby the innate high molecular weight hyaluronan is anti-angiogenic, anti-inflammatory, and anti-oncogenic [69]. However, following degradation by the ROS/RNS, the low molecular weight oligosaccharides produced become pro-angiogenic, pro-inflammatory, and oncogenic [69]. Changes in hyaluronan synthesis and degradation have mainly been associated with cancer and inflammatory conditions [70,71,72,73]. Indeed, elevated levels of serum hyaluronan have been previously reported in patients with rheumatic mitral stenosis and pulmonary arterial thromboembolism [74]. In addition, there is also excessive abnormal angiogenesis, which is observed in the progression of cancer, diabetic retinopathy and in atherosclerosis [75,76,77,78]. Atherosclerosis is one of the major cardiovascular disease conditions involving inflammation, where hyaluronan is thought to play a role [79]. The differentially expressed hyaluronan in atheromatic aortas was found to play a role in platelet-derived growth factor (PDGF)-induced vascular smooth muscle cell proliferation and migration, which is important during atherogenesis [80]. The CD44-hyaluronan axis is believed to regulate the inflammatory process involved in atherosclerosis, making it an important potential drug target in order to bring about disease remission [81]. Indeed, overexpression of hyaluronan in the tunica media of an aorta has been demonstrated to promote the development of atherosclerosis [82]. Moreover, increased expression of some of the biosynthetic enzymes of hyaluronan, such as HAS-3, was found to increase inflammation in an atherosclerotic plaque and promote atheroprogression [83]. This suggests that hyaluronan biosynthetic enzymes such as HAS-3 could be important therapeutic targets in the fight against atherosclerosis. The inflammatory process in atherosclerosis also promotes endothelial and platelet dysfunction that exacerbate the pathogenesis of this condition [84]. More evidence showing the impairment of the endothelial glycocalyx in atherosclerosis and obesity was recently summarized by Sang Joon Ahn et al. [85].

3. Glycocalyx and Salt Interactions

Since the endothelial glycocalyx covers the luminal surface of the cells, where it forms a gel-like structure, it is believed to protect the endothelial cells from direct exposure to excessive NaCl salt (above 160 mEq/L) dissolved in plasma [86]. The normal sodium levels in plasma are kept within a narrow range of 135–145 mEq/L by a combination of ‘thirst’/water intake and hormonal (aldosterone-anti-diuretic/vasopressin) systems [87,88]. The GAG chains (including heparan sulfate, chondroitin sulfate, and hyaluronan) are major components of the glycocalyx and are highly negatively charged, making them attractive to the positively charged sodium ions flowing in circulation [89], Figure 2. Thus, the glycocalyx is able to play a positive role in the sodium buffering by transiently binding sodium on the luminal side of the blood vessels [90]. The glycocalyx, therefore, is a major player in buffering the intravascular sodium and stores a great amount sodium creating a hypertonic environment [91,92].
Increased sodium levels in plasma are associated with rapid degradation of the endothelial glycocalyx, which may cause endothelial dysfunction characteristic of most cardiovascular diseases [13,92,93]. Sodium overload in the blood vessels arises as a result of excessive dietary intake beyond the capacity of the kidney to excrete it, which affects the function of not only the blood vessels but also of other organs including the heart and kidneys [91,94,95].

3.1. Proposed Glycocalyx-Salt Interaction Mechanisms Contributing to Hypertension and Cardiovascular Disease

The mechanism underlying the rapid degradation of the endothelial glycocalyx is that excess sodium diminishes the buffering capacity of the glycocalyx leading to increased sodium reaching endothelial cells as well as reducing the repelling effect between the vascular and erythrocyte glycocalyces; this leads to corrosion of both the erythrocyte and endothelial glycocalyces, which result in endothelial activation and dysfunction [93,96,97]. Damage to the vascular glycocalyx also leads to extravasation of the excess sodium ions into the interstitial glycosaminoglycan networks where sodium disrupts the function of the glycosaminoglycans and also activates immune cells [91,98,99]. The augmented interaction between the erythrocytes and endothelial glycocalyces increases the thrombotic events [100] and the interaction between the endothelial cell and innate cells in the lumen via the adhesion molecules [101]. The entry of excess sodium through the epithelial sodium channel (ENaC) on the endothelial cells and innate immune cells (such as dendritic cells [102,103,104,105] and macrophages) activates nicotinamide adenine dinucleotide phosphate (NADPH) oxidase resulting in the generation of super oxides, peroxynitrite, and other reactive oxygen species (ROS) [106]. Normally, nitric oxide (NO) is produced to dilate blood vessels by converting L-arginine to L-citrulline and NO catalyzed by the endothelial nitric oxide synthase. The ROS react directly with the NO production pathway and inhibit the production of NO [106,107,108]. The increased oxidative stress from ROS production by the NADPH oxidase enzyme also activates the nuclear factor kappa-light-chain-enhancer of activated B (NF-κB) mediated by the NLR Family Pyrin Domain Containing 3 (NLRP3) inflammasome, leading to the production of inflammatory cytokines such as tumor necrosis factor-alpha (TNF-α), IL-1β, and IL-6 [105,109,110]. The increased production of inflammatory cytokines coupled with the reduced production of NO resulting from ROS reactions leads to the stiffening of blood vessels and endothelial activation/dysfunction that contributes to hypertension and CVDs [109]. The produced inflammatory cytokines disrupt the gap junctions between the endothelial cells and connexin hemichannels leading to increased permeability and endothelial dysfunction [111]. Further, the activated endothelial cells begin to increase their expression of adhesion molecules leading to increased rolling and adhesion of monocytes and other cells to the endothelial cells in the vasculature leading to thrombotic and increased endothelial dysfunction [112]. This is further augmented by the increased activation of NADPH oxidase that is mediated by increased salt entry via the ENaC leading to the formation of isolevuglandin (IsoLG)-protein adducts, adaptive immune activation, and secretion of inflammatory cytokines such as IL-17A, TNF-α, and interferon-gamma (IFN-γ) that contributes to the development of hypertension and exacerbates the already existing CVDs [110,113,114,115] (see Figure 3). IsoLGs are formed when ROS peroxidize arachidonic acid, a structural component of cell membranes [114]. The resulting IsoLGs bind non-covalently to lysine residues of intracellular proteins, altering their structure and function [114]. The IsoLG-protein adduct is presented to the T cells on the major histocompatibility complex (MHC) molecules of the dendritic cells as neoantigens to activate the T cells [114]. The inflammatory cytokines secreted by the T cells injure the vascular lining and cause endothelial dysfunction, and the resulting healing by fibrosis reduces the vascular lumen, stiffens the blood vessels, and accelerates atherosclerosis leading to hypertension [114]. Moreover, the delta sub-unit of the ENaC has been reported to be expressed in human arteries and its elevated levels of expression are potentially associated with hypertension [116].
The endothelial cells with excessively degraded glycocalyx from excess sodium overload are unable to produce sufficient nitric oxide due to the reduced activation of endothelial nitric oxide synthase [117,118]. Recent evidence has shown that the endothelial glycocalyx plays a role in initiating the signal transduction pathways that contribute to NO production. Specifically, Bartosch et al. demonstrated that the proteoglycan glypican-1, not syndecan-1, plays a dominant role in propagating extracellular forces into the endothelial cells to activate signal transduction for the production of NO [119]. Moreover, this plasma sodium overload contributes not only to increased loss of the endothelial glycocalyx but also to vascular inflammation, which is characteristic of various cardiovascular diseases as explained above [120]. For instance, the hypertension experienced in pre-eclampsia has been associated with increased degradation of the endothelial glycocalyx with subsequent endothelial dysfunction and vascular injury [121,122].
The endothelial glycocalyx is therefore highly sensitive to high salt intake. Not only does high salt disrupt the repulsive forces between erythrocytes and endothelial glycocalyces by saturating their buffering capacity for sodium ions, but, as mentioned earlier, high sodium also facilitates and increases the adhesion forces occurring between monocytes and endothelial surfaces leading to monocyte and endothelial cell activation that results in endothelial dysfunction and inflammation [120]. Activation of the endothelial cells increases their expression of P- and E-selectins and additional adhesion molecules that facilitate rolling and adhesion of the leukocyte to the endothelium [123,124].
It is also important to mention that the damage to the glycocalyx induced by high salt intake occurs more with chronic salt overload as opposed to acute salt loading and deterioration of the glycocalyx advances with ageing independent of the salt intake [120,125].

