Next Article in Journal
Upcycling Polystyrene
Next Article in Special Issue
Starch Physicochemical Properties of Normal Maize under Different Fertilization Modes
Previous Article in Journal
Optimal Surface Pre-Reacted Glass Filler Ratio in a Dental Varnish Effective for Inhibition of Biofilm-Induced Root Dentin Demineralization
Previous Article in Special Issue
Grafted Pullulan Derivatives for Reducing the Content of Some Pesticides from Simulated Wastewater
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Alterations and Roles of Glycosaminoglycans in Human Diseases

National Glycoengineering Research Center, Shandong University, Qingdao 266237, China
*
Author to whom correspondence should be addressed.
Polymers 2022, 14(22), 5014; https://doi.org/10.3390/polym14225014
Submission received: 14 October 2022 / Revised: 14 November 2022 / Accepted: 15 November 2022 / Published: 18 November 2022
(This article belongs to the Special Issue Natural Polysaccharide: Synthesis, Modification and Application)

Abstract

:
Glycosaminoglycans (GAGs) are a heterogeneous family of linear polysaccharides which are composed of a repeating disaccharide unit. They are also linked to core proteins to form proteoglycans (PGs). GAGs/PGs are major components of the cell surface and the extracellular matrix (ECM), and they display critical roles in development, normal function, and damage response in the body. Some properties (such as expression quantity, molecular weight, and sulfation pattern) of GAGs may be altered under pathological conditions. Due to the close connection between these properties and the function of GAGs/PGs, the alterations are often associated with enormous changes in the physiological/pathological status of cells and organs. Therefore, these GAGs/PGs may serve as marker molecules of disease. This review aimed to investigate the structural alterations and roles of GAGs/PGs in a range of diseases, such as atherosclerosis, cancer, diabetes, neurodegenerative disease, and virus infection. It is hoped to provide a reference for disease diagnosis, monitoring, prognosis, and drug development.

1. Introduction

Glycosaminoglycan is a kind of unbranched linear anionic polysaccharide composed of 10 to 200 repeating disaccharide units [1]. Each disaccharide unit consists of one hexuronic acid (except keratan sulfate) and one hexosamine. The hydroxyls and/or acetyls on these disaccharide units are substituted by sulfate groups to varying degrees. Modification and isomerization of sugar residues leads to greater molecular diversity of GAGs. According to the structural characteristics of the disaccharide unit, GAGs can be divided into four classes: heparin/heparan sulfate (HP/HS), chondroitin/dermatan sulfate (CS/DS), keratan sulfate (KS), and hyaluronan (HA) [2].
The disaccharide structure of HP/HS isα-L-IdoA/β-D-GlcA (1→4) α-D-GlcNS/GlcNAc (1→4). HP/HS is typically 2-O-sulfated (2-O-S) at uronic acid residues and N-sulfated (N-S), 6-O-sulfated (6-O-S), and 3-O-sulfated (3-O-S) at glucosamine (GlcN) residues. Compared with HS, HP has a higher iduronic acid (IdoA) content (>70% in uronic acid) and sulfate/disaccharide ratio (about 2.3 sulfate groups per disaccharide in HP compared to 0.8 sulfate groups per disaccharide in HS) [3]. HP mainly exists in the intracellular granules of mast cells, and it possesses significant anticoagulant and other activities, such as antiviral and antitumor metastasis [4]. HS is in the extracellular matrix (ECM) and plays an important role in cell growth, immune response, tissue homeostasis, and embryonic development [5,6]. The disaccharide structure of CS/DS isβ-D-GlcA/α-L-IdoA (1→3) β-D-GalNAc (1→4). The 4-O, 6-O positions on GalNAc and 2-O position on Glc/IdoA can be sulfated. In addition to playing a structural role in organs and tissues (e.g., skin and cartilage), CS/DS was, in recent years, found to be involved in important biological processes [7,8] such as tumorigenesis and metastasis [9], nervous system development [10,11], and immune regulation [12]. KS differs from HS and CS/DS in that it takesβ-D-Gal (1→4) β-D-GlcNAc (1→3) as a disaccharide unit with 6-O-S at galactose and glucosamine [13]. KS is widely distributed in the body, such as in the eyes, brain, cartilage, and epithelial tissue. Therefore, KS has multiple physiological functions and plays an important role in the regulation of neuronal charge and ion gradients [14], cell adhesion, proliferation, and differentiation [15]. HA is the only nonsulfated polysaccharide in the GAG family, with a repeating disaccharide unit ofβ-D-GlcA (1→3) β-D-GlcNAc (1→4). HA is the largest-molecular-weight GAG (from 105 Da to 107 Da) and is abundant in connective tissues, such as synovial and vitreous fluid. HA not only provides compressive strength, lubricity, and hydration in the ECM, but also modulates cell adhesion metastasis, inflammation, and tissue homeostasis [16].
Most GAGs usually do not exist alone in vivo. In addition to HA, GAGs are linked to core proteins to form N-linked/O-linked proteoglycans (PGs) [17]. For example, HS and CS/DS chains are linked to the serine of core proteins via a tetrasaccharide unit of GlcA (1→3) β-Gal (1→3) β-Gal (1→4) β-Xyl (1→4). In fact, they are products of posttranslational modifications (glycosylation) of proteins. PGs are widely present on the cell surface, extracellular matrix (ECM), and basement membrane (BMs) (Figure 1). They participate in various life activities. GAGs/PGs are considered to be the most complex and informative biomolecules in organisms due to the differences in glycosidic bond types, polymerization, sulfation patterns, monosaccharide types, and core proteins [18]. Therefore, GAGs/PGs are indispensable for exerting the normal physiological functions of cells. They not only act as traditional supporting structures, but also interact with many extracellular signaling molecules, binding proteins, and enzymes to participate in different biological processes [17,19,20,21].
It is well known that the structure and distribution of GAG/PGs are different in various cells/tissues because they are affected by tissue-specific expression, activity level, and specificity of biosynthetic and degradative enzymes [22,23,24]. The decoration patterns of GAG chains influence and determine their specific interactions with natural protein ligands. This also reflects the physiological or pathological state of cells or tissue to some extent. In this review, we focus on the structural alterations and functions of GAGs/PGs in major human diseases (atherosclerosis, cancer, diabetes, neurodegenerative disease, and virus infections), hoping to provide a reference for disease diagnosis, monitoring, prognosis, and drug development.

2. GAGs in Atherosclerosis

Cardiovascular disease (CVD) is the leading cause of death in many countries [25]. CVD can be caused by many factors, such as genetics, diseases (e.g., diabetes [26,27]), and an unhealthy lifestyle (e.g., alcoholism and smoking). In addition, oxidative stress [28] and inflammation [29,30] are thought to be involved in the pathogenesis of CVD. Atherosclerosis is the most prevalent and clinically significant CVD, and it is closely related to other diseases, such as retinopathy, neuropathy, and nephropathy [31,32,33]. Human atherosclerosis develops in different stages, usually including intimal hyperplasia and lipid accumulation, foam cell formation, plaque formation and growth, plaque rupture, and thrombosis [34]. GAGs/PGs are the main components of the ECM in vascular wall cells, such as endothelial cells, smooth muscle cells (SMCs), and adventitial fibroblasts. Therefore, GAGs/PGs play a crucial role in regulating vascular permeability and maintaining homeostasis of the vascular environment [35,36].
CS and DS are the major GAG components of arterial vessels. The most abundant CSPG in the vascular ECM is versican [37,38]. DSPGs mainly include decorin and biglycan, which belong to the small leucine-rich repeat family. GAG chains of CS/DSPGs perform important functions in binding to lipoproteins and regulating elastin synthesis [39]. Moreover, CS/DSPGs have been found to be significantly elevated in early atherosclerotic lesions. They can promote lipid retention and accumulation, which is considered to be the initial factor in the development of atherosclerosis [40,41]. For instance, versican and biglycan strongly bind both native LDL and OxLDL [42]. Decorin binding collagen type I can promote LDL binding and enhance lipoprotein accumulation in the vascular wall [43]. Moreover, LDL–GAG complexes can be internalized by macrophages and SMCs [12,44] together with proliferation and migration of SMCs. This leads to intracellular lipid accumulation, foam cell formation, and intimal hyperplasia, which are considered hallmarks of early atherosclerosis [45]. In addition, CS chains can interact with elastin-binding protein (EBP) on the cell surface, inhibit the assembly of elastic fibers, and increase the rigidity of the vascular wall [46]. The CS/DS chain length, charge density, and sulfation pattern influences CS/DSPG binding to lipoproteins [47]. A study showed that the domains of 6-O-S or 4-O-S galactosamine (GalN) underlie CS/DS binding to lipoproteins [48]. The content of CS containing 4-O-S or 6-O-S in the aorta is 30% higher than in other arteries, which binds lipoproteins more tightly [49]. Theocharis et al. demonstrated an increase in both total CS content and the ratio of 6-O-S/4-O-S disaccharides in type II atherosclerosis arteries. This also denotes an increase in the CS/DS ratio, since CS and DS are the main providers of 6-O-S disaccharides and 4-O-S disaccharides, respectively [50]. In addition, transforming growth factor (TGF-β) in atherosclerotic vessels prolongs CS chains in arterial smooth muscle cells (ASMCs), thereby increasing versican binding to LDL [51]. Therefore, CS/DS can promote the development of atherosclerosis under pathological conditions.
The major HSPG in vascular endothelial cells and SMCs is perlecan [52]. Earlier studies showed that vascular HSPG levels are decreased and negatively correlated with cholesterol levels in atherosclerosis patients [53,54,55]. Thus, HSPG shows the opposite trend to CS/DSPGs in atherosclerosis. The reduction in HSPG could increase the binding of lipoprotein A to the ECM in endothelial cells, which may be related to the fact that OxLDL encourages endothelial cells to produce more heparinase [56]. Heparanase leads to abnormal HS degradation, which, in turn, results in increasing vascular endothelial permeability and SMC migration. This suggests that rising HSPG synthesis may protect against atherosclerosis [57]. Moreover, as mentioned earlier, large numbers of macrophages are recruited in atherosclerotic lesions. HSPG in macrophage ECM has also been implicated in atherogenesis. Asplund et al. found that HS depletion on the surface of human macrophages (HMDM) under hypoxia enhanced cell motility and accelerated plaque formation [58].
Deposition of lipids in the vascular wall promotes proliferation and migration of SMCs, where HA appears to play a major role [59]. HA in the arterial wall is mainly produced by SMCs, and synthesis increases in atherosclerosis [60]. A study showed that HA accumulation could promote SMC metastasis via ERK1/2 modulation of the CD44 signaling pathway, resulting in intimal hyperplasia [61]. HA also binds to LDL and is more readily internalized by macrophages than native LDL [62]. Low-molecular-weight hyaluronic acid (LMW-HA) is considered to induce inflammation in vivo, whereas high-molecular-weight HA (HMW-HA) does not [63,64]. Tabata et al. found that LMW-HA, which is abundantly produced in atherosclerotic lesions, participates in the inflammatory mechanism of atherosclerotic plaque formation by promoting monocyte migration and foam macrophage differentiation through binding to CD44 [65]. In addition, HA can interact with versican to remodel the ECM of diseased SMCs and promote the proliferation and migration of SMCs [44]. In short, LDL internalization impels macrophages to release more cytokines and growth factors, which changes the behavior of SMCs. Meanwhile, overexpression of HA and its receptor CD44 further aggravated cell migration and intimal hyperplasia.
In the process of atherosclerosis, the content of total CS and 6-O-S of CS are increased, enhancing binding to lipids, and LDL-GAG complexes can be internalized by macrophages and SMCs, promoting the formation of sclerotic plaques. In addition, HS depletion in the ECM at the lesion resulted in increased vascular endothelial permeability, promoting inflammatory cell infiltration and cell migration. The synthesis of LMW-HA is increased, which is involved in the inflammatory mechanism of atherosclerotic plaque formation. In conclusion, GAGs/PGs participate in ECM remodeling and play multiple roles in regulating immune adhesion, as well as promoting lipid accumulation, intimal hyperplasia, and thrombosis. Therefore, vascular GAGs/PGs have potential as a target for the prevention of atherosclerosis.