3.2. Possible Strategies for Reducing the Damaging Effects of NaCl-Salt Overload on Vascular Endothelium

Studies that explore the mechanisms associated with reducing the damaging effects of salt overload on vascular endothelium are scarce, but a few studies have reported promising results and are highlighted below.
High dietary salt intake remains a big challenge as many people in various populations around the world are still unable to stop consuming high amounts of salt due to the diverse sources of dietary salt available in common foods [126,127,128]. It is therefore understandable that efforts are being made to find alternative ways of overcoming the deleterious effects of high salt overload on human health, other than simply advising people to reduce salt intake. For example, a recent clinical trial conducted among black women aged between 20 and 60 years in the USA provided evidence showing that ‘hot yoga’ can reduce the harmful effects of salt overload on endothelial function [129]. The exact mechanism is unknown. Similarly, regular aerobic exercise has also been reported to reduce endothelin-1-mediated vasoconstriction and endothelial dysfunction in postmenopausal women, as well as in obese or overweight adults [130,131]. Replacement or substitution of NaCl salt with other forms of salt with similar ‘saltness’ taste, such as potassium chloride (KCl) and monosodium glutamate (MSG), have also been piloted by the Department of Food Science at Cornell University (New York) and been found to have relatively high acceptability among the study subjects, although with a caveat of not disclosing the specific names of the salt substitute [132]. Consumer acceptability of the salt substitute is indeed considered to be necessary in the reduction in NaCl dietary salt intake [133]. It has also been suggested that the increased intake of potassium, rather than sodium, can help to alleviate the deleterious effects of excessive sodium during the treatment of hypertension [134,135]. Other substitutes that have been used in the reduction in dietary NaCl intake include yeast extract, taste peptides, and odor compounds [136]. Since the ENaC subunits are expressed in several vascular beds including mesenteric, cerebral, and renal [137,138,139], where the increased ENaC expression is also associated with salt-sensitive hypertension by causing endothelial dysfunction or vascular smooth muscle activation [103,140], it is a therapeutic target to be blocked using drugs such as benzamil or amiloride in conjunction with other potential anti-hypertensive agents [141]. Because high extracellular salt concentration increases ENaC expression [90,142], it is logical that blocking it with amiloride helps to restore endothelial function through increased phosphorylation of endothelial nitric oxide synthase (eNOS) [138]. As the endothelial glycocalyx plays a central role in buffering against excessive salt, with its degradation preceding vascular endothelial dysfunction, preventing this damage and restoring the glycocalyx has been proposed as an important strategy in managing CVD disorders [46]. Some of the therapies that can be used to protect against degradation and help in regenerating the damaged endothelial glycocalyx include berberin, doxycycline, and sphingosine-1-phosphate (S1P) [143,144,145]. These drugs act to prevent the shedding of the glycocalyx by inhibiting the increased secretion of heparinase and MMP enzymes [145,146]. For instance, S1P binds on its receptor on the endothelial cells and increases the synthesis of syndecan-1 and heparan sulfate through phosphatidyl inositol-3 kinase (PI3K) signaling [147].

4. Current Diagnostic Tools to Determine Glycocalyx and Endothelial Health Status

Since the glycocalyx consists of both protein and carbohydrate components, its breakdown products include monomers or shorter/smaller molecules of each of the two large macromolecules, as well as shed syndecans and glypicans [27,148]. These include small peptides or amino acids and the breakdown products of proteins: glypicans, syndecan-1, shorter disaccharides, or individual GlcA and GlcNAc—these are sugar residues, which are the major breakdown products of the polysaccharide component of the glycocalyx [148]. The much longer HA polysaccharides are usually reduced to shorter sugar chains that are released into various body fluids including saliva, cerebrospinal fluid (CSF), urine, and blood plasma and can easily be isolated from such body fluids [149,150]. The smaller peptides or amino acids can be re-used in the synthesis of new proteins by body cells [151]. Most of the breakdown products of the polysaccharide component of the glycocalyx end up being deposited in body tissues and body fluids from which they can be measured or determined to assess the level of glycocalyx degradation as biomarkers of health or disease [148,150]. For example, elevated plasma levels of the glycocalyx components seen in sepsis-associated encephalopathy can be used as early biomarkers of cognitive impairment among sepsis patients [152]. However, Hahn et al. have summarized evidence, which seems to suggest that since glycocalyx shedding is widespread in both acute and chronic inflammatory conditions, there is a lack of adequate sensitivity or specificity for any particular disease by measuring the plasma levels of the glycocalyx components [153]. A combination of biochemical and immunological techniques such as ELISA are commonly used in the determination of the concentration of glycocalyx breakdown products [154,155]. The thickness of the glycocalyx may also be determined indirectly by using a side stream dark field imaging technique [156]. Other techniques such as atomic force microscopy and liquid chromatography with selected reaction monitoring in tandem with mass spectrometry (LC-SRM/MS) have been used to successfully determine the glycocalyx thickness disruption in both in vivo and in vitro assays [157]. Dimethylmethylene blue (GAG-DMMB) and liquid chromatography-tandem mass spectrometry (GAG-MS) assay have also been used to determine the concentration of the glycocalyx components in urine [158].

Erythrocyte Salt Sensitivity Test

The sodium buffering capacity of both the endothelial and erythrocyte glycocalyx can be measured using a simple “salt blood test” as a way of assessing the functional capacity of the glycocalyx [159,160]. We have previously shown that a high intake of salt is associated with a damaged glycocalyx and could be potentially used to predict future CVD [161]. The salt blood test or erythrocyte salt sensitivity test has been used in several studies as a surrogate for endothelial function and should be used more often in cost limited settings [161,162]. A recent pilot study by McNally et al. demonstrated that the erythrocyte salt sensitivity test can be used as a marker for cellular salt sensitivity in hypertension [163].

5. Future Directions

Future research efforts should be directed toward interventions that can ameliorate the damaging effects of excessive dietary salt, since many people seem to be unable to avoid it. Additional research is needed to specifically evaluate and validate the diagnostic and prognostic potential of these glycocalyx components that are found in body fluids such as the plasma and urine of patients with cardiovascular disease.

6. Conclusions

Endothelial glycocalyx is important in protecting endothelial cells from direct exposure to excessive amounts of salt and helps in maintaining normal endothelial function, which is necessary in the prevention of cardiovascular diseases. The degradation of the glycocalyx is increased in certain cardiovascular diseases due to enhanced sheer force of the rapid blood flow in hypertension, inflammatory changes, and oxidative stress in atherosclerosis and, alternatively, because of the increased expression of degradative enzymes. Excessive salt due to salt overload may also lead to rapid degradation of the endothelial glycocalyx, resulting in an endothelial dysfunction and consequent cardiovascular disease. This makes dietary modification, particularly reducing sodium-salt intake or using substitutes for dietary salt, a key strategy for preserving glycocalyx integrity. The increased presence of breakdown components of the glycocalyx, such as syndecans and hyaluronan in the plasma or urine of cardiovascular disease patients can be measured and utilized as a potential prognostic biomarker for specific cardiovascular disorders.

Author Contributions

L.F.S. wrote the original draft and edited the manuscript; A.M.S., A.N. (Agnes Namaganda), H.M., J.N.K., R.K., A.N. (Annettee Nakimuli) and M.N. wrote different sections of the manuscript and contributed to all subsequent revisions; K.P.P.: wrote different sections, responded to some reviewer comments and contributed to the overall scientific content; S.K.M. wrote different sections of the manuscript, made revisions and created all figures. A.K. supervised the overall writing, edited, and contributed to the scientific content and submitted the final manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Fogarty International Center of the National Institutes of Health grants; Reference numbers: R03HL155041, R01HL147818, R01HL144941 (AK), and 2D43TW009744 (SKM). The content is solely the responsibility of the authors and does not represent the official views of the National Institutes of Health.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