3. GAGs in Cancers

Cancer is the second leading cause of death worldwide [66,67]. Cancer possesses biological characteristics, such as uncontrolled cell differentiation and proliferation, invasion, and metastasis. Its occurrence is a multifactorial and multistep complex process [68]. Cancer is defined as many types, such as liver, lung, breast, and ovarian cancer, depending on where the cancer is located. During cancer development, growth, metastasis, and invasion require specific interactions between tumor cells and the tumor microenvironment (TME) [69,70,71]. GAGs/PGs, as the critical effectors of the cell surface and TME, are involved in tumor growth and metastasis through interacting with growth factors, growth factor receptors, and cytokines [69,72,73,74]. Importantly, GAGs/PGs play a vital role in cancer regulation in terms of types, molecular weight, distribution, and fine modification. Thus, GAGs/PGs can become potential targets for anticancer therapy [75,76].
HSPG (mainly perlecans, syndecans, and glypicans) are considered central molecules regulating cell behavior and cancer progression [77]. HSPGs are differentially expressed in diverse cancers. For example, in breast cancer, perlecan was absent in epithelial cell BM, while being markedly upregulated in stroma. Furthermore, plasma perlecan level was significantly higher in estrogen receptor (ER)+ patients than ER patients [78]. Perlecan expression is increased in invasive and metastatic prostate cancer cells [79]. Syndecan-2 was significantly increased in well-differentiated neuroendocrine tumors (NETs) and significantly decreased in poorly differentiated NETs. Glypican-5 was overexpressed in high-grade tumors with epithelial differentiation, but not in tumors with neuroendocrine phenotype [80]. Indeed, the sulfation pattern of GAG chains is also strongly related to cancer type and differentiated degree [81,82,83,84,85,86]. For instance, Weyers et al. examined changes in the GAGs of fatal and nonfatal breast cancer tissues. The GAG length increased by approximately 15% in tumor tissue compared to normal tissue. Both the 6-O-S CS and the total sulfation of HS increased. Compared to nonfatal breast cancers, the sulfation degree of HS, particularly 6-O-S, was decreased in fatal breast cancers, whereas the proportion of non-sulfated disaccharides was increased [87]. In addition, the expression profile of HS in cancer and its role in cancer regulation have been detailed in a recent review [88].
Many studies have indicated that N-S, 2-O-S, and 6-O-S of HS play an important regulatory role in tumor metastasis and invasion, especially 6-O-S [89]. For example, fibroblast growth factor/fibroblast growth factor receptor (FGF/FGFR) promotes cardiovascular generation and endothelial cell repair, playing a key role in the regulation of lesion metabolism [90] (Figure 2). Therefore, FGF/FGFR is widely regarded as a potential target for antitumor therapy [91,92]. HS promotes cell signal transduction by binding to FGF2/FGFRs to form ternary complexes [93,94]. In this process, HS binding to FGF2 requires the N-S of GlcN and 2-O-S of IdoA [95]. In addition to N-S and 2-O-S, HS requires 6-O-S of GlcN to bind to FGFR. Similarly, IdoA with 2-O-S and GlcN with 6-O-S and N-S are also essential for binding HS to FGF1 [96]. Specific sequences for HS binding to FGF family proteins were further identified by Kreuger et al. They found that HS octasaccharides binding to FGF1 contained an IdoA (2S)–GlcNS (6S)–IdoA (2S) trisaccharide sequence, and HS binding to FGF2 contained an IdoA (2S)–GlcNS–IdoA (2S) trisaccharide sequence [97]. Schultz et al. determined the fine structure of HS binding to FGF12–FGFR1c2 from chemically enzymatically synthesized HS octasaccharide, and they pointed out that the N-S of the nonreducing terminal residue is essential for binding [98]. In addition, HS on the tumor cells surface showed a stronger affinity for NT4 (a tetrapeptide) compared with other GAGs [99,100]. NT4 can target cell lines of different human cancers; therefore, it may serve as a carrier for the delivery of anticancer drugs or tumor imaging tracers [101,102,103,104,105]. More importantly, NT4 may inhibit the migration of pancreatic cancer cells, as well as the growth factor-induced invasion of breast cancer cells, by binding to HS [99,106]. Brunetti et al. demonstrated the decisive role of sulfate groups on HS for binding to NT4 and determined that possible binding sequences include repeated disaccharide units of uronic acid with 2-O-S and GlcN with 6-O-S and N-S [107]. Interestingly, Liu et al. treated tumor cells with heparinases I and III, respectively. They demonstrated that HS on the cell surface has specific sequences for both tumor activation and inhibitory activity [108]. It was proven that the tumor metabolic regulatory effect of HS is closely linked to its specific sulfation patterns.
CS/DS is equally important during tumor cell proliferation, migration, adhesion, and invasion [109]. In some cancer tissues, the CS content is increased [110,111,112,113,114]. For example, glioblastoma multiforme (GBM) tumors with increased CSPG content accounted for 65% of the total number of samples through clinical studies by Tsidulko et al. [115]. CSPG, such as versican, is highly expressed in breast cancer, which is associated with an unfavorable prognosis [116]. For a few cases, such as invasive gliomas, several studies have shown that the GAG chain of CS/DSPGs in the tumor ECM is severely absent [117,118,119,120,121]. Abnormal glycosylation may be associated with isomeric conversion and lyase activity of CSPGs [120,121,122,123]. The change of DS depends on cancer type. DS levels are elevated in liver cancer [124], lung cancer [125], gastric cancer [126], pancreatic cancer [127], and colorectal cancer [113]. Although the content and distribution of CS/DS are heterogeneous in tumor tissues, several studies have shown that 6-O-S and non-sulfated disaccharide levels are increased in some tumors, while 4-O-S disaccharide levels are decreased [128,129]. For example, increased CS content is observed in pancreatic cancer, characterized by a significantly enhanced expression of 6-O-S and non-sulfated disaccharide units [127,130]. Prostate cancer has elevated 4-O-S CS content in the ECM, which may be due to inhibited androgen receptor (AR) signaling, thus resulting in increased 4-O-sulfotransferase CHST11 expression [131]. CS testing of human gastric cancer tissues revealed a 10-fold increase in 6-O-S and non-disaccharide units, while 4S disaccharides were correspondingly decreased [132]. In addition, CS with 4, 6-O-S (CS-E) is increased in a variety of cancers. A high expression of CS-E in the ECM of ovarian adenocarcinoma enhances vascular endothelial growth factor (VEGF) mediation [133]. The proportion of Δ4.5HexA-GalNAc-4, 6-O-disulfate was higher in highly metastatic lung cancer cell lines than in low metastatic cell lines [134]. The results suggest complex changes in the sulfuryl modification of CS/DS during carcinogenesis. A possible reason is that, on the one hand, aberrant expression of the PG core protein leads to changes in the type and structure of the linked GAG chain [135]. On the other hand, CS/DS synthesis or modification is mediated by abnormalities in enzyme activity or levels [136,137,138]. However, the reduction in DS in tumor tissue may be more conducive to cancer development and metastasis. Therefore, it has been suggested that DS has antitumor activity. For instance, DS/DSPG has an inhibitory effect on the proliferation and migration of certain osteosarcoma and melanoma cell lines [139,140,141]. However, there seems to be controversy regarding the role of DS in cancer. DS was found to promote proliferation of esophageal squamous cells [142]. Therefore, the role of DS in cancer needs further investigation.
HA has the dual properties of tumor promotion and tumor inhibition. LMW-HA predominates in normal tissues and is essential for maintaining tissue homeostasis. Studies have shown that HA content increases in many types of human cancers [143,144,145]. Moreover, HA molecular weight decreases in the TME [146,147,148]. LMW-HA stimulates the expression of chemokines and growth factors, as well as promotes tumor cell adhesion and migration [149,150]. The role of LMW-HA in cancer is associated with HA receptors (e.g., CD44, and HA-mediated mobility receptor (RHAMM)) [151,152]. In addition, HA has been reported to be degraded by hyaluronidase into smaller oligosaccharide fragments, inducing the division of CD44, thereby enhancing tumor cell viability in breast, ovarian, and glial tumors, and colon cancers [153,154].
For other GAGs, Yukinari et al. found that highly sulfated KS was overexpressed in malignant astrocytic tumors using histochemistry [155].
In short, GAGs/PGs are critical regulators for cancer cell proliferation and metastasis. Although changes in GAGs/PGs vary widely in different types and stages of cancer, overall HS expression is upregulated in cancer cells relative to normal cells. As important signaling molecules on the cell surface, HS overexpression increases the communication between cancer cells and the external environment, which supports the characteristics of cancer cells that are prone to proliferation and metastasis. During this period, N-S, 2-O-S, 6-O-S of HS play an important regulatory role. CS/CSPGs are also upregulated in some tumors, and at the same time, they are accompanied by increased 6-O-S levels and decreased 4-O-S levels. DS/DSPG is detected in some tumors, which suggests that DS may have antitumor activity. LMW-HA increases in many types of cancer and promotes tumor cell adhesion and migration. Thus, GAGs/PGs play a crucial role in tumor cell activity. In addition, GAGs perform their functions by interacting with proteins in the body. Therefore, the development of some GAGs analogues to inhibit their interactions may become another approach for cancer treatment [156].

4. GAGs in Diabetes Mellitus

Diabetes mellitus (diabetes, DM) is a metabolic disorder caused by decreased insulin levels due to autoimmune β-cell destruction (T1D) and/or insulin resistance (T2D) [127,157]. Diabetes is one of the fastest-growing diseases, with approximately 415 million patients worldwide in 2015, and this number is expected to grow to 693 million by 2045 [157,158]. The development of diabetes can cause systemic diseases, such as cardiovascular disease, diabetic nephropathy, retinopathy, and neuropathy. The ECM of various tissues or organs acts as the first line of defense against pathological factors, and ECM remodeling plays a crucial part in the development of diabetes and complications [159].
In normal pancreatic β-cells, high levels of HSPG are necessary for cell survival. In normal metabolism, HSPG can not only prevent the invasion of immune cells and prevent the degradation by heparanase, but also act as a nonenzymatic antioxidant that can scavenge ROS in a timely manner and avoid oxidative damage to cells [160,161]. In studies of T1D, HSPGs such as Col18 and syndecan-1 were found to be highly expressed in normal human islets. Importantly, highly sulfated HS was specifically expressed in normal human β-cells, while highly sulfated HS was expressed in α-cells [162]. Further studies revealed that HS in β-cells largely contain 2-O-S, 6-O-S, and N-S, whereas in α cells, HS contains N-acetyl, N-S, and 2-O-S, with fewer 6-O-S. Moreover, HS mediates cell activity through FGF/FGFRs [163] (the effect of HS on the FGF/FGFR signaling pathway was presented in the Section 3). When β-cell dysfunction occurs, it is often accompanied by inflammation such as pancreatitis; leukocytes infiltrate islet cells, release heparanase to degrade HSPG, gradually reduce islet HS, and injure β-cells [160,164,165]. Low-sulfated HS decreases β-cell proliferation and viability by mediating FGF/FGFR signaling [163]. At the same time, the decrease in the content of highly sulfated HS renders β-cells highly susceptible to oxidant-mediated damage, while high levels of endogenous ROS can depolymerize HS [166,167]. In addition, for HSPG alterations in T2D, Simeonovic et al. proposed the “ER stress” model: β-cell endoplasmic reticulum (ER) homeostasis is disrupted during the pathological process of T2D, resulting in ER luminal misfolded protein accumulation and ER stress [168,169]. ER stress initiates the unfolded protein response (UPR), which alleviates stress by increasing chaperones for protein folding and decreasing normal protein synthesis [170]. This impaired HSPG core protein synthesis further hinders HS synthesis. HS acts as a nonenzymatic antioxidant in β-cells, and its lower expression increases intracellular ROS and promotes oxidative stress, ultimately leading to the apoptosis of β-cells [171,172]. Thus, a high expression of highly sulfated HS is a hallmark of healthy β-cells. Alternatively, β-cells are metabolically disturbed under pathological conditions, and islet amyloid polypeptide (IAPP) undergoes misfolded aggregation, resulting in deposition. IAPP is a hormonal peptide implicated in T2D pathogenesis and progression [173]. GAGs were found to be involved in the deposition of IAPP within or around pancreatic β-cells [174]. Castillo et al. investigated the interaction of different sulfated GAGs, including perlecan, with amyloid in vitro and found that GAG sulfation patterns affected amyloid fibril formation. The order of impact was heparin > N-desulfated acetylated heparin > fully desulfated N-sulfated heparin > fully desulfated N-acetylated heparin [175]. Recent studies have shown that PGs on the surface of islet cells, especially HS/HSPGs, are able to promote islet amyloid deposition and IAPP-induced cytotoxicity, thereby accelerating T1D progression [176,177]. Nevertheless, there is little information on other GAGs involved in islet cell lesions. Only a few reports mentioned that HA levels were significantly elevated in immune cells both inside and outside islet cells and at inflammatory sites in T1D [178,179].
Diabetic nephropathy (DN) is one of the most important complications of diabetes and is characterized by proteinuria [180]. Earlier studies have shown that reduced HS levels in diabetic glomerular basement membranes (GBM) are associated with proteinuria [181]. The altered HS sulfation pattern of GBM in diabetic nephropathy may be due to increased heparanase levels [182] or altered sulfatase regulation, and it has been demonstrated that 6-O-S of HS plays an important role in maintaining the glomerular filtration barrier [183]. In another study, HS was found to have less N-S in the GBM of diabetic rats compared to the normal group [184]. Paradoxically, however, some researchers have demonstrated that the structure of glomerular HS is not affected in diabetic rat models [185,186]. Compared with HS, CS/DS alterations were more significant in the kidney. In diabetic rats, renal CS/DS content decreased, accompanied by a decrease in the degree of sulfation, particularly 4, 6-O-sulfated GalN content [187]. Another study described CS alterations in the kidney in more detail. Reine et al. found that 4-O-disaccharide sulfate significantly decreased from 65% to 40%, whereas 6-O-S disaccharide decreased from 11% to 6% and non-sulfated disaccharide increased from 21.5% to 51% in the renal cortex of diabetic db/db mice [186]. Alterations in CS/DS structure in the kidney of patients with DN have an impact on the composition and function of the ECM in which it is located. Glomerular filtration of urine is also compromised. A study from diabetic patients with T2D found that the contents of total GAGs, CS/DS, and HS in urine were significantly higher than those in healthy subjects [188]. Through quantitative analysis, it was found that 6-O-S and the 6-O-S/4-O-S ratio in the urine of diabetic patients with microalbuminuria were significantly increased compared with the healthy group [189]. In addition, CS/DS in erythrocytes was investigated in a diabetic rat model by Srikanth et al. They found a twofold increase in CS/DS content, mainly consisting of 4-O-S disaccharide units and a small number of non-sulfated disaccharides [190]. This suggests that the level of GAG in urine and blood or the ratio between them has the potential to be a diagnostic indicator of diabetes. However, the alteration of GAGs was mainly detected in advanced diabetic nephropathy, the elevation of GAG levels in urine/serum in the early stage of the disease is not very clear.
Diabetes begins with islet cell dysfunction, but persistent hyperglycemia causes a variety of complications. The ECM remodeling exhibited by various complications is also different. Therefore, finding a link between these GAG alterations is expected to provide useful information for the treatment of diabetes and its complications.

5. GAGs in Neurodegenerative Disease

Neurodegenerative disease is a type of disease caused by the degenerative loss of neurons in the brain and spinal cord. Its occurrence may be multifactorial (including genetics, oxidative stress, neuroinflammation, mitochondrial damage, and abnormal protein folding) [191], but its pathogenesis has not yet been determined [192,193]. Neurodegenerative diseases are classified according to pathological features, mainly including Alzheimer’s disease (AD) and Parkinson’s disease (PD), Huntington’s chorea, and amyotrophic lateral sclerosis. Protein misfolding and aggregation is one of the features of neurodegenerative diseases, such as β-amyloid (Aβ) and phosphorylated tau in AD, and α-synuclein (α-syn) aggregation and fibrogenesis in PD [194,195]. Neuronal cells are the main site of lesions in this kind of disease. Perineuronal nets (PNNs) are specialized extracellular matrices of neurons. They are mainly composed of CSPGs and HA, and they play an important part in the normal function of the central nervous system [196,197,198]. At the same time, changes in PNN composition also reflect a variety of neuropathological states [199].
The most abundant PG found in the nervous system is CSPG [200], and its sulfation pattern is thought to be directly linked to pathological development [196,201]. PGs in most neuronal PNNs mainly contain versicans, aggreicans, neuroicans, and brevicans [199]. It has been shown that CS-C (6-O-S) has the highest proportion in the early stages of brain development and is gradually replaced by CS-A (4-O-S) as the brain develops and matures [202]. This change illustrates the important role of CS-A in stabilizing PNNs and limiting neuronal plasticity [203,204]. However, this state shifts in AD. A recent study showed that CS quantification in prefrontal neocortical (middle frontal gyrus) samples from AD patients showed an increased CS-C and CS-E content and decreased non-sulfated CS disaccharides, but total GAG levels did not change [201]. In addition, a few reports have shown that CSPGs are also involved in the formation of amyloid precipitation [205,206]. Early studies certified that 4-O-S and 6-O-S CS are found in neurofibrillary tangles (NFTs) of AD patients, while only 4-O-S CS is found in senile plaques (SPs) [207]. In addition, in multiple sclerosis (MS), inflammatory cell infiltration, demyelination, and axonal injury result in sclerosing lesions in the white matter [208]. Studies have shown that CS/DSPGs are the main PG components of plaques [209]. It was found that CSPGs (versican, neurocan, and aggrecan) and DSPGs were mainly located at the edge of active plaques, while the content of CSPGs in the active center of MS plaques was significantly decreased, possibly due to the internalization of PGs in PNNs by their foam macrophages together with myelin [208]. Moreover, increased CSPG content at the plaque edge can inhibit oligodendrocyte precursor cells (OPCs) [210,211,212,213], which are essential for reconstructing myelin sheaths and protecting neurons. Further studies showed that the CS chain of CSPG exerts its inhibitory effect on OPCs mainly through 4-O-S and 6-O-S disaccharide units [214].
Although HS is less abundant in central neurons, it is the most studied PG in neurodegenerative diseases. HS has been demonstrated to be involved in abnormal protein accumulation within and around neurons, such as amyloid plaques and neurofibrillary tangles [197,206,215]. Extracellular Aβ plaques and intracellular NFTs are neurotoxic in AD, ultimately leading to neuronal loss [216]. In the brains of AD patients, PGs were more abundant in areas with amyloid plaques and neurofibrillary tangles. For example, relative to healthy individuals, the total PGs increased 1.6-fold in the AD hippocampus and 3.4-fold in the superior frontal gyrus (superior gyrus frontalis). Among them, HSPGs increased the most [217]. As described earlier, GAGs promote amyloid fibril formation, as determined by the chain type and sulfation pattern [175]. Staining of occipital neocortical and hippocampal tissue from AD patients revealed that fibrillar Aβ plaques and nonfibrillar Aβ plaques contained high levels of N-sulfated HS, while N-sulfation was very low in nonfibrillar Aβ plaques [218]. Lindahl et al. identified critical sites for the binding of heparin sulfate to Aβ fibrils containing 2-O-S IdoA and N-S from the human cerebral cortex, whereas the binding of Aβ monomers requires 6-O-S on GlcN residues [219]. The sequence of heparin oligosaccharides interacting with Aβ was determined using 2D NMR and molecular simulation docking techniques by Zhou et al. [220]. They found that the binding motif of HS is HexA–GlcNS–IdoA2S–GlcNS6S; IdoA and 6-O-S are required for binding [220]. This is consistent with previous findings. At the same time, they also identified the amino acid sequence of the Aβ-binding site as V12HHQKL17 using hydrogen–deuterium exchange mass spectrometry (HDX-MS) [220]. Coincidentally, the HS sequence profiles identified in the above studies are identical to FGF-2-binding sites [219]. Thus, the binding of HS to Aβ fibrils may competitively inhibit FGF2-mediated neuroprotection. In addition, GAGs can interact with tau protein, stabilize tau conformation, and promote its phosphorylation [221]. A recent study demonstrated that 3-O-S in HS enhances HS and tau binding and promotes tau transport across membranes [203]. Thus, tau is one of proteins that recognizes 3-O-S of HS. α-Synuclein (α-syn) is a major pathogenic protein in PD and a major component of LBs. HS interacts with α-syn and influences α-syn fibril conformation choice. Liu et al. found that HSPG (agrin) accelerated the formation of α-syn fibrils and induced α-syn protein β-sheets in an HS-dependent manner, enhancing the insolubility of α-syn [222]. A recent study determined the atomic structure of α-syn fibrils formed by heparin involvement and discovered a novel folding mode of α-syn, the “Z” fold [223]. These results indicate that sulfated GAGs play a crucial role in protein aggregation. Furthermore, Ishe et al. confirmed that, in neuronal cells, internalization of α-syn aggregates strongly depends on the cell surface HS and is associated with their total sulfation level [224]. These results indicate that HS acts on α-syn similarly to Aβ. In addition to HS playing an important role in PD progression, the presence of CS (4-O-S and 6-O-S) with different degrees of sulfation in LBs was reported by DeWitt et al. [225].
At present, the pathogenesis of most neurodegenerative diseases is still unclear. However, it is certain that GAGs are indeed involved in the development and progression of the disease. Therefore, an in-depth study of GAGs is an important aspect of mechanistic studies of neurodegenerative diseases.