All data are contained within the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Mandrycky, C.J.; Howard, C.C.; Rayner, S.G.; Shin, Y.J.; Zheng, Y. Organ-on-a-chip systems for vascular biology. J. Mol. Cell. Cardiol. 2021, 159, 1–13. [Google Scholar] [CrossRef] [PubMed]
  2. Daly, C.J. Examining Vascular Structure and Function Using Confocal Microscopy and 3D Imaging Techniques. Adv. Exp. Med. Biol. 2019, 1120, 97–106. [Google Scholar]
  3. Eelen, G.; Treps, L.; Li, X.; Carmeliet, P. Basic and Therapeutic Aspects of Angiogenesis Updated. Circ. Res. 2020, 127, 310–329. [Google Scholar] [CrossRef] [PubMed]
  4. Clough, G. Relationship between microvascular permeability and ultrastructure. Prog. Biophys. Mol. Biol. 1991, 55, 47–69. [Google Scholar] [CrossRef] [PubMed]
  5. Stan, R.V.; Tse, D.; Deharvengt, S.J.; Smits, N.C.; Xu, Y.; Luciano, M.R.; McGarry, C.L.; Buitendijk, M.; Nemani, K.V.; Elgueta, R.; et al. The diaphragms of fenestrated endothelia: Gatekeepers of vascular permeability and blood composition. Dev. Cell 2012, 23, 1203–1218. [Google Scholar] [CrossRef] [Green Version]
  6. Dull, R.O.; Hahn, R.G. The glycocalyx as a permeability barrier: Basic science and clinical evidence. Crit. Care 2022, 26, 273. [Google Scholar] [CrossRef]
  7. Jin, J.; Fang, F.; Gao, W.; Chen, H.; Wen, J.; Wen, X.; Chen, J. The Structure and Function of the Glycocalyx and Its Connection with Blood-Brain Barrier. Front. Cell. Neurosci. 2021, 15, 739699. [Google Scholar] [CrossRef]
  8. Haymet, A.B.; Bartnikowski, N.; Wood, E.S.; Vallely, M.P.; McBride, A.; Yacoub, S.; Biering, S.B.; Harris, E.; Suen, J.Y.; Fraser, J.F. Studying the Endothelial Glycocalyx in vitro: What Is Missing? Front. Cardiovasc. Med. 2021, 8, 647086. [Google Scholar] [CrossRef]
  9. Wang, G.; Tiemeier, G.L.; van den Berg, B.M.; Rabelink, T.J. Endothelial Glycocalyx Hyaluronan: Regulation and Role in Prevention of Diabetic Complications. Am. J. Pathol. 2020, 190, 781–790. [Google Scholar] [CrossRef]
  10. Sarrazin, S.; Lamanna, W.C.; Esko, J.D. Heparan sulfate proteoglycans. Cold Spring Harb. Perspect. Biol. 2011, 3, a004952. [Google Scholar] [CrossRef] [Green Version]
  11. Melrose, J. Perlecan, a modular instructive proteoglycan with diverse functional properties. Int. J. Biochem. Cell Biol. 2020, 128, 105849. [Google Scholar] [CrossRef] [PubMed]
  12. Reitsma, S.; Slaaf, D.W.; Vink, H.; van Zandvoort, M.A.; oude Egbrink, M.G. The endothelial glycocalyx: Composition, functions, and visualization. Pflug. Arch. 2007, 454, 345–359. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Oberleithner, H. Vascular endothelium: A vulnerable transit zone for mercilesssodium. Nephrol. Dial. Transplant. 2014, 29, 240–246. [Google Scholar] [CrossRef] [Green Version]
  14. Bkaily, G.; Jacques, D. Morphological and Functional Remodeling of Vascular Endothelium in Cardiovascular Diseases. Int. J. Mol. Sci. 2023, 24, 1998. [Google Scholar] [CrossRef]
  15. Knežević, D.; Ćurko-Cofek, B.; Batinac, T.; Laškarin, G.; Rakić, M.; Šoštarič, M.; Zdravković, M.; Šustić, A.; Sotošek, V.; Batičić, L. Endothelial Dysfunction in Patients Undergoing Cardiac Surgery: A Narrative Review and Clinical Implications. J. Cardiovasc. Dev. Dis. 2023, 10, 213. [Google Scholar] [CrossRef]
  16. Foote, C.A.; Soares, R.N.; Ramirez-Perez, F.I.; Ghiarone, T.; Aroor, A.; Manrique-Acevedo, C.; Padilla, J.; Martinez-Lemus, L. Endothelial Glycocalyx. Compr. Physiol. 2022, 12, 3781–3811. [Google Scholar] [PubMed]
  17. Mortazavi, C.M.; Hoyt, J.M.; Patel, A.; Chignalia, A.Z. The glycocalyx and calcium dynamics in endothelial cells. Curr. Top. Membr. 2023, 91, 21–41. [Google Scholar]
  18. Askari, H.; Sadeghinejad, M.; Fancher, I.S. Mechanotransduction and the endothelial glycocalyx: Interactions with membrane and cytoskeletal proteins to transduce force. Curr. Top. Membr. 2023, 91, 43–60. [Google Scholar]
  19. Pot, C.; Chen, A.Y.; Ha, J.N.; Schmid-Schönbein, G.W. Proteolytic Cleavage of the Red Blood Cell Glycocalyx in a Genetic Form of Hypertension. Cell. Mol. Bioeng. 2011, 4, 678–692. [Google Scholar] [CrossRef]
  20. Salmon, A.H.; Ferguson, J.K.; Burford, J.L.; Gevorgyan, H.; Nakano, D.; Harper, S.J.; Harper, D.O. Bates, and J. Peti-Peterdi, Loss of the endothelial glycocalyx links albuminuria and vascular dysfunction. J. Am. Soc. Nephrol. 2012, 23, 1339–1350. [Google Scholar] [CrossRef] [Green Version]
  21. Tarbell, J.M.; Cancel, L.M. The glycocalyx and its significance in human medicine. J. Intern. Med. 2016, 280, 97–113. [Google Scholar] [CrossRef] [Green Version]
  22. Esko, J.D.; Rostand, K.S.; Weinke, J.L. Tumor formation dependent on proteoglycan biosynthesis. Science 1988, 241, 1092–1096. [Google Scholar] [CrossRef]
  23. Llaneza, A.; Vizoso, F.; Rodriguez, J.C.; Raigoso, P.; Garcia-Muniz, J.L.; Allende, M.T.; Garcia-Moran, M. Hyaluronic acid as prognostic marker in resectable colorectal cancer. Br. J. Surg. 2000, 87, 1690–1696. [Google Scholar] [CrossRef]
  24. Adamia, S.; Maxwell, C.A.; Pilarski, L.M. Hyaluronan and hyaluronan synthases: Potential therapeutic targets in cancer. Curr. Drug Targets Cardiovasc. Haematol. Disord. 2005, 5, 3–14. [Google Scholar] [CrossRef] [Green Version]
  25. Dogné, S.; Flamion, B. Endothelial Glycocalyx Impairment in Disease: Focus on Hyaluronan Shedding. Am. J. Pathol. 2020, 190, 768–780. [Google Scholar] [CrossRef]
  26. Csóka, A.B.; Scherer, S.W.; Stern, R. Expression analysis of six paralogous human hyaluronidase genes clustered on chromosomes 3p21 and 7q31. Genomics 1999, 60, 356–361. [Google Scholar] [CrossRef]
  27. Becker, B.F.; Jacob, M.; Leipert, S.; Salmon, A.H.; Chappell, D. Degradation of the endothelial glycocalyx in clinical settings: Searching for the sheddases. Br. J. Clin. Pharmacol. 2015, 80, 389–402. [Google Scholar] [CrossRef]
  28. Johansson, P.I.; Stensballe, J.; Rasmussen, L.S.; Ostrowski, S.R. A high admission syndecan-1 level, a marker of endothelial glycocalyx degradation, is associated with inflammation, protein C depletion, fibrinolysis, and increased mortality in trauma patients. Ann. Surg. 2011, 254, 194–200. [Google Scholar] [CrossRef] [PubMed]
  29. Robich, M.; Ryzhov, S.; Kacer, D.; Palmeri, M.; Peterson, S.M.; Quinn, R.D.; Carter, D.; Sheppard, F.; Hayes, T.; Sawyer, D.B.; et al. Prolonged Cardiopulmonary Bypass is Associated with Endothelial Glycocalyx Degradation. J. Surg. Res. 2020, 251, 287–295. [Google Scholar] [CrossRef] [PubMed]
  30. Pretorius, D.; Richter, R.P.; Anand, T.; Cardenas, J.C.; Richter, J.R. Alterations in heparan sulfate proteoglycan synthesis and sulfation and the impact on vascular endothelial function. Matrix Biol. Plus 2022, 16, 100121. [Google Scholar] [CrossRef] [PubMed]