6. GAGs in Virus Infection

Viruses are the main pathogens threatening human health, and they are characterized by species diversity, strong infectivity, diverse transmission routes, and easy variation. The worldwide spread of COVID-19 disease caused by severe acute respiratory syndrome-associated coronavirus 2 (SARS-CoV-2) in recent years is a living example. Furthermore, Ebola virus, HIV virus, SARS virus, and hantavirus have a high mortality. A common feature of viruses infecting humans is that they enter host cells by binding to cell-surface receptors. GAGs in the ECM have been found to play an important role in regulating immune defense and pathogenic mechanisms [226,227,228]. For instance, when SARS-CoV-2 infects host cells, HS acts as a cofactor involved in the interaction between the SARS-CoV-2 spike glycoprotein receptor-binding domain (RBD) and angiotensin-converting enzyme 2 (ACE2) [229,230,231]. In addition, dengue virus (DENV) [232,233], hepatitis C virus (HCV) [234], herpes simplex virus [235,236], human papillomavirus (HPV) [237], arboviruses [238], respiratory syncytial virus (RSV) [239], and monkeypox virus (MPXV) [240] have all been found to invade host cells by binding HS and/or CS/DS in the ECM through receptor proteins. Specific sulfation patterns of GAGs are critical for viral adsorption and invasion. For example, DENV-secreted NS1 protein accumulates on infected cell membranes and interacts with HS and CS-E on the cell surface, ultimately leading to selective vascular leak syndrome [232]. Kim et al. found that N-S, 2-O-S, and 6-O-S in HS and 6-O-S, 2-O-S, 3-O-S, and N-S in HP were critical for competitive binding to SARS-CoV-2 spike protein [241]. The amino-acid sequences of spike protein trimer-binding sites to GAGs are YRLFRKS, PRRARS, and SKPSKRS. Thus, HS is a potential competitive inhibitor against SARS-CoV-2 infection [241]. In parallel, Tiwari et al. confirmed that the presence of 3-O-S in HS contributes to the recognition and binding of SARS-CoV-2 spikes in vitro [242]. This idea was confirmed in another report, where 3-O-sulfotransferase 3B overexpression and glycocalyx sulfation degree were too low to promote SARS-CoV-2 infection under pathological conditions [243]. However, there are few reports on the alterations of GAGs in virus infection in vivo [244]. In brief, GAGs in cellular ECM execute critical roles in regulating viral adhesion and invasion.

7. GAGs in Other Diseases

GAGs/PGs, as the main component of ECM, are involved in the development and progression of almost all human diseases. Pulmonary diseases have a high morbidity and mortality, such as pulmonary fibrosis, asthma, and chronic obstructive pulmonary disease (COPD) [245,246]. In idiopathic pulmonary fibrosis, CS/DS, HA, and the CS/DS ratio increased significantly. CS/DS increased in 4-O-S, 6-O-S, and 2-O-S disaccharide units and decreased in non-sulfated disaccharides, resulting in significant increases in sulfated levels. Similarly, significant increases were observed in N-S, 2-O-S, and 6-O-S disaccharides of HS, particularly the UA2S–GlcNS6S unit [247]. In addition, significant increases in PG (versican, biglycan, and decorin) content at the lesion were observed in biopsy specimens from asthma cases [248]. In COPD, although HS increased significantly, its sulfation pattern was related to the COPD stage, while CS/DS did not change significantly. The 2-O-S and N-S of HS increased during the fourth phase of COPD, while 6-O-S did not change [249]. GAG expression is elevated in cystic fibrosis [250]. Moreover, abnormal sulfation of GAG can be found in bronchial epithelial cells of patients [251]. Kim et al. investigated CS expression in lupus erythematosus (LE) and dermatomyositis (DM) [252]. Expression of 4-O-S CS was found to be increased only in discoid lupus erythematosus (DLE) and DM, whereas 6-O-S CS was found to be significantly increased in dermal endothelium with DM [252].
Kidney stones are a common urinary disorder. Due to metabolic abnormalities, crystals such as uric acid and calcium oxalate (CaOx) accumulate in the renal pelvis or calyces of patients’ urine, resulting in stone formation. It has been found that the type and content of GAGs in urine may be closely related to the formation process of renal calculi [253,254,255]. Some earlier studies demonstrated that CS and HS inhibited the formation of kidney stones, while HA promoted stone formation and growth [256,257,258]. Dissayabutra et al. tested GAGs in urine from familial urolithiasis cases and found that total sulfated GAGs, CS, and HS contents in urine were all decreased, while HA content was increased, and the proportion of HS in total sulfated GAGs was increased [259]. Jappie et al. detected urine from healthy white and black South Africans. Blacks were found to have higher CS levels in urine than whites (kidney stones were significantly more prevalent in whites than blacks in South Africans), suggesting that higher CS levels may inhibit kidney stone formation [260]. These findings are consistent with previous conclusions. However, a recent study found that HA inhibited aggregation of CaOx crystals in artificial urine, but did not have any effect on the crystalline properties of CaOx in real urine. In addition, it has been pointed out that the regulation of nucleation and growth of crystals in urine is the result of the combined action of various GAGs [261].
In human diseases, inflammation is deemed to be the “source of all diseases”. The human body is an organic system, and there are tied connections between various diseases. A normal inflammatory response is beneficial for the human body, but chronic inflammation is a common pathological basis for various diseases and may be an important factor in the increased morbidity and mortality of most diseases. For example, many cancers develop in the presence of chronic inflammation; cancer metastasis is also akin to an inflammatory response, and even many inflammatory cells participate in and assist in the metastasis of cancer cells. Changes in the inflammatory response, from short to long term, can lead to the collapse of immune tolerance and lead to major alterations in the physiology of all tissues and organs, as well as normal cells. This increases the risk of noncommunicable diseases in humans. The ECM remodeling involved in inflammation, particularly the alterations and roles of HS, CS, and HA, were extensively discussed in several recent articles [262,263,264,265].

8. Prospects and Challenges

GAGs/PGs are the most complex biological macromolecules in the human body, and their role in cellular life processes is justifiable. Various GAGs function in vivo by interacting with proteins via ionic, hydrogen, and hydrophobic bonds [3,266]. The structural basis for these interactions is a specific sulfation sequence in the GAGs’ sugar chain. Because GAG biosynthesis is a non-template-driven process, their structures are stochastic and variable. GAGs also display structural alterations during pathology (the alterations of GAGs in different diseases are summarized in Table 1). Thus, complex sugar chains also have huge information density and extensive structural and functional heterogeneity. Over the last few decades, numerous studies were conducted on the structure and function of GAG sugar chains, as well as related regulatory enzymes, and their changes in time and space were identified, thus providing substantial theoretical support for the elucidation of disease mechanisms and the exploration of therapeutic targets.
However, there are still great challenges for the study of GAGs in human diseases. On the one hand, it has been debated whether the binding of GAG sequences to proteins is specific. However, there is increasing evidence that proteins are highly selective for GAG sequences. In addition to the examples listed here, classical binding of heparin pentasaccharides to antithrombin III has been demonstrated. However, most studies on GAG chains binding to proteins were performed in vitro; hence, whether they can truly reflect in vivo behavior needs further investigation. On the other hand, due to the high heterogeneity of the GAG molecular structure, the current structural analysis of GAG chains lags behind that of other biomacromolecules (proteins and DNA). Despite the rapid development of modern analytical techniques, determination of the fine structure of GAGs remains a challenging task [2]. This has caused great hindrance to in-depth studies of the structure–function relationship of endogenous GAGs in physiological or pathological conditions.
At present, GAGs have become another “life code” to be deciphered after nucleic acids and proteins, and the elucidation of the relationship between GAG structure and function is of great significance for the prevention of human diseases and the implementation of precision medicine.

Author Contributions

Writing, drawing, review and editing, Q.W.; editing, supervision, review, and funding acquisition, L.C. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by the Shandong Province Natural Science Foundation (Grant Number: ZR2022QB116) and the National Key Research and Development Program of China (Grant Number: 2021YFC2103104).

Institutional Review Board Statement

Not Applicable.

Informed Consent Statement

Not Applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

glycosaminoglycans (GAGs); proteoglycans (PGs); extracellular matrix (ECM); heparin (HP); heparan sulfate (HS); chondroitin sulfate (CS); dermatan sulfate (DS); keratan sulfate (KS); hyaluronan (HA); chondroitin sulfate proteoglycans (CSPGs); dermatan sulfate proteoglycans (DSPGs); heparan sulfate proteoglycans (HSPGs); basement membrane (BMs); Cardiovascular disease (CVD); smooth muscle cells (SMCs); arterial smooth muscle cells (ASMCs); elastin-binding protein (EBP); human macrophages (HMDM); low-molecular-weight hyaluronic acid (LMW-HA); high-molecular-weight HA (HMW-HA); tumor microenvironment (TME); neuroendocrine tumors (NETs); fibroblast growth factor (FGF); fibroblast growth factor receptor (FGFR); vascular endothelial growth factor (VEGF); HA-mediated mobility receptor (RHAMM); diabetes mellitus (diabetes, DM); type 1 diabetes mellitus (T1D); type 2 diabetes mellitus (T1D); unfolded protein response (UPR); islet amyloid polypeptide (IAPP); glomerular basement membranes (GBM); Alzheimer’s disease (AD); Parkinson’s disease (PD); β-amyloid (Aβ); α-synuclein (α-syn); perineuronal nets (PNNs); neurofibrillary tangles (NFTs); senile plaques (SPs); multiple sclerosis (MS); hydrogen–deuterium exchange mass spectrometry (HDX-MS); syndrome-associated coronavirus 2 (SARS-CoV-2); receptor-binding domain (RBD); angiotensin-converting enzyme 2 (ACE2); dengue virus (DENV); hepatitis C virus (HCV); human papillomavirus (HPV); respiratory syncytial virus (RSV); monkeypox virus (MPXV); chronic obstructive pulmonary disease (COPD); calcium oxalate (CaOx).

References

  1. Li, L.; Ly, M.; Linhardt, R.J. Proteoglycan Sequence. Mol. Biosyst. 2012, 8, 1613–1625. [Google Scholar] [CrossRef] [PubMed]
  2. Song, Y.; Zhang, F.; Linhardt, R.J. Analysis of the Glycosaminoglycan Chains of Proteoglycans. J. Histochem. Cytochem. 2021, 69, 121–135. [Google Scholar] [CrossRef] [PubMed]
  3. Shi, D.; Sheng, A.; Chi, L. Glycosaminoglycan-Protein Interactions and Their Roles in Human Disease. Front. Mol. Biosci. 2021, 8, 639666. [Google Scholar] [CrossRef] [PubMed]
  4. Buijsers, B.; Yanginlar, C.; Maciej-Hulme, M.L.; de Mast, Q.; van der Vlag, J. Beneficial Non-Anticoagulant Mechanisms Underlying Heparin Treatment of Covid-19 Patients. EBioMedicine 2020, 59, 102969. [Google Scholar] [CrossRef] [PubMed]
  5. De Pasquale, V.; Pavone, L.M. Heparan Sulfate Proteoglycans: The Sweet Side of Development Turns Sour in Mucopolysaccharidoses. Biochim. Biophys. Acta Mol. Basis Dis. 2019, 1865, 165539. [Google Scholar] [CrossRef] [PubMed]
  6. Aquino, R.S.; Lee, E.S.; Park, P.W. Diverse Functions of Glycosaminoglycans in Infectious Diseases. In Progress in Molecular Biology and Translational Science; Zhang, L., Ed.; Academic Press: New York, NY, USA, 2010; Volume 93, pp. 373–394. [Google Scholar]
  7. Sugahara, K.; Mikami, T.; Uyama, T.; Mizuguchi, S.; Nomura, K.; Kitagawa, H. Recent Advances in the Structural Biology of Chondroitin Sulfate and Dermatan Sulfate. Curr. Opin. Struct. Biol. 2003, 13, 612–620. [Google Scholar] [CrossRef]
  8. Malavaki, C.; Mizumoto, S.; Karamanos, N.; Sugahara, K. Recent Advances in the Structural Study of Functional Chondroitin Sulfate and Dermatan Sulfate in Health and Disease. Connect. Tissue Res. 2008, 49, 133–139. [Google Scholar] [CrossRef]
  9. Tang, F.; Lord, M.S.; Stallcup, W.B.; Whitelock, J.M. Cell Surface Chondroitin Sulphate Proteoglycan 4 (Cspg4) Binds to the Basement Membrane Heparan Sulphate Proteoglycan, Perlecan, and Is Involved in Cell Adhesion. J. Biochem. 2018, 163, 399–412. [Google Scholar] [CrossRef] [Green Version]
  10. Avram, S.; Shaposhnikov, S.; Buiu, C.; Mernea, M. Chondroitin Sulfate Proteoglycans: Structure-Function Relationship with Implication in Neural Development and Brain Disorders. Biomed Res. Int. 2014. [Google Scholar] [CrossRef] [Green Version]
  11. Sugahara, K.; Mikami, T. Chondroitin/Dermatan Sulfate in the Central Nervous System. Curr. Opin. Struct. Biol. 2007, 17, 536–545. [Google Scholar] [CrossRef]
  12. Du Souich, P.; Garcia, A.G.; Verges, J.; Montell, E. Immunomodulatory and Anti-Inflammatory Efects of Chondroitin Sulphate. J. Cell. Mol. Med. 2009, 13, 1451–1463. [Google Scholar] [CrossRef] [PubMed]
  13. Sodhi, H.; Panitch, A. Glycosaminoglycans in Tissue Engineering: A Review. Biomolecules 2020, 11, 29. [Google Scholar] [CrossRef] [PubMed]
  14. Caterson, B.; Melrose, J. Keratan Sulfate, a Complex Glycosaminoglycan with Unique Functional Capability. Glycobiology 2018, 28, 182–206. [Google Scholar] [CrossRef] [Green Version]
  15. Iozzo, R.V.; Schaefer, L. Proteoglycan Form and Function: A Comprehensive Nomenclature of Proteoglycans. Matrix Biol. 2015, 42, 11–55. [Google Scholar] [CrossRef]
  16. Necas, J.; Bartosikova, L.; Brauner, P.; Kolar, J. Hyaluronic Acid (Hyaluronan): A Review. Vet. Med. 2008, 53, 397–411. [Google Scholar] [CrossRef] [Green Version]
  17. Gesslbauer, B.; Rek, A.; Falsone, F.; Rajkovic, E.; Kungl, A.J. Proteoglycanomics: Tools to Unravel the Biological Function of Glycosaminoglycans. Proteomics 2007, 7, 2870–2880. [Google Scholar] [CrossRef]
  18. Bulow, H.E.; Hobert, O. The Molecular Diversity of Glycosaminoglycans Shapes Animal Development. Annu. Rev. Cell Dev. Biol. 2006, 22, 375–407. [Google Scholar] [CrossRef]
  19. Esko, J.D.; Selleck, S.B. Order out of Chaos: Assembly of Ligand Binding Sites in Heparan Sulfate. Annu. Rev. Biochem. 2002, 71, 435–471. [Google Scholar] [CrossRef] [PubMed]
  20. Capila, I.; Linhardt, R.J. Heparin-Protein Interactions. Angew. Chem. Int. Ed. 2002, 41, 390–412. [Google Scholar] [CrossRef]
  21. Couchman, J.R. Transmembrane Signaling Proteoglycans. Annu. Rev. Cell Dev. Biol. 2010, 26, 89–114. [Google Scholar] [CrossRef]
  22. Nakato, H.; Kimata, K. Heparan Sulfate Fine Structure and Specificity of Proteoglycan Functions. Biochim. Biophys. Acta 2002, 1573, 312–318. [Google Scholar] [CrossRef]
  23. Gallagher, J.T. Multiprotein Signalling Complexes: Regional Assembly on Heparan Sulphate. Biochem. Soc. Trans. 2006, 34, 438–441. [Google Scholar] [CrossRef] [PubMed]
  24. Hiroko, H.; Goichiro, M.; Ken, N.; Asato, K.; Yoichi, M.; Marion, K.G.; Osami, H.; Masayuki, T.; Koji, K. Biosynthesis of Heparan Sulphate with Diverse Structures and Functions Two Alternatively Spliced Forms of Human Heparan Sulphate 6-O-Sulphotransferase-2 Having Different Expression Patterns and Properties. Biochem. J. 2003, 371, 131–142. [Google Scholar]
  25. Dagenais, G.R.; Leong, D.P.; Rangarajan, S.; Lanas, F.; Lopez-Jaramillo, P.; Gupta, R.; Diaz, R.; Avezum, A.; Oliveira, G.B.F.; Wielgosz, A.; et al. Variations in Common Diseases, Hospital Admissions, and Deaths in Middle-Aged Adults in 21 Countries from Five Continents (Pure): A Prospective Cohort Study. Lancet 2020, 395, 785–794. [Google Scholar] [CrossRef]
  26. La Sala, L.; Prattichizzo, F.; Ceriello, A. The Link between Diabetes and Atherosclerosis. Eur. J. Prev. Cardiol. 2019, 26, 15–24. [Google Scholar] [CrossRef]
  27. Ye, J.; Li, L.; Wang, M.; Ma, Q.; Tian, Y.; Zhang, Q.; Liu, J.; Li, B.; Zhang, B.; Liu, H.; et al. Diabetes Mellitus Promotes the Development of Atherosclerosis: The Role of Nlrp3. Front. Immunol. 2022, 13, 900254. [Google Scholar] [CrossRef]
  28. Pennathur, S.; Heinecke, J.W. Oxidative Stress and Endothelial Dysfunction in Vascular Disease. Curr. Diabetes Rep. 2007, 7, 257–264. [Google Scholar] [CrossRef]
  29. Luscher, T.F. Inflammation: The New Cardiovascular Risk Factor. Eur. Heart J. 2018, 39, 3483–3487. [Google Scholar] [CrossRef]
  30. Lancellotti, P.; Marechal, P.; Donis, N.; Oury, C. Inflammation, Cardiovascular Disease, and Cancer: A Common Link with Far-Reaching Implications. Eur. Heart J. 2019, 40, 3910–3912. [Google Scholar] [CrossRef] [Green Version]
  31. Valdivielso, J.M.; Rodriguez-Puyol, D.; Pascual, J.; Barrios, C.; Bermudez-Lopez, M.; Sanchez-Nino, M.D.; Perez-Fernandez, M.; Ortiz, A. Atherosclerosis in Chronic Kidney Disease: More, Less, or Just Different? Arterioscler. Thromb. Vasc. Biol. 2019, 39, 1938–1966. [Google Scholar] [CrossRef]
  32. Choi, J.; Haan, J.D.; Sharma, A. Animal Models of Diabetes-Associated Vascular Diseases: An Update on Available Models and Experimental Analysis. Br. J. Pharmacol. 2022, 179, 748–769. [Google Scholar] [CrossRef] [PubMed]
  33. Drinkwater, J.J.; Davis, T.M.E.; Davis, W.A. The Relationship between Carotiddisease and Retinopathy in Diabetes: A Systematic Review. Cardiovasc. Diabetol. 2020, 19, 54. [Google Scholar] [CrossRef] [PubMed]
  34. Kim, M.J.; Jung, S.K. Nutraceuticals for Prevention of Atherosclerosis: Targeting Monocyte Infiltration to the Vascular Endothelium. J. Food Biochem. 2020, 44, e13200. [Google Scholar] [CrossRef] [PubMed]
  35. Masola, V.; Zaza, G.; Arduini, A.; Onisto, M.; Gambaro, G. Endothelial Glycocalyx as a Regulator of Fibrotic Processes. Int. J. Mol. Sci. 2021, 22, 2996. [Google Scholar] [CrossRef]
  36. Ghadie, N.M.; St-Pierre, J.P.; Labrosse, M.R. The Contribution of Glycosaminoglycans/Proteoglycans to Aortic Mechanics in Health and Disease: A Critical Review. IEEE Trans. Biomed. Eng. 2021, 68, 3491–3500. [Google Scholar] [CrossRef]
  37. Naso, M.F.; Zimmermann, D.R.; Iozzo, R.V. Characterization of the Complete Genomic Structure of the Human Versican Gene and Functional Analysis of Its Promoter. J. Biol. Chem. 1994, 269, 32999–33008. [Google Scholar] [CrossRef]
  38. Zimmermann, D.R.; Ruoslahti, E. Multiple Domains of the Large Fibroblast Proteoglycan, Versican. EMBO J. 1989, 8, 2975–2981. [Google Scholar] [CrossRef]
  39. Jackson, R.L.; Busch, S.J.; Cardin, A.D. Glycosaminoglycans: Molecular Properties, Protein Interactions, and Role in Physiological Processes. Physiol. Rev. 1991, 71, 481–539. [Google Scholar] [CrossRef]
  40. Hultgardh-Nilsson, A.; Boren, J.; Chakravarti, S. The Small Leucine-Rich Repeat Proteoglycans in Tissue Repair and Atherosclerosis. J. Intern. Med. 2015, 278, 447–461. [Google Scholar] [CrossRef] [Green Version]
  41. Boren, J.; Olin, K.; Lee, I.; Chait, A.; Wight, T.N.; Innerarity, T.L. Identification of the Principal Proteoglycan-Binding Site in Ldl. A Single-Point Mutation in Apo-B100 Severely Affects Proteoglycan Interaction without Affecting Ldl Receptor Binding. J. Clin. Investig. 1998, 101, 2658–2664. [Google Scholar] [CrossRef] [Green Version]
  42. Olin, K.L.; Potter-Perigo, S.; Barrett, P.H.; Wight, T.N.; Chait, A. Lipoprotein Lipase Enhances the Binding of Native and Oxidized Low Density Lipoproteins to Versican and Biglycan Synthesized by Cultured Arterial Smooth Muscle Cells. J. Biol. Chem. 1999, 274, 34629–34636. [Google Scholar] [CrossRef] [PubMed]