  31. Gondelaud, F.; Ricard-Blum, S. Structures and interactions of syndecans. FEBS J. 2019, 286, 2994–3007. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Couchman, J.R.; Gopal, S.; Lim, H.C.; Nørgaard, S.; Multhaupt, H.A. Fell-Muir Lecture: Syndecans: From peripheral coreceptors to mainstream regulators of cell behaviour. Int. J. Exp. Pathol. 2015, 96, 1–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Deepa, S.S.; Yamada, S.; Zako, M.; Goldberger, O.; Sugahara, K. Chondroitin sulfate chains on syndecan-1 and syndecan-4 from normal murine mammary gland epithelial cells are structurally and functionally distinct and cooperate with heparan sulfate chains to bind growth factors. A novel function to control binding of midkine, pleiotrophin, and basic fibroblast growth factor. J. Biol. Chem. 2004, 279, 37368–37376. [Google Scholar]
  34. Iozzo, R.V.; Schaefer, L. Proteoglycan form and function: A comprehensive nomenclature of proteoglycans. Matrix Biol. 2015, 42, 11–55. [Google Scholar] [CrossRef] [PubMed]
  35. Bertrand, J.; Bollmann, M. Soluble syndecans: Biomarkers for diseases and therapeutic options. Br. J. Pharmacol. 2019, 176, 67–81. [Google Scholar] [CrossRef] [PubMed]
  36. Pruessmeyer, J.; Martin, C.; Hess, F.M.; Schwarz, N.; Schmidt, S.; Kogel, T.; Hoettecke, N.; Schmidt, B.; Sechi, A.; Uhlig, S.; et al. A disintegrin and metalloproteinase 17 (ADAM17) mediates inflammation-induced shedding of syndecan-1 and -4 by lung epithelial cells. J. Biol. Chem. 2010, 285, 555–564. [Google Scholar] [CrossRef] [Green Version]
  37. Itoh, Y. Modulation of Microenvironment Signals by Proteolytic Shedding of Cell Surface Extracellular Matrix Receptors. Front. Cell Dev. Biol. 2021, 9, 736735. [Google Scholar] [CrossRef]
  38. Gopal, S.; Arokiasamy, S.; Pataki, C.; Whiteford, J.R.; Couchman, J.R. Syndecan receptors: Pericellular regulators in development and inflammatory disease. Open Biol. 2021, 11, 200377. [Google Scholar] [CrossRef]
  39. Ha, E.; Kim, M.J.; Choi, B.K.; Rho, J.J.; Oh, D.J.; Rho, T.H.; Kim, K.H.; Lee, H.J.; Shin, D.H.; Yim, S.V.; et al. Positive association of obesity with single nucleotide polymorphisms of syndecan 3 in the Korean population. J. Clin. Endocrinol. Metab. 2006, 91, 5095–5099. [Google Scholar] [CrossRef] [Green Version]
  40. Chang, B.C.; Hwang, L.C.; Huang, W.H. Positive Association of Metabolic Syndrome with a Single Nucleotide Polymorphism of Syndecan-3 (rs2282440) in the Taiwanese Population. Int. J. Endocrinol. 2018, 2018, 9282598. [Google Scholar] [CrossRef] [Green Version]
  41. Huang, W.H.; Hwang, L.C.; Chan, H.L.; Lin, H.Y.; Lin, Y.H. Study of seven single-nucleotide polymorphisms identified in East Asians for association with obesity in a Taiwanese population. BMJ Open 2016, 6, e011713. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Kunnas, T.; Nikkari, S.T. Contribution of syndecan-4 genetic variants to hypertension, the TAMRISK study. BMC Res. Notes 2014, 7, 815. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Russo, T.A.; Banuth, A.M.M.; Nader, H.B.; Dreyfuss, J.L. Altered shear stress on endothelial cells leads to remodeling of extracellular matrix and induction of angiogenesis. PLoS ONE 2020, 15, e0241040. [Google Scholar] [CrossRef] [PubMed]
  44. Lunde, I.G.; Herum, K.M.; Carlson, C.C.; Christensen, G. Syndecans in heart fibrosis. Cell Tissue Res. 2016, 365, 539–552. [Google Scholar] [CrossRef] [PubMed]
  45. Lunde, I.G.; Aronsen, J.M.; Melleby, A.O.; Strand, M.E.; Skogestad, J.; Bendiksen, B.A.; Ahmed, M.S.; Sjaastad, I.; Attramadal, H.; Carlson, C.R.; et al. Cardiomyocyte-specific overexpression of syndecan-4 in mice results in activation of calcineurin-NFAT signalling and exacerbated cardiac hypertrophy. Mol. Biol. Rep. 2022, 49, 11795–11809. [Google Scholar] [CrossRef]
  46. Milusev, A.; Rieben, R.; Sorvillo, N. The Endothelial Glycocalyx: A Possible Therapeutic Target in Cardiovascular Disorders. Front. Cardiovasc. Med. 2022, 9, 897087. [Google Scholar] [CrossRef]
  47. Abassi, Z.; Armaly, Z.; Heyman, S.N. Glycocalyx Degradation in Ischemia-Reperfusion Injury. Am. J. Pathol. 2020, 190, 752–767. [Google Scholar] [CrossRef]
  48. Kim, Y.H.; Nijst, P.; Kiefer, K.; Tang, W.H. Endothelial Glycocalyx as Biomarker for Cardiovascular Diseases: Mechanistic and Clinical Implications. Curr. Heart Fail Rep. 2017, 14, 117–126. [Google Scholar] [CrossRef] [Green Version]
  49. Ajaero, C.N.; Procter, N.E.K.; Chirkov, Y.Y.; Heresztyn, T.; Arstall, M.A.; McGavigan, A.D.; Frenneaux, M.P.; Horowitz, J.D. Endothelial dysfunction and glycocalyx shedding in heart failure: Insights from patients receiving cardiac resynchronisation therapy. Heart Vessel. 2020, 35, 197–206. [Google Scholar] [CrossRef]
  50. Filmus, J.; Capurro, M.; Rast, J. Glypicans. Genome Biol. 2008, 9, 224. [Google Scholar] [CrossRef] [Green Version]
  51. Filmus, J. The function of glypicans in the mammalian embryo. Am. J. Physiol. Cell Physiol. 2022, 322, C694–C698. [Google Scholar] [CrossRef] [PubMed]
  52. Filmus, J.; Capurro, M. The role of glypicans in Hedgehog signaling. Matrix Biol. 2014, 35, 248–252. [Google Scholar] [CrossRef] [PubMed]
  53. Su, G.; Meyer, K.; Nandini, C.D.; Qiao, D.; Salamat, S.; Friedl, A. Glypican-1 is frequently overexpressed in human gliomas and enhances FGF-2 signaling in glioma cells. Am. J. Pathol. 2006, 168, 2014–2026. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Traister, A.; Shi, W.; Filmus, J. Mammalian Notum induces the release of glypicans and other GPI-anchored proteins from the cell surface. Biochem. J. 2008, 410, 503–511. [Google Scholar] [CrossRef] [Green Version]
  55. Zeng, Y. Endothelial glycocalyx as a critical signalling platform integrating the extracellular haemodynamic forces and chemical signalling. J. Cell. Mol. Med. 2017, 21, 1457–1462. [Google Scholar] [CrossRef] [Green Version]
  56. Bartosch, A.M.W.; Mathews, R.; Mahmoud, M.M.; Cancel, L.M.; Haq, Z.S.; Tarbell, J.M. Heparan sulfate proteoglycan glypican-1 and PECAM-1 cooperate in shear-induced endothelial nitric oxide production. Sci. Rep. 2021, 11, 11386. [Google Scholar] [CrossRef]
  57. Kobayashi, T.; Chanmee, T.; Itano, N. Hyaluronan: Metabolism and Function. Biomolecules 2020, 10, 1525. [Google Scholar] [CrossRef]
  58. Itano, N.; Sawai, T.; Yoshida, M.; Lenas, P.; Yamada, Y.; Imagawa, M.; Shinomura, T.; Hamaguchi, M.; Yoshida, Y.; Ohnuki, Y.; et al. Three isoforms of mammalian hyaluronan synthases have distinct enzymatic properties. J. Biol. Chem. 1999, 274, 25085–25092. [Google Scholar] [CrossRef] [Green Version]
  59. Rilla, K.; Oikari, S.; Jokela, T.A.; Hyttinen, J.M.; Kärnä, R.; Tammi, R.H.; Tammi, M.I. Hyaluronan synthase 1 (HAS1) requires higher cellular UDP-GlcNAc concentration than HAS2 and HAS3. J. Biol. Chem. 2013, 288, 5973–5983. [Google Scholar] [CrossRef] [Green Version]