  43. Pentikäinen, M.O.; Oörni, K.; Lassila, R.; Kovanen, P.T. The Proteoglycan Decorin Links Low Density Lipoproteins with Collagen Type I. J. Biol. Chem. 1997, 272, 7633–7638. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Wight, T.N.; Merrilees, M.J. Proteoglycans in Atherosclerosis and Restenosis: Key Roles for Versican. Circ. Res. 2004, 94, 1158–1167. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Vijayagopal, P.; Figueroa, J.E.; Fontenot, J.D.; Glancy, D.L. Isolation and Characterization of a Proteoglycan Variant from Human Aorta Exhibiting a Marked Affinity for Low Density Lipoprotein and Demonstration of Its Enhanced Expression in Atherosclerotic Plaques. Atherosclerosis 1996, 127, 195–203. [Google Scholar] [CrossRef]
  46. Hinek, A.; Mecham, R.P.; Keeley, F.; Rabinovitch, M. Impaired Elastin Fiber Assembly Related to Reduced 67-KD Elastin-Binding Protein in Fetal Lamb Ductus Arteriosus and in Cultured Aortic Smooth Muscle Cells Treated with Chondroitin Sulfate. J. Clin. Investig. 1991, 88, 2083–2094. [Google Scholar] [CrossRef]
  47. Wight, T.N. A Role for Proteoglycans in Vascular Disease. Matrix Biol. 2018, 71–72, 396–420. [Google Scholar] [CrossRef]
  48. Sambandam, T.; Baker, J.R.; Christner, J.E.; Eborg, S.L. Specificity of the Low Density Lipoproteins-Glycosaminoglycan Interaction. Arterioscler. Thromb. 1991, 11, 561–568. [Google Scholar] [CrossRef] [Green Version]
  49. Cardoso, L.E.; Mourão, P.A. Glycosaminoglycan Fractions from Human Arteries Presenting Diverse Susceptibilities to Atherosclerosis Have Different Binding Affinities to Plasma Ldl. Arterioscler. Thromb. 1994, 14, 115–124. [Google Scholar] [CrossRef] [Green Version]
  50. Theocharis, A.D.; Theocharis, D.A.; Luca, G.D.; Hjerpe, A.; Karamanos, N.K. Compositional and Structural Aterations of Chondroitin and Dermatan Sulfates During the Progression of Atherosclerosis and Aneurysmal Dilatation of the Human Abdominal Aorta. Biochimie 2002, 84, 667–674. [Google Scholar] [CrossRef]
  51. Little, P.J.; Tannock, L.; Olin, K.L.; Chait, A.; Wight, T.N. Proteoglycans Synthesized by Arterial Smooth Muscle Cells in the Presence of Transforming Growth Factor-Beta1 Exhibit Increased Binding to Ldls. Arterioscler. Thromb. Vasc. Biol. 2002, 22, 55–60. [Google Scholar] [CrossRef] [Green Version]
  52. Pillarisetti, S. Lipoprotein Modulation of Subendothelial Heparan Sulfate Proteoglycans (Perlecan) and Atherogenicity. Trends Cardiovasc. Med. 2000, 10, 60–65. [Google Scholar] [CrossRef]
  53. Tran, P.K.; Agardh, H.E.; Tran-Lundmark, K.; Ekstrand, J.; Roy, J.; Henderson, B.; Gabrielsen, A.; Hansson, G.K.; Swedenborg, J.; Paulsson-Berne, G.; et al. Reduced Perlecan Expression and Accumulation in Human Carotid Atherosclerotic Lesions. Atherosclerosis 2007, 190, 264–270. [Google Scholar] [CrossRef] [PubMed]
  54. Nakamura, M.; Imaizumi, K.; Shigemi, U.; Nakashima, Y.; Kikuchi, Y. Relationship of Sulfated Glycosaminoglycans and Cholesterol Content in Normal and Atherosclerotic Human Aorta. Stroke 1976, 7, 594–598. [Google Scholar] [CrossRef] [Green Version]
  55. Völker, W.; Schmidt, A.; Oortmann, W.; Broszey, T.; Faber, V.; Buddecke, E. Mapping of Proteoglycans in Atherosclerotic Lesions. Eur. Heart J. 1990, 11, 29–40. [Google Scholar] [CrossRef] [PubMed]
  56. Pillarisetti, S.; Paka, L.; Obunike, J.C.; Berglund, L.; Goldberg, I.J. Subendothelial Retention of Lipoprotein (a). Evidence That Teduced Heparan Sulfate Promotes Lipoprotein Binding to Subendothelial Matrix. J. Clin. Investig. 1997, 100, 867–874. [Google Scholar] [CrossRef] [PubMed]
  57. Duan, W.; Paka, L.; Pillarisetti, S. Distinct Effects of Glucose and Glucosamine on Vascular Endothelial and Smooth Muscle Cells: Evidence for a Protective Role for Glucosamine in Atherosclerosis. Cardiovasc. Diabetol. 2005, 4, 16. [Google Scholar] [CrossRef] [Green Version]
  58. Asplund, A.; Ostergren-Lunden, G.; Camejo, G.; Stillemark-Billton, P.; Bondjers, G. Hypoxia Increases Macrophage Motility, Possibly by Decreasing the Heparan Sulfate Proteoglycan Biosynthesis. J. Leukoc. Biol. 2009, 86, 381–388. [Google Scholar] [CrossRef] [Green Version]
  59. Fischer, J.W. Role of Hyaluronan in Atherosclerosis: Current Knowledge and Open Questions. Matrix Biol. 2019, 78–79, 324–336. [Google Scholar] [CrossRef]
  60. Karangelis, D.E.; Kanakis, I.; Asimakopoulou, A.P.; Karousou, E.; Passi, A.; Theocharis, A.D.; Triposkiadis, F.; Tsilimingas, N.B.; Karamanos, N. Glycosaminoglycans as Key Molecules in Atherosclerosis: The Role of Versican and Hyaluronan. Curr. Med. Chem. 2010, 17, 4018–4026. [Google Scholar] [CrossRef]
  61. Vigetti, D.; Viola, M.; Karousou, E.; Rizzi, M.; Moretto, P.; Genasetti, A.; Clerici, M.; Hascall, V.C.; De Luca, G.; Passi, A. Hyaluronan-Cd44-Erk1/2 Regulate Human Aortic Smooth Muscle Cell Motility During Aging. J. Biol. Chem. 2008, 283, 4448–4458. [Google Scholar] [CrossRef] [Green Version]
  62. 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. [Google Scholar] [CrossRef] [PubMed]
  63. Noble, P.W. Hyaluronan and Its Catabolic Products in Tissue Injury and Repair. Matrix Biol. 2002, 21, 25–29. [Google Scholar] [CrossRef]
  64. Grandoch, M.; Bollyky, P.L.; Fischer, J.W. Hyaluronan: A Master Switch between Vascular Homeostasis and Inflammation. Circ. Res. 2018, 122, 1341–1343. [Google Scholar] [CrossRef] [PubMed]
  65. Tabata, T.; Mine, S.; Okada, Y.; Tanaka, Y. Low Molecular Weight Hyaluronan Increases the Uptaking of Oxidized Ldl into Monocytes. Endocr. J. 2007, 54, 685–693. [Google Scholar] [CrossRef] [Green Version]
  66. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: Globocan Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
  67. Bergers, G.; Fendt, S.M. The Metabolism of Cancer Cells During Metastasis. Nat. Rev. Cancer 2021, 21, 162–180. [Google Scholar] [CrossRef]
  68. Lemaire, J.; Larrue, R.; Perrais, M.; Cauffiez, C.; Pottier, N. Fundamental Aspects of Oncogenesis. Bull. Cancer 2020, 107, 1148–1160. [Google Scholar]
  69. Poltavets, V.; Kochetkova, M.; Pitson, S.M.; Samuel, M.S. The Role of the Extracellular Matrix and Its Molecular and Cellular Regulators in Cancer Cell Plasticity. Front. Oncol. 2018, 8, 431. [Google Scholar] [CrossRef] [Green Version]
  70. Xiao, Y.; Yu, D. Tumor Microenvironment as a Therapeutic Target in Cancer. Pharmacol. Ther. 2021, 221, 107753. [Google Scholar] [CrossRef]
  71. Arneth, B. Tumor Microenvironment. Medicina 2020, 56, 15. [Google Scholar] [CrossRef] [Green Version]
  72. Walker, C.; Mojares, E.; Del Rio Hernandez, A. Role of Extracellular Matrix in Development and Cancer Progression. Int. J. Mol. Sci. 2018, 19, 3028. [Google Scholar] [CrossRef] [Green Version]
  73. Wei, J.; Hu, M.; Huang, K.; Lin, S.; Du, H. Roles of Proteoglycans and Glycosaminoglycans in Cancer Development and Progression. Int. J. Mol. Sci. 2020, 21, 5983. [Google Scholar] [CrossRef]
  74. Afratis, N.; Gialeli, C.; Nikitovic, D.; Tsegenidis, T.; Karousou, E.; Theocharis, A.D.; Pavao, M.S.; Tzanakakis, G.N.; Karamanos, N.K. Glycosaminoglycans: Key Players in Cancer Cell Biology and Treatment. FEBS J. 2012, 279, 1177–1197. [Google Scholar] [CrossRef]
  75. Berdiaki, A.; Neagu, M.; Giatagana, E.M.; Kuskov, A.; Tsatsakis, A.M.; Tzanakakis, G.N.; Nikitovic, D. Glycosaminoglycans: Carriers and Targets for Tailored Anti-Cancer Therapy. Biomolecules 2021, 11, 395. [Google Scholar] [CrossRef]
  76. Nagarajan, A.; Malvi, P.; Wajapeyee, N. Heparan Sulfate and Heparan Sulfate Proteoglycans in Cancer Initiation and Progression. Front. Endocrinol. 2018, 9, 483. [Google Scholar] [CrossRef] [Green Version]
  77. Soares da Costa, D.; Reis, R.L.; Pashkuleva, I. Sulfation of Glycosaminoglycans and Its Implications in Human Health and Disorders. Annu. Rev. Biomed. Eng. 2017, 19, 1–26. [Google Scholar] [CrossRef] [Green Version]
  78. Jansson, M.; Billing, O.; Herdenberg, C.; Lundin, C.; Tolockiene, E.; Nazemroaya, A.; Sund, M. Expression and Circulating Levels of Perlecan in Breast Cancer—Implications for Oestrogen Dependent Stromal Remodeling. J. Mammary Gland Biol. Neoplasia 2020, 25, 69–77. [Google Scholar] [CrossRef] [Green Version]
  79. Datta, S.; Pierce, M.; Datta, M.W. Perlecan Signaling: Helping Hedgehog Stimulate Prostate Cancer Growth. Int. J. Biochem. Cell Biol. 2006, 38, 1855–1861. [Google Scholar] [CrossRef]
  80. Garcia-Suarez, O.; Garcia, B.; Fernandez-Vega, I.; Astudillo, A.; Quiros, L.M. Neuroendocrine Tumors Show Altered Expression of Chondroitin Sulfate, Glypican 1, Glypican 5, and Syndecan 2 Depending on Their Differentiation Grade. Front. Oncol. 2014, 4, 15. [Google Scholar] [CrossRef] [Green Version]
  81. Fernández-Vega, I.; García, O.; Crespo, A.; Castañón, S.; Menéndez, P.; Astudillo, A.; Quirós, L.M. Specific Genes Involved in Synthesis and Editing of Heparan Sulfate Proteoglycans Show Altered Expression Patterns in Breast Cancer. BMC Cancer 2013, 17, 13–24. [Google Scholar] [CrossRef] [Green Version]
  82. Cooney, C.A.; Jousheghany, F.; Yao-Borengasser, A.Y.; Phanavanh, B.; Gomes, T.; Kieber-Emmons, A.M.; Siegel, E.R.; Suva, L.J.; Ferrone, S.; Kieber-Emmons, T.; et al. Chondroitin Sulfates Play a Major Role in Breast Cancer Metastasis: A Role for Cspg4 and Chst11 Gene Expression in Forming Surface P-Selectin Ligands in Aggressive Breast Cancer Cells. Breast Cancer Res. 2011, 13, R58. [Google Scholar] [CrossRef] [Green Version]