  60. Harada, H.; Takahashi, M. CD44-dependent intracellular and extracellular catabolism of hyaluronic acid by hyaluronidase-1 and -2. J. Biol. Chem. 2007, 282, 5597–5607. [Google Scholar] [CrossRef] [Green Version]
  61. Kaul, A.; Short, W.D.; Wang, X.; Keswani, S.G. Hyaluronidases in Human Diseases. Int. J. Mol. Sci. 2021, 22, 3204. [Google Scholar] [CrossRef] [PubMed]
  62. Patterson, E.K.; Cepinskas, G.; Fraser, D.D. Endothelial Glycocalyx Degradation in Critical Illness and Injury. Front. Med. 2022, 9, 898592. [Google Scholar] [CrossRef] [PubMed]
  63. Anderegg, U.; Eichenberg, T.; Parthaune, T.; Haiduk, C.; Saalbach, A.; Milkova, L.; Ludwig, A.; Grosche, J.; Averbeck, M.; Gebhardt, C.; et al. ADAM10 is the constitutive functional sheddase of CD44 in human melanoma cells. J. Investig. Dermatol. 2009, 129, 1471–1482. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Okamoto, I.; Kawano, Y.; Tsuiki, H.; Sasaki, J.I.; Nakao, M.; Matsumoto, M.; Suga, M.; Ando, M.; Nakajima, M.; Saya, H. CD44 cleavage induced by a membrane-associated metalloprotease plays a critical role in tumor cell migration. Oncogene 1999, 18, 1435–1446. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Pardue, E.L.; Ibrahim, S.; Ramamurthi, A. Role of hyaluronan in angiogenesis and its utility to angiogenic tissue engineering. Organogenesis 2008, 4, 203–214. [Google Scholar] [CrossRef] [Green Version]
  66. Ding, H.Y.; Xie, Y.N.; Dong, Q.; Kimata, K.; Nishida, Y.; Ishiguro, N.; Zhuo, L.S. Roles of hyaluronan in cardiovascular and nervous system disorders. J. Zhejiang Univ. Sci. B 2019, 20, 428–436. [Google Scholar] [CrossRef]
  67. West, D.C.; Kumar, S. Hyaluronan and angiogenesis. Ciba Found. Symp. 1989, 143, 187–201, discussion 81–85. [Google Scholar]
  68. Slevin, M.; Krupinski, J.; Gaffney, J.; Matou, S.; West, D.; Delisser, H.; Savani, R.C.; Kumar, S. Hyaluronan-mediated angiogenesis in vascular disease: Uncovering RHAMM and CD44 receptor signaling pathways. Matrix Biol. 2007, 26, 58–68. [Google Scholar] [CrossRef]
  69. Berdiaki, A.; Neagu, M.; Spyridaki, I.; Kuskov, A.; Perez, S.; Nikitovic, D. Hyaluronan and Reactive Oxygen Species Signaling-Novel Cues from the Matrix? Antioxidants 2023, 12, 824. [Google Scholar] [CrossRef]
  70. Day, A.J.; de la Motte, C.A. Hyaluronan cross-linking: A protective mechanism in inflammation? Trends Immunol. 2005, 26, 637–643. [Google Scholar] [CrossRef]
  71. Band, P.A.; Heeter, J.; Wisniewski, H.G.; Liublinska, V.; Pattanayak, C.W.; Karia, R.J.; Stabler, T.; Balazs, E.A.; Kraus, V.B. Hyaluronan molecular weight distribution is associated with the risk of knee osteoarthritis progression. Osteoarthr. Cartil. 2015, 23, 70–76. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Heldin, P.; Kolliopoulos, C.; Lin, C.Y.; Heldin, C.H. Involvement of hyaluronan and CD44 in cancer and viral infections. Cell. Signal. 2020, 65, 109427. [Google Scholar] [CrossRef] [PubMed]
  73. Karousou, E.; Misra, S.; Ghatak, S.; Dobra, K.; Götte, M.; Vigetti, D.; Passi, A.; Karamanos, N.K.; Skandalis, S.S. Roles and targeting of the HAS/hyaluronan/CD44 molecular system in cancer. Matrix Biol. 2017, 59, 3–22. [Google Scholar] [CrossRef] [PubMed]
  74. Kalay, N.; Elcik, D.; Savaş, G.; Altın, P.; Şakalar, Ç.; Kaya, Ö.; Aytekin, M. Elevated hyaluronan levels in patients with rheumatic mitral stenosis and pulmonary arterial thromboembolism. Heart Lung Circ. 2014, 23, 649–654. [Google Scholar] [CrossRef] [PubMed]
  75. Unterleuthner, D.; Neuhold, P.; Schwarz, K.; Janker, L.; Neuditschko, B.; Nivarthi, H.; Crncec, I.; Kramer, N.; Unger, C.; Hengstschläger, M.; et al. Cancer-associated fibroblast-derived WNT2 increases tumor angiogenesis in colon cancer. Angiogenesis 2020, 23, 159–177. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Viallard, C.; Larrivée, B. Tumor angiogenesis and vascular normalization: Alternative therapeutic targets. Angiogenesis 2017, 20, 409–426. [Google Scholar] [CrossRef]
  77. Wang, W.; Lo, A.C.Y. Diabetic Retinopathy: Pathophysiology and Treatments. Int. J. Mol. Sci. 2018, 19, 1816. [Google Scholar] [CrossRef] [Green Version]
  78. Perrotta, P.; Emini Veseli, B.; Van der Veken, B.; Roth, L.; Martinet, W.; De Meyer, G.R.Y. Pharmacological strategies to inhibit intra-plaque angiogenesis in atherosclerosis. Vasc. Pharmacol. 2019, 112, 72–78. [Google Scholar] [CrossRef]
  79. Falk, E. Pathogenesis of atherosclerosis. J. Am. Coll. Cardiol. 2006, 47 (Suppl. 8), C7–C12. [Google Scholar] [CrossRef] [Green Version]
  80. Papakonstantinou, E.; Roth, M.; Block, L.H.; Mirtsou-Fidani, V.; Argiriadis, P.; Karakiulakis, G. The differential distribution of hyaluronic acid in the layers of human atheromatic aortas is associated with vascular smooth muscle cell proliferation and migration. Atherosclerosis 1998, 138, 79–89. [Google Scholar] [CrossRef]
  81. Krolikoski, M.; Monslow, J.; Puré, E. The CD44-HA axis and inflammation in atherosclerosis: A temporal perspective. Matrix Biol. 2019, 78–79, 201–218. [Google Scholar] [CrossRef]
  82. Chai, S.; Chai, Q.; Danielsen, C.C.; Hjorth, P.; Nyengaard, J.R.; Ledet, T.; Yamaguchi, Y.; Rasmussen, L.M.; Wogensen, L. Overexpression of hyaluronan in the tunica media promotes the development of atherosclerosis. Circ. Res. 2005, 96, 583–591. [Google Scholar] [CrossRef] [Green Version]
  83. Homann, S.; Grandoch, M.; Kiene, L.S.; Podsvyadek, Y.; Feldmann, K.; Rabausch, B.; Nagy, N.; Lehr, S.; Kretschmer, I.; Oberhuber, A.; et al. Hyaluronan synthase 3 promotes plaque inflammation and atheroprogression. Matrix Biol. 2018, 66, 67–80. [Google Scholar] [CrossRef] [PubMed]
  84. van der Poll, T.; Parker, R.I. Platelet Activation and Endothelial Cell Dysfunction. Crit. Care Clin. 2020, 36, 233–253. [Google Scholar] [CrossRef] [PubMed]
  85. Ahn, S.J.; Le Master, E.; Granados, S.T.; Levitan, I. Impairment of endothelial glycocalyx in atherosclerosis and obesity. Curr. Top. Membr. 2023, 91, 1–19. [Google Scholar] [PubMed]
  86. Weinbaum, S.; Cancel, L.M.; Fu, B.M.; Tarbell, J.M. The Glycocalyx and Its Role in Vascular Physiology and Vascular Related Diseases. Cardiovasc. Eng. Technol. 2021, 12, 37–71. [Google Scholar] [CrossRef] [PubMed]
  87. Ackerman, G. Serum Sodium. In Clinical Methods: The History, Physical, and Laboratory Examinations, 3rd ed.; Walker, H.K., Hall, W.D., Hurst, J.W., Eds.; Butterworths: Oxford, UK, 1990. [Google Scholar]
  88. Hyndman, K.A.; Mironova, E.V.; Giani, J.F.; Dugas, C.; Collins, J.; McDonough, A.A.; Stockand, J.D.; Pollock, J.S. Collecting Duct Nitric Oxide Synthase 1ß Activation Maintains Sodium Homeostasis During High Sodium Intake Through Suppression of Aldosterone and Renal Angiotensin II Pathways. J. Am. Heart Assoc. 2017, 6, e006896. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Oberleithner, H.; Wilhelmi, M. Vascular glycocalyx sodium store—Determinant of salt sensitivity? Blood Purif. 2015, 39, 7–10. [Google Scholar] [CrossRef]
  90. Korte, S.; Wiesinger, A.; Straeter, A.S.; Peters, W.; Oberleithner, H.; Kusche-Vihrog, K. Firewall function of the endothelial glycocalyx in the regulation of sodium homeostasis. Pflug. Arch. 2012, 463, 269–278. [Google Scholar] [CrossRef]
  91. Nijst, P.; Verbrugge, F.H.; Grieten, L.; Dupont, M.; Steels, P.; Tang, W.H.W.; Mullens, W. The pathophysiological role of interstitial sodium in heart failure. J. Am. Coll. Cardiol. 2015, 65, 378–388. [Google Scholar] [CrossRef] [Green Version]
  92. Titze, J.; Machnik, A. Sodium sensing in the interstitium and relationship to hypertension. Curr. Opin. Nephrol. Hypertens. 2010, 19, 385–392. [Google Scholar] [CrossRef]
  93. Kusche-Vihrog, K.; Oberleithner, H. An emerging concept of vascular salt sensitivity. F1000 Biol. Rep. 2012, 4, 20. [Google Scholar] [CrossRef] [PubMed]
  94. Robinson, A.T.; Edwards, D.G.; Farquhar, W.B. The Influence of Dietary Salt Beyond Blood Pressure. Curr. Hypertens. Rep. 2019, 21, 42. [Google Scholar] [CrossRef] [PubMed]
  95. Farquhar, W.B.; Edwards, D.G.; Jurkovitz, C.T.; Weintraub, W.S. Dietary sodium and health: More than just blood pressure. J. Am. Coll. Cardiol. 2015, 65, 1042–1050. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Oberleithner, H.; Peters, W.; Kusche-Vihrog, K.; Korte, S.; Schillers, H.; Kliche, K.; Oberleithner, K. Salt overload damages the glycocalyx sodium barrier of vascular endothelium. Pflug. Arch. 2011, 462, 519–528. [Google Scholar] [CrossRef] [Green Version]
  97. Sulyok, E.; Farkas, B.; Nagy, B.; Várnagy, Á.; Kovács, K.; Bódis, J. Tissue Sodium Accumulation: Pathophysiology and Clinical Implications. Antioxidants 2022, 11, 750. [Google Scholar] [CrossRef] [PubMed]
  98. Li, X.; Alu, A.; Wei, Y.; Wei, X.; Luo, M. The modulatory effect of high salt on immune cells and related diseases. Cell Prolif. 2022, 55, e13250. [Google Scholar] [CrossRef]
  99. Kirabo, A. A new paradigm of sodium regulation in inflammation and hypertension. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2017, 313, R706–R710. [Google Scholar] [CrossRef]
  100. Oberleithner, H.; Wälte, M.; Kusche-Vihrog, K. Sodium renders endothelial cells sticky for red blood cells. Front. Physiol. 2015, 6, 188. [Google Scholar] [CrossRef] [Green Version]
  101. Mitra, R.; O’Neil, G.L.; Harding, I.C.; Cheng, M.J.; Mensah, S.A.; Ebong, E.E. Glycocalyx in Atherosclerosis-Relevant Endothelium Function and as a Therapeutic Target. Curr. Atheroscler. Rep. 2017, 19, 63. [Google Scholar] [CrossRef] [Green Version]
  102. Mutchler, S.M.; Kleyman, T.R. New insights regarding epithelial Na+ channel regulation and its role in the kidney, immune system and vasculature. Curr. Opin. Nephrol. Hypertens. 2019, 28, 113–119. [Google Scholar] [CrossRef]
  103. Ertuglu, L.A.; Kirabo, A. Dendritic Cell Epithelial Sodium Channel in Inflammation, Salt-Sensitive Hypertension, and Kidney Damage. Kidney360 2022, 3, 1620–1629. [Google Scholar] [CrossRef] [PubMed]
  104. Barbaro, N.R.; Foss, J.D.; Kryshtal, D.O.; Tsyba, N.; Kumaresan, S.; Xiao, L.; Mernaugh, R.L.; Itani, H.A.; Loperena, R.; Chen, W.; et al. Dendritic Cell Amiloride-Sensitive Channels Mediate Sodium-Induced Inflammation and Hypertension. Cell Rep. 2017, 21, 1009–1020. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Pitzer, A.; Elijovich, F.; Laffer, C.L.; Ertuglu, L.A.; Sahinoz, M.; Saleem, M.; Krishnan, J.; Dola, T.; Aden, L.A.; Sheng, Q.; et al. DC ENaC-Dependent Inflammasome Activation Contributes to Salt-Sensitive Hypertension. Circ. Res. 2022, 131, 328–344. [Google Scholar] [CrossRef] [PubMed]
  106. Datla, S.R.; Griendling, K.K. Reactive oxygen species, NADPH oxidases, and hypertension. Hypertension 2010, 56, 325–330. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Patik, J.C.; Lennon, S.L.; Farquhar, W.B.; Edwards, D.G. Mechanisms of Dietary Sodium-Induced Impairments in Endothelial Function and Potential Countermeasures. Nutrients 2021, 13, 270. [Google Scholar] [CrossRef] [PubMed]
  108. Ruggeri Barbaro, N.; Van Beusecum, J.; Xiao, L.; do Carmo, L.; Pitzer, A.; Loperena, R.; Foss, J.D.; Elijovich, F.; Laffer, C.L.; Montaniel, K.R.; et al. Sodium activates human monocytes via the NADPH oxidase and isolevuglandin formation. Cardiovasc. Res. 2021, 117, 1358–1371. [Google Scholar] [CrossRef]
  109. Masenga, S.K.; Kabwe, L.S.; Chakulya, M.; Kirabo, A. Mechanisms of Oxidative Stress in Metabolic Syndrome. Int. J. Mol. Sci. 2023, 24, 7898. [Google Scholar] [CrossRef]
  110. Ertuglu, L.A.; Mutchler, A.P.; Yu, J.; Kirabo, A. Inflammation and oxidative stress in salt sensitive hypertension; The role of the NLRP3 inflammasome. Front. Physiol. 2022, 13, 1096296. [Google Scholar] [CrossRef]
  111. Okamoto, T.; Suzuki, K. The Role of Gap Junction-Mediated Endothelial Cell-Cell Interaction in the Crosstalk between Inflammation and Blood Coagulation. Int. J. Mol. Sci. 2017, 18, 2254. [Google Scholar] [CrossRef] [Green Version]
  112. Chadjichristos, C.E.; Scheckenbach, K.E.L.; Van Veen, T.; Richani Sarieddine, M.Z.; De Wit, C.; Yang, Z.; Roth, I.; Bacchetta, M.; Viswambharan, H.; Foglia, B.; et al. Endothelial-specific deletion of connexin40 promotes atherosclerosis by increasing CD73-dependent leukocyte adhesion. Circulation 2010, 121, 123–131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Rucker, A.J.; Rudemiller, N.P.; Crowley, S.D. Salt, Hypertension, and Immunity. Annu. Rev. Physiol. 2018, 80, 283–307. [Google Scholar] [CrossRef] [PubMed]
  114. Xiao, L.; Patrick, D.M.; Aden, L.A.; Kirabo, A. Mechanisms of isolevuglandin-protein adduct formation in inflammation and hypertension. Prostaglandins Other Lipid Mediat. 2018, 139, 48–53. [Google Scholar] [CrossRef] [PubMed]
  115. Dixon, K.B.; Davies, S.S.; Kirabo, A. Dendritic cells and isolevuglandins in immunity, inflammation, and hypertension. Am. J. Physiol. Heart Circ. Physiol. 2017, 312, H368–H374. [Google Scholar] [CrossRef]
  116. Paudel, P.; van Hout, I.; Bunton, R.W.; Parry, D.J.; Coffey, S.; McDonald, F.J.; Fronius, M. Epithelial Sodium Channel δ Subunit Is Expressed in Human Arteries and Has Potential Association with Hypertension. Hypertension 2022, 79, 1385–1394. [Google Scholar] [CrossRef]
  117. Yen, W.; Cai, B.; Yang, J.; Zhang, L.; Zeng, M.; Tarbell, J.M.; Fu, B.M. Endothelial surface glycocalyx can regulate flow-induced nitric oxide production in microvessels in vivo. PLoS ONE 2015, 10, e0117133. [Google Scholar] [CrossRef]
  118. Florian, J.A.; Kosky, J.R.; Ainslie, K.; Pang, Z.; Dull, R.O.; Tarbell, J.M. Heparan sulfate proteoglycan is a mechanosensor on endothelial cells. Circ. Res. 2003, 93, e136–e142. [Google Scholar] [CrossRef] [Green Version]