  83. Vynios, D.H.; Theocharis, D.A.; Papageorgakopoulou, N.; Papadas, T.A.; Mastronikolis, N.S.; Goumas, P.D.; Stylianou, M.; Skandalis, S.S. Biochemical Changes of Extracellular Proteoglycans in Squamous Cell Laryngeal Carcinoma. Connect. Tissue Res. 2008, 49, 239–243. [Google Scholar] [CrossRef]
  84. Backen, A.C.; Cole, C.L.; Lau, S.C.; Clamp, A.R.; McVey, R.; Gallagher, J.T.; Jayson, G.C. Heparan Sulphate Synthetic and Editing Enzymes in Ovarian Cancer. Br. J. Cancer 2007, 96, 1544–1548. [Google Scholar] [CrossRef] [Green Version]
  85. Vallen, M.J.; Massuger, L.F.; ten Dam, G.B.; Bulten, J.; van Kuppevelt, T.H. Highly Sulfated Chondroitin Sulfates, a Novel Class of Prognostic Biomarkers in Ovarian Cancer Tissue. Gynecol. Oncol. 2012, 127, 202–209. [Google Scholar] [CrossRef]
  86. Kalathas, D.; Theocharis, D.A.; Bounias, D.; Kyriakopoulou, D.; Papageorgakopoulou, N.; Stavropoulos, M.S.; Vynios, D.H. Alterations of Glycosaminoglycan Disaccharide Content and Composition in Colorectal Cancer: Structural and Expressional Studies. Oncol. Rep. 2009, 22, 369–375. [Google Scholar]
  87. Weyers, A.; Yang, B.; Yoon, D.S.; Park, J.H.; Zhang, F.; Lee, K.B.; Linhardt, R.J. A Structural Analysis of Glycosaminoglycans from Lethal and Nonlethal Breast Cancer Tissues: Toward a Novel Class of Theragnostics for Personalized Medicine in Oncology? OMICS 2012, 16, 79–89. [Google Scholar] [CrossRef] [Green Version]
  88. Faria-Ramos, I.; Poças, J.; Marques, C.; Santos-Antunes, J.; Macedo, G.; Reis, C.A.; Magalhães, A. Heparan Sulfate Glycosaminoglycans: (Un)Expected Allies in Cancer Clinical Management. Biomolecules 2021, 11, 136. [Google Scholar] [CrossRef]
  89. El Masri, R.; Seffouh, A.; Lortat-Jacob, H.; Vives, R.R. The “in and out” of Glucosamine 6-O-Sulfation: The 6th Sense of Heparan Sulfate. Glycoconj. J. 2017, 34, 285–298. [Google Scholar] [CrossRef]
  90. Wang, Y.; Liu, D.; Zhang, T.; Xia, L. Fgf/Fgfr Signaling in Hepatocellular Carcinoma: From Carcinogenesis to Recent Therapeutic Intervention. Cancers 2021, 13, 1360. [Google Scholar] [CrossRef]
  91. Jomrich, G.; Hudec, X.; Harpain, F.; Winkler, D.; Timelthaler, G.; Mohr, T.; Marian, B.; Schoppmann, S.F. Expression of Fgf8, Fgf18, and Fgfr4 in Gastroesophageal Adenocarcinomas. Cells 2019, 8, 1092. [Google Scholar] [CrossRef] [Green Version]
  92. Stehbens, S.J.; Ju, R.J.; Adams, M.N.; Perry, S.R.; Haass, N.K.; Bryant, D.M.; Pollock, P.M. Fgfr2-Activating Mutations Disrupt Cell Polarity to Potentiate Migration and Invasion in Endometrial Cancer Cell Models. J. Cell Sci. 2018, 131, jcs213678. [Google Scholar] [CrossRef] [Green Version]
  93. Stauber, D.J.; DiGabriele, A.D.; Hendrickson, W.A. Structural Interactions of Fibroblast Growth Factor Receptor with Its Ligands. Proc. Natl. Acad. Sci. USA 2000, 97, 49–54. [Google Scholar] [CrossRef] [Green Version]
  94. Elgundi, Z.; Papanicolaou, M.; Major, G.; Cox, T.R.; Melrose, J.; Whitelock, J.M.; Farrugia, B.L. Cancer Metastasis: The Role of the Extracellular Matrix and the Heparan Sulfate Proteoglycan Perlecan. Front. Oncol. 2019, 9, 1482. [Google Scholar] [CrossRef]
  95. Lundin, L.; Larsson, H.; Kreuger, J.; Kanda, S.; Lindahl, U.; Salmivirta, M.; Claesson-Welsh, L. Selectively Desulfated Heparin Inhibits Fibroblast Growth Factor-Induced Mitogenicity and Angiogenesis. J. Biol. Chem. 2000, 275, 24653–24660. [Google Scholar] [CrossRef] [Green Version]
  96. Pye, D.A.; Vivès, R.R.; Hyde, P.; Gallagher, J.T. Regulation of Fgf-1 Mitogenic Activity by Heparan Sulfate Oligosaccharides Is Dependent on Specific Structural Features: Differential Requirements for the Modulation of Fgf-1 and Fgf-2. Glycobiology 2000, 10, 1183–1192. [Google Scholar] [CrossRef]
  97. Kreuger, J.; Salmivirta, M.; Sturiale, L.; Gimenez-Gallego, G.; Lindahl, U. Sequence Analysis of Heparan Sulfate Epitopes with Graded Affinities for Fibroblast Growth Factors 1 and 2. J. Biol. Chem. 2001, 276, 30744–30752. [Google Scholar] [CrossRef] [Green Version]
  98. Schultz, V.; Suflita, M.; Liu, X.; Zhang, X.; Yu, Y.; Li, L.; Green, D.E.; Xu, Y.; Zhang, F.; DeAngelis, P.L.; et al. Heparan Sulfate Domains Required for Fibroblast Growth Factor 1 and 2 Signaling through Fibroblast Growth Factor Receptor 1c. J. Biol. Chem. 2017, 292, 2495–2509. [Google Scholar] [CrossRef] [Green Version]
  99. Brunetti, J.; Depau, L.; Falciani, C.; Gentile, M.; Mandarini, E.; Riolo, G.; Lupetti, P.; Pini, A.; Bracci, L. Insights into the Role of Sulfated Glycans in Cancer Cell Adhesion and Migration through Use of Branched Peptide Probe. Sci. Rep. 2016, 6, 27174. [Google Scholar] [CrossRef] [Green Version]
  100. Falciani, C.; Brunetti, J.; Lelli, B.; Ravenni, N.; Lozzi, L.; Depau, L.; Scali, S.; Bernini, A.; Pini, A.; Bracci, L. Cancer Selectivity of Tetrabranched Neurotensin Peptides Is Generated by Simultaneous Binding to Sulfated Glycosaminoglycans and Protein Receptors. J. Med. Chem. 2013, 56, 5009–5018. [Google Scholar] [CrossRef]
  101. Falciani, C.; Fabbrini, M.; Pini, A.; Lozzi, L.; Lelli, B.; Pileri, S.; Brunetti, J.; Bindi, S.; Scali, S.; Bracci, L. Synthesis and Biological Activity of Stable Branched Neurotensin Peptides for Tumortargeting. Mol. Cancer Ther. 2007, 6, 2441–2448. [Google Scholar] [CrossRef] [Green Version]
  102. Falciani, C.; Brunetti, J.; Pagliuca, C.; Menichetti, S.; Vitellozzi, L.; Lelli, B.; Pini, A.; Bracci, L. Design and in Vitro Evaluation of Branched Peptide Conjugates: Turning Nonspecific Cytotoxic Drugs into Tumor-Selective Agents. ChemMedChem 2010, 5, 567–574. [Google Scholar] [CrossRef] [PubMed]
  103. Brunetti, J.; Falciani, C.; Lelli, B.; Minervini, A.; Ravenni, N.; Depau, L.; Siena, G.; Tenori, E.; Menichetti, S.; Pini, A.; et al. Neurotensin Branched Peptide as a Tumor-Targeting Agent for Human Bladder Cancer. Biomed Res. Int. 2015. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Brunetti, J.; Pillozzi, S.; Falciani, C.; Depau, L.; Tenori, E.; Scali, S.; Lozzi, L.; Pini, A.; Arcangeli, A.; Menichetti, S.; et al. Tumor-Selective Peptide-Carrier Delivery of Paclitaxel Increases in Vivo Activity of the Drug. Sci. Rep. 2015, 5, 17736. [Google Scholar] [CrossRef] [Green Version]
  105. Brunetti, J.; Riolo, G.; Gentile, M.; Bernini, A.; Paccagnini, E.; Falciani, C.; Lozzi, L.; Scali, S.; Depau, L.; Pini, A.; et al. Near-Infrared Quantum Dots Labelled with a Tumor Selective Tetrabranched Peptide for in Vivo Imaging. J. Nanobiotechnol. 2018, 16, 21. [Google Scholar] [CrossRef] [PubMed]
  106. Bracci, L.; Mandarini, E.; Brunetti, J.; Depau, L.; Pini, A.; Terzuoli, L.; Scali, S.; Falciani, C. The Gag-Specific Branched Peptide Nt4 Reduces Angiogenesis and Invasiveness of Tumor Cells. PLoS ONE 2018, 13, e0194744. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Brunetti, J.; Riolo, G.; Depau, L.; Mandarini, E.; Bernini, A.; Karousou, E.; Passi, A.; Pini, A.; Bracci, L.; Falciani, C. Unraveling Heparan Sulfate Proteoglycan Binding Motif for Cancer Cell Selectivity. Front. Oncol. 2019, 9, 843. [Google Scholar] [CrossRef]
  108. Liu, D.F.; Shriver, Z.; Venkataraman, G.; Shabrawi, Y.E.; Sasisekharan, R. Tumor Cell Surface Heparan Sulfate as Cryptic Promoters or Inhibitors of Tumor Growth and Metastasis. PNAS 2002, 99, 568–573. [Google Scholar] [CrossRef] [Green Version]
  109. Mikami, T.; Kitagawa, H. Biosynthesis and Function of Chondroitin Sulfate. Biochim. Biophys. Acta 2013, 1830, 4719–4733. [Google Scholar] [CrossRef]
  110. Batista, L.T.; Matos, L.L.; Machado, L.R.; Suarez, E.R.; Theodoro, T.R.; Martins, J.R.; Nader, H.B.; Pompeo, A.C.; Pinhal, M.A. Heparanase Expression and Glycosaminoglycans Profile in Renal Cell Carcinoma. Int. J. Urol. 2012, 19, 1036–1040. [Google Scholar] [CrossRef]
  111. Brauchle, E.; Kasper, J.; Daum, R.; Schierbaum, N.; Falch, C.; Kirschniak, A.; Schaffer, T.E.; Schenke-Layland, K. Biomechanical and Biomolecular Characterization of Extracellular Matrix Structures in Human Colon Carcinomas. Matrix Biol. 2018, 68–69, 180–193. [Google Scholar] [CrossRef]
  112. Rangel, M.P.; de Sa, V.K.; Prieto, T.; Martins, J.R.M.; Olivieri, E.R.; Carraro, D.; Takagaki, T.; Capelozzi, V.L. Biomolecular Analysis of Matrix Proteoglycans as Biomarkers in Non Small Cell Lung Cancer. Glycoconj. J. 2018, 35, 233–242. [Google Scholar] [CrossRef]
  113. Marolla, A.P.; Waisberg, J.; Saba, G.T.; Waisberg, D.R.; Margeotto, F.B.; Pinhal, M.A. Glycomics Expression Analysis of Sulfated Glycosaminoglycans of Human Colorectal Cancer Tissues and Non-Neoplastic Mucosa by Electrospray Ionization Mass Spectrometry. Einstein 2015, 13, 510–517. [Google Scholar] [CrossRef] [Green Version]
  114. Svensson, K.J.; Christianson, H.C.; Kucharzewska, P.; Fagerstrom, V.; Lundstedt, L.; Borgquist, S.; Jirstrom, K.; Belting, M. Chondroitin Sulfate Expression Predicts Poor Outcome in Breast Cancer. Int. J. Oncol. 2011, 39, 1421–1428. [Google Scholar] [PubMed]
  115. Tsidulko, A.Y.; Kazanskaya, G.M.; Volkov, A.M.; Suhovskih, A.V.; Kiselev, R.S.; Kobozev, V.V.; Gaytan, A.S.; Krivoshapkin, A.L.; Aidagulova, S.V.; Grigorieva, E.V. Chondroitin Sulfate Content and Decorin Expression in Glioblastoma Are Associated with Proliferative Activity of Glioma Cells and Disease Prognosis. Cell Tissue Res. 2020, 379, 147–155. [Google Scholar] [CrossRef] [PubMed]
  116. Ricciardelli, C.; Brooks, J.H.; Suwiwat, S.; Sakko, A.J.; Mayne, K.; Raymond, W.A.; Seshadri, R.; LeBaron, R.G.; Horsfall, D.J. Regulation of Stromal Versican Expression by Breast Cancer Cells and Importance to Relapse-Free Survival in Patients with Node-Negative Primary Breast Cancer. Clin. Cancer Res. 2002, 8, 1054–1060. [Google Scholar] [PubMed]
  117. Silver, D.J.; Silver, J. Contributions of Chondroitin Sulfate Proteoglycans to Neurodevelopment, Injury, and Cancer. Curr. Opin. Neurobiol. 2014, 27, 171–178. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Zhang, H.; Kelly, G.; Zerillo, C.; Jaworski, D.M.; Hockfield, S. Expression of a Cleaved Brain-Specific Extracellular Matrix Protein Mediates Glioma Cell Invasion in Vivo. J. Neurosci. 1998, 18, 2370–2376. [Google Scholar] [CrossRef] [Green Version]
  119. Silver, D.J.; Siebzehnrubl, F.A.; Schildts, M.J.; Yachnis, A.T.; Smith, G.M.; Smith, A.A.; Scheffler, B.; Reynolds, B.A.; Silver, J.; Steindler, D.A. Chondroitin Sulfate Proteoglycans Potently Inhibit Invasion and Serve as a Central Organizer of the Brain Tumor Microenvironment. J. Neurosci. 2013, 33, 15603–15617. [Google Scholar] [CrossRef] [Green Version]
  120. Viapiano, M.S.; Bi, W.L.; Piepmeier, J.; Hockfield, S.; Matthews, R.T. Novel Tumor-Specific Isoforms of Behab/Brevican Identified in Human Malignant Gliomas. Cancer Res. 2005, 65, 6726–6733. [Google Scholar] [CrossRef] [Green Version]
  121. Muller, S.; Kunkel, P.; Lamszus, K.; Ulbricht, U.; Lorente, G.A.; Nelson, A.M.; von Schack, D.; Chin, D.J.; Lohr, S.C.; Westphal, M.; et al. A Role for Receptor Tyrosine Phosphatase Zeta in Glioma Cell Migration. Oncogene 2003, 22, 6661–6668. [Google Scholar] [CrossRef] [Green Version]
  122. Arslan, F.; Bosserhoff, A.K.; Nickl-Jockschat, T.; Doerfelt, A.; Bogdahn, U.; Hau, P. The Role of Versican Isoforms V0/V1 in Glioma Migration Mediated by Transforming Growth Factor-Β2. Br. J. Cancer 2007, 96, 1560–1568. [Google Scholar] [CrossRef]
  123. Viapiano, M.S.; Hockfield, S.; Matthews, R.T. Behab/Brevican Requires Adamts-Mediated Proteolytic Cleavage to Promote Glioma Invasion. J. Neurooncol. 2008, 88, 261–272. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Kovalszky, I.; Pogany, G.; Molnar, G.; Jeney, A.; Lapis, K.; Karacsonyi, S.; Szecseny, A.; Iozzo, R.V. Altered Glycosaminoglycan Composition in Reactive and Neoplastic Human Liver. Biochem. Biophys. Res. Commun. 1990, 167, 883–890. [Google Scholar] [CrossRef]
  125. Lan, Y.; Li, X.; Liu, Y.; He, Y.; Hao, C.; Wang, H.; Jin, L.; Zhang, G.; Zhang, S.; Zhou, A.; et al. Pingyangmycin Inhibits Glycosaminoglycan Sulphation in Both Cancer Cells and Tumour Tissues. J. Cell. Mol. Med. 2020, 24, 3419–3430. [Google Scholar] [CrossRef] [Green Version]
  126. Sobue, M.; Takeuchi, J.; Miura, K.; Kawase, K.; Mizuno, F.; Sato, E. Glycosaminoglycan Content and Synthesis in Gastric Carcinoma. Br. J. Cancer 1980, 42, 79–84. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Theocharis, A.D.; Tsara, M.E.; Papageorgacopoulou, N.; Karavias, D.D.; Theocharis, D.A. Pancreatic Carcinoma Is Characterized by Elevated Content of Hyaluronan and Chondroitin Sulfate with Altered Disaccharide Composition. Biochim. Biophys. Acta 2000, 1502, 201–206. [Google Scholar] [CrossRef] [Green Version]
  128. Pudelko, A.; Wisowski, G.; Olczyk, K.; Kozma, E.M. The Dual Role of the Glycosaminoglycan Chondroitin-6-Sulfate in the Development, Progression and Metastasis of Cancer. FEBS J. 2019, 286, 1815–1837. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Morla, S. Glycosaminoglycans and Glycosaminoglycan Mimetics in Cancer and Inflammation. Int. J. Mol. Sci. 2019, 20, 1963. [Google Scholar] [CrossRef] [Green Version]
  130. Skandalis, S.S.; Kletsas, D.; Kyriakopoulou, D.; Stavropoulos, M.; Theocharis, D.A. The Greatly Increased Amounts of Accumulated Versican and Decorin with Specific Post-Translational Modifications May Be Closely Associated with the Malignant Phenotype of Pancreatic Cancer. Biochim. Biophys. Acta 2006, 1760, 1217–1225. [Google Scholar] [CrossRef]
  131. Al-Nakouzi, N.; Wang, C.K.; Oo, H.Z.; Nelepcu, I.; Lallous, N.; Spliid, C.B.; Khazamipour, N.; Lo, J.; Truong, S.; Collins, C.; et al. Reformation of the Chondroitin Sulfate Glycocalyx Enables Progression of Ar-Independent Prostate Cancer. Nat. Commun. 2022, 13, 4760. [Google Scholar] [CrossRef]
  132. Theocharis, A.D.; Vynios, D.H.; Papageorgakopoulou, N.; Skandalis, S.S.; Theocharis, D.A. Altered Content Composition and Structure of Glycosaminoglycans and Proteoglycans in Gastric Carcinoma. Int. J. Biochem. Cell Biol. 2003, 35, 376–390. [Google Scholar] [CrossRef]
  133. Ten Dam, G.B.; van de Westerlo, E.M.; Purushothaman, A.; Stan, R.V.; Bulten, J.; Sweep, F.C.; Massuger, L.F.; Sugahara, K.; van Kuppevelt, T.H. Antibody Gd3g7 Selected against Embryonic Glycosaminoglycans Defines Chondroitin Sulfate-E Domains Highly up-Regulated in Ovarian Cancer and Involved in Vascular Endothelial Growth Factor Binding. Am. J. Pathol. 2007, 171, 1324–1333. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Li, F.; Ten Dam, G.B.; Murugan, S.; Yamada, S.; Hashiguchi, T.; Mizumoto, S.; Oguri, K.; Okayama, M.; van Kuppevelt, T.H.; Sugahara, K. Involvement of Highly Sulfated Chondroitin Sulfate in the Metastasis of the Lewis Lung Carcinoma Cells. J. Biol. Chem. 2008, 283, 34294–34304. [Google Scholar] [CrossRef] [PubMed]
  135. Seidler, D.G.; Breuer, E.; Grande-Allen, K.J.; Hascall, V.C.; Kresse, H. Core Protein Dependence of Epimerization of Glucuronosyl Residues in Galactosaminoglycans. J. Biol. Chem. 2002, 277, 42409–42416. [Google Scholar] [CrossRef] [Green Version]
  136. Momose, T.; Yoshimura, Y.; Harumiya, S.; Isobe, K.; Kito, M.; Fukushima, M.; Kato, H.; Nakayama, J. Chondroitin Sulfate Synthase 1 Expression Is Associated with Malignant Potential of Soft Tissue Sarcomas with Myxoid Substance. Hum. Pathol. 2016, 50, 15–23. [Google Scholar] [CrossRef] [Green Version]
  137. Liu, C.H.; Lan, C.T.; Chou, J.F.; Tseng, T.J.; Liao, W.C. Chsy1 Promotes Aggressive Phenotypes of Hepatocellular Carcinoma Cells Via Activation of the Hedgehog Signaling Pathway. Cancer Lett. 2017, 403, 280–288. [Google Scholar] [CrossRef]
  138. Honda, T.; Kaneiwa, T.; Mizumoto, S.; Sugahara, K.; Yamada, S. Hyaluronidases Have Strong Hydrolytic Activity toward Chondroitin 4-Sulfate Comparable to That for Hyaluronan. Biomolecules 2012, 2, 549–563. [Google Scholar] [CrossRef] [Green Version]
  139. Nikitovic, D.; Assouti, M.; Sifaki, M.; Katonis, P.; Krasagakis, K.; Karamanos, N.K.; Tzanakakis, G.N. Chondroitin Sulfate and Heparan Sulfate-Containing Proteoglycans Are Both Partners and Targets of Basic Fibroblast Growth Factor-Mediated Proliferation in Human Metastatic Melanoma Cell Lines. Int. J. Biochem. Cell Biol. 2008, 40, 72–83. [Google Scholar] [CrossRef]
  140. Nikitovic, D.; Zafiropoulos, A.; Tzanakakis, G.N.; Karamanos, N.K.; Tsatsakis, A.M. Effects of Glycosaminoglycans on Cell Proliferation of Normal Osteoblasts and Human Osteosarcoma Cells Depend on Their Type and Fine Chemical Compositions. Anticancer Res. 2005, 25, 2851–2856. [Google Scholar]
  141. Merle, B.; Durussel, L.; Delmas, P.D.; Clézardin, P. Decorin Inhibits Cell Migration through a Process Requiring Its Glycosaminoglycan Side Chain. J. Cell Biochem. 1999, 75, 538–546. [Google Scholar] [CrossRef]
  142. Thelin, M.A.; Svensson, K.J.; Shi, X.; Bagher, M.; Axelsson, J.; Isinger-Ekstrand, A.; van Kuppevelt, T.H.; Johansson, J.; Nilbert, M.; Zaia, J.; et al. Dermatan Sulfate Is Involved in the Tumorigenic Properties of Esophagus Squamous Cell Carcinoma. Cancer Res. 2012, 72, 1943–1952. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Auvinen, P.; Tammi, R.; Parkkinen, J.; Tammi, M.; Ågren, U.; Johansson, R.; Hirvikoski, P.; Eskelinen, M.; Kosma, V.M. Hyaluronan in Peritumoral Stroma and Malignant Cells Associates with Breast Cancer Spreading and Predicts Survival. Am. J. Pathol. 2000, 156, 529–536. [Google Scholar] [CrossRef]
  144. Pirinen, R.; Tammi, R.; Tammi, M.; Hirvikoski, P.; Parkkinen, J.J.; Johansson, R.; Böhm, J.; Hollmén, S.; Kosma, V.M. Prognostic Value of Hyaluronan Expression in Non-Small-Cell Lung Cancer: Increased Stromal Expression Indicates Unfavorable Outcome in Patients with Adenocarcinoma. Int. J. Cancer 2001, 95, 12–17. [Google Scholar] [CrossRef]
  145. Anttila, M.; Tammi, R.; Tammi, M.; Syrjänen, K.J.; Saarikoski, S.V.; Kosma, V.M. High Levels of Stromal Hyaluronan Predict Poor Disease Outcome in Epithelial Ovarian Cancer. Cancer Res. 2000, 60, 150–155. [Google Scholar]
  146. Spinelli, F.M.; Vitale, D.L.; Sevic, I.; Alaniz, L. Hyaluronan in the Tumor Microenvironment. Adv. Exp. Med. Biol. 2020, 1245, 67–83. [Google Scholar]
  147. Itano, N.; Kimata, K. Mammalian Hyaluronan Synthases. IUBMB Life 2002, 54, 195–199. [Google