  119. Bartosch, A.M.W.; Mathews, R.; Tarbell, J.M. Endothelial Glycocalyx-Mediated Nitric Oxide Production in Response to Selective AFM Pulling. Biophys. J. 2017, 113, 101–108. [Google Scholar] [CrossRef] [Green Version]
  120. Schierke, F.; Wyrwoll, M.J.; Wisdorf, M.; Niedzielski, L.; Maase, M.; Ruck, T.; Meuth, S.G.; Kusche-Vihrog, K. Nanomechanics of the endothelial glycocalyx contribute to Na(+)-induced vascular inflammation. Sci. Rep. 2017, 7, 46476. [Google Scholar] [CrossRef] [Green Version]
  121. Weissgerber, T.L.; Garcia-Valencia, O.; Milic, N.M.; Codsi, E.; Cubro, H.; Nath, M.C.; White, W.M.; Nath, K.A.; Garovic, V.D. Early Onset Preeclampsia Is Associated with Glycocalyx Degradation and Reduced Microvascular Perfusion. J. Am. Heart Assoc. 2019, 8, e010647. [Google Scholar] [CrossRef] [Green Version]
  122. Stanhewicz, A.E.; Nuckols, V.R.; Pierce, G.L. Maternal microvascular dysfunction during preeclamptic pregnancy. Clin. Sci. 2021, 135, 1083–1101. [Google Scholar] [CrossRef] [PubMed]
  123. Ley, K.; Laudanna, C.; Cybulsky, M.I.; Nourshargh, S. Getting to the site of inflammation: The leukocyte adhesion cascade updated. Nat. Rev. Immunol. 2007, 7, 678–689. [Google Scholar] [CrossRef] [PubMed]
  124. Sperandio, M.; Pickard, J.; Unnikrishnan, S.; Acton, S.T.; Ley, K. Analysis of leukocyte rolling in vivo and in vitro. Methods Enzymol. 2006, 416, 346–371. [Google Scholar]
  125. Machin, D.R.; Bloom, S.I.; Campbell, R.A.; Phuong, T.T.T.; Gates, P.E.; Lesniewski, L.A.; Rondina, M.T.; Donato, A.J. Advanced age results in a diminished endothelial glycocalyx. Am. J. Physiol. Heart Circ. Physiol. 2018, 315, H531–H539. [Google Scholar] [CrossRef]
  126. Bhat, S.; Marklund, M.; Henry, M.E.; Appel, L.J.; Croft, K.D.; Neal, B.; Wu, J.H.Y. A Systematic Review of the Sources of Dietary Salt Around the World. Adv. Nutr. 2020, 11, 677–686. [Google Scholar] [CrossRef] [PubMed]
  127. Hao, Z.; Liang, L.; Pu, D.; Zhang, Y. Analysis of Sodium Content in 4082 Kinds of Commercial Foods in China. Nutrients 2022, 14, 2908. [Google Scholar] [CrossRef] [PubMed]
  128. Ojo, A.E.; Jones, A.; Okoro, C.E.; Alfa, V.O.; Okoli, R.; Shedul, G.L.; Orji, I.A.; Osagie, S.; Chopra, A.; Van Horn, L.V.; et al. Sodium Content and Labelling of Packaged Foods and Beverages in Nigeria: A Cross-Sectional Study. Nutrients 2022, 15, 27. [Google Scholar] [CrossRef]
  129. Hunter, S.D.; Kavouras, S.A.; Rahimi, M. Exploring heated exercise as a means of preventing the deleterious effects of high-sodium intake in Black women. Am. J. Physiol. Heart Circ. Physiol. 2023, 324, H833–H839. [Google Scholar] [CrossRef]
  130. Wenner, M.M.; Welti, L.M.; Dow, C.A.; Greiner, J.J.; Stauffer, B.L.; DeSouza, C.A. Aerobic exercise training reduces ET-1-mediated vasoconstriction and improves endothelium-dependent vasodilation in postmenopausal women. Am. J. Physiol. Heart Circ. Physiol. 2023, 324, H732–H738. [Google Scholar] [CrossRef]
  131. Dow, C.A.; Stauffer, B.L.; Brunjes, D.L.; Greiner, J.J.; DeSouza, C.A. Regular aerobic exercise reduces endothelin-1-mediated vasoconstrictor tone in overweight and obese adults. Exp. Physiol. 2017, 102, 1133–1142. [Google Scholar] [CrossRef] [Green Version]
  132. Walker, J.C.; Dando, R. Sodium Replacement with KCl and MSG: Attitudes, Perception and Acceptance in Reduced Salt Soups. Foods 2023, 12, 2063. [Google Scholar] [CrossRef] [PubMed]
  133. Lee, C.L.; Lee, S.M.; Kim, K.O. Use of Consumer Acceptability as a Tool to Determine the Level of Sodium Reduction: A Case Study on Beef Soup Substituted with Potassium Chloride and Soy-Sauce Odor. J. Food Sci. 2015, 80, S2570–S2577. [Google Scholar] [CrossRef] [PubMed]
  134. Levings, J.L.; Gunn, J.P. The imbalance of sodium and potassium intake: Implications for dietetic practice. J. Acad. Nutr. Diet. 2014, 114, 838–841. [Google Scholar] [CrossRef] [PubMed]
  135. Carey, R.M.; Moran, A.E.; Whelton, P.K. Whelton, Treatment of Hypertension: A Review. JAMA 2022, 328, 1849–1861. [Google Scholar] [CrossRef] [PubMed]
  136. Shen, D.; Song, H.; Zou, T.; Raza, A.; Li, P.; Li, K.; Xiong, J. Reduction of sodium chloride: A review. J. Sci. Food Agric. 2022, 102, 3931–3939. [Google Scholar] [CrossRef]
  137. Drummond, H.A.; Gebremedhin, D.; Harder, D.R. Degenerin/epithelial Na+ channel proteins: Components of a vascular mechanosensor. Hypertension 2004, 44, 643–648. [Google Scholar] [CrossRef] [Green Version]
  138. Pérez, F.R.; Venegas, F.; González, M.; Andrés, S.; Vallejos, C.; Riquelme, G.; Sierralta, J.; Michea, L. Endothelial epithelial sodium channel inhibition activates endothelial nitric oxide synthase via phosphoinositide 3-kinase/Akt in small-diameter mesenteric arteries. Hypertension 2009, 53, 1000–1007. [Google Scholar] [CrossRef] [Green Version]
  139. Guan, Z.; Pollock, J.S.; Cook, A.K.; Hobbs, J.L.; Inscho, E.W. Effect of epithelial sodium channel blockade on the myogenic response of rat juxtamedullary afferent arterioles. Hypertension 2009, 54, 1062–1069. [Google Scholar] [CrossRef] [Green Version]
  140. Pitzer, A.L.; Van Beusecum, J.P.; Kleyman, T.R.; Kirabo, A. ENaC in Salt-Sensitive Hypertension: Kidney and Beyond. Curr. Hypertens. Rep. 2020, 22, 69. [Google Scholar] [CrossRef]
  141. Lemmens-Gruber, R.; Tzotzos, S. The Epithelial Sodium Channel—An Underestimated Drug Target. Int. J. Mol. Sci. 2023, 24, 7775. [Google Scholar] [CrossRef]
  142. Fedorov, D.A.; Sidorenko, S.V.; Yusipovich, A.I.; Bukach, O.V.; Gorbunov, A.M.; Lopina, O.D.; Klimanova, E.A. Increased Extracellular Sodium Concentration as a Factor Regulating Gene Expression in Endothelium. Biochemistry 2022, 87, 489–499. [Google Scholar] [CrossRef]
  143. Huang, L.; Zhang, X.; Ma, X.; Zhang, D.; Li, D.; Feng, J.; Pan, X.; Lü, J.; Wang, X.; Liu, X. Berberine alleviates endothelial glycocalyx degradation and promotes glycocalyx restoration in LPS-induced ARDS. Int. Immunopharmacol. 2018, 65, 96–107. [Google Scholar] [CrossRef] [PubMed]
  144. Mulivor, A.W.; Lipowsky, H.H. Inhibition of glycan shedding and leukocyte-endothelial adhesion in postcapillary venules by suppression of matrixmetalloprotease activity with doxycycline. Microcirculation 2009, 16, 657–666. [Google Scholar] [CrossRef] [PubMed]
  145. Zeng, Y.; Adamson, R.H.; Curry, F.R.; Tarbell, J.M. Sphingosine-1-phosphate protects endothelial glycocalyx by inhibiting syndecan-1 shedding. Am. J. Physiol. Heart Circ. Physiol. 2014, 306, H363–H372. [Google Scholar] [CrossRef]
  146. Ramnath, R.D.; Butler, M.J.; Newman, G.; Desideri, S.; Russell, A.; Lay, A.C.; Neal, C.R.; Qiu, Y.; Fawaz, S.; Onions, K.L.; et al. Blocking matrix metalloproteinase-mediated syndecan-4 shedding restores the endothelial glycocalyx and glomerular filtration barrier function in early diabetic kidney disease. Kidney Int. 2020, 97, 951–965. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Zeng, Y.; Liu, X.H.; Tarbell, J.; Fu, B. Sphingosine 1-phosphate induced synthesis of glycocalyx on endothelial cells. Exp. Cell Res. 2015, 339, 90–95. [Google Scholar] [CrossRef] [PubMed]
  148. Lepedda, A.J.; Nieddu, G.; Piperigkou, Z.; Kyriakopoulou, K.; Karamanos, N.; Formato, M. Circulating Heparan Sulfate Proteoglycans as Biomarkers in Health and Disease. Semin. Thromb. Hemost. 2021, 47, 295–307. [Google Scholar] [CrossRef]
  149. Huang, X.; Hu, H.; Sun, T.; Zhu, W.; Tian, H.; Hao, D.; Wang, T.; Wang, X. Plasma Endothelial Glycocalyx Components as a Potential Biomarker for Predicting the Development of Disseminated Intravascular Coagulation in Patients with Sepsis. J. Intensive Care Med. 2021, 36, 1286–1295. [Google Scholar] [CrossRef] [PubMed]
  150. Uchimido, R.; Schmidt, E.P.; Shapiro, N.I. The glycocalyx: A novel diagnostic and therapeutic target in sepsis. Crit. Care 2019, 23, 16. [Google Scholar] [CrossRef] [Green Version]
  151. Mony, V.K.; Benjamin, S.; O’Rourke, E.J. A lysosome-centered view of nutrient homeostasis. Autophagy 2016, 12, 619–631. [Google Scholar] [CrossRef] [Green Version]
  152. Baby, S.; Reljic, T.; Villalba, N.; Kumar, A.; Yuan, S.Y. Endothelial glycocalyx-associated molecules as potential serological markers for sepsis-associated encephalopathy: A systematic review and meta-analysis. PLoS ONE 2023, 18, e0281941. [Google Scholar] [CrossRef] [PubMed]
  153. Hahn, R.G.; Patel, V.; Dull, R.O. Human glycocalyx shedding: Systematic review and critical appraisal. Acta Anaesthesiol. Scand. 2021, 65, 590–606. [Google Scholar] [CrossRef]
  154. Rehm, M.; Bruegger, D.; Christ, F.; Conzen, P.; Thiel, M.; Jacob, M.; Chappell, D.; Stoeckelhuber, M.; Welsch, U.; Reichart, B.; et al. Shedding of the endothelial glycocalyx in patients undergoing major vascular surgery with global and regional ischemia. Circulation 2007, 116, 1896–1906. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Torres Filho, I.P.; Torres, L.N.; Salgado, C.; Dubick, M.A. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Bol, M.E.; Huckriede, J.B.; van de Pas, K.G.H.; Delhaas, T.; Lorusso, R.; Nicolaes, G.A.F.; Sels, J.E.M.; van de Poll, M.C.G. Multimodal measurement of glycocalyx degradation during coronary artery bypass grafting. Front. Med. 2022, 9, 1045728. [Google Scholar] [CrossRef]
  157. Matyjaszczyk-Gwarda, K.; Kij, A.; Olkowicz, M.; Fels, B.; Kusche-Vihrog, K.; Walczak, M.; Chlopicki, S. Simultaneous quantification of selected glycosaminoglycans by butanolysis-based derivatization and LC-SRM/MS analysis for assessing glycocalyx disruption in vitro and in vivo. Talanta 2022, 238 Pt 1, 123008. [Google Scholar] [CrossRef]
  158. Yeo, T.W.; Weinberg, J.B.; Lampah, D.A.; Kenangalem, E.; Bush, P.; Chen, Y.; Price, R.N.; Young, S.; Zhang, H.Y.; Millington, D.; et al. Glycocalyx Breakdown Is Associated with Severe Disease and Fatal Outcome in Plasmodium falciparum Malaria. Clin. Infect. Dis. 2019, 69, 1712–1720. [Google Scholar] [CrossRef] [Green Version]
  159. Oberleithner, H. Quantifying salt sensitivity. Biol. Chem. 2021, 402, 1597–1602. [Google Scholar] [CrossRef]
  160. Oberleithner, H.; Wilhelmi, M. Salt Sensitivity Determined From Capillary Blood. Kidney Blood Press. Res 2016, 41, 355–364. [Google Scholar] [CrossRef]
  161. Masenga, S.K.; Pilic, L.; Malumani, M.; Hamooya, B.M. Erythrocyte sodium buffering capacity status correlates with self-reported salt intake in a population from Livingstone, Zambia. PLoS ONE 2022, 17, e0264650. [Google Scholar] [CrossRef]
  162. Vinaiphat, A.; Pazhanchamy, K.; JebaMercy, G.; Ngan, S.C.; Leow, M.K.S.; Ho, H.H.; Gao, Y.G.; Lim, K.L.; Richards, A.M.; de Kleijn, D.P.; et al. Endothelial Damage Arising From High Salt Hypertension Is Elucidated by Vascular Bed Systematic Profiling. Arterioscler. Thromb. Vasc. Biol. 2023, 43, 427–442. [Google Scholar] [CrossRef] [PubMed]
  163. McNally, R.J.; Morselli, F.; Farukh, B.; Chowienczyk, P.J.; Faconti, L. A pilot study to evaluate the erythrocyte glycocalyx sensitivity to sodium as a marker for cellular salt sensitivity in hypertension. J. Hum. Hypertens. 2023, 37, 286–291. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Components of the vascular endothelial cell surface glycocalyx. CD-44: cluster of differentiation-44.
Figure 1. Components of the vascular endothelial cell surface glycocalyx. CD-44: cluster of differentiation-44.
Nutrients 15 02873 g001
Figure 2. Vascular endothelial glycocalyx buffering sodium.
Figure 2. Vascular endothelial glycocalyx buffering sodium.
Nutrients 15 02873 g002
Figure 3. Proposed model of salt-induced damage to the glycocalyx that results in hypertension and cardiovascular disease. High intake of salt damages the glycocalyx and induces inflammation, oxidative stress, and immune activation, thus leading to the development of hypertension and cardiovascular disease. ROS, reactive oxygen species; ENaC, epithelial sodium channel; NADPH, reduced nicotinamide adenine dinucleotide phosphate; NLRP3, NLR Family Pyrin Domain Containing 3; NF-Κb, Nuclear factor kappa-light-chain-enhancer of activated B cells; IsoLGs, Islovuglandins; TNF-α, tumor necrosis factor alpha; IFN-γ, Interferon gamma.
Figure 3. Proposed model of salt-induced damage to the glycocalyx that results in hypertension and cardiovascular disease. High intake of salt damages the glycocalyx and induces inflammation, oxidative stress, and immune activation, thus leading to the development of hypertension and cardiovascular disease. ROS, reactive oxygen species; ENaC, epithelial sodium channel; NADPH, reduced nicotinamide adenine dinucleotide phosphate; NLRP3, NLR Family Pyrin Domain Containing 3; NF-Κb, Nuclear factor kappa-light-chain-enhancer of activated B cells; IsoLGs, Islovuglandins; TNF-α, tumor necrosis factor alpha; IFN-γ, Interferon gamma.
Nutrients 15 02873 g003
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Sembajwe, L.F.; Ssekandi, A.M.; Namaganda, A.; Muwonge, H.; Kasolo, J.N.; Kalyesubula, R.; Nakimuli, A.; Naome, M.; Patel, K.P.; Masenga, S.K.; et al. Glycocalyx–Sodium Interaction in Vascular Endothelium. Nutrients 2023, 15, 2873. https://doi.org/10.3390/nu15132873

AMA Style

Sembajwe LF, Ssekandi AM, Namaganda A, Muwonge H, Kasolo JN, Kalyesubula R, Nakimuli A, Naome M, Patel KP, Masenga SK, et al. Glycocalyx–Sodium Interaction in Vascular Endothelium. Nutrients. 2023; 15(13):2873. https://doi.org/10.3390/nu15132873

Chicago/Turabian Style

Sembajwe, Lawrence Fred, Abdul M. Ssekandi, Agnes Namaganda, Haruna Muwonge, Josephine N. Kasolo, Robert Kalyesubula, Annettee Nakimuli, Mwesigwa Naome, Kaushik P. Patel, Sepiso K. Masenga, and et al. 2023. "Glycocalyx–Sodium Interaction in Vascular Endothelium" Nutrients 15, no. 13: 2873. https://doi.org/10.3390/nu15132873

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop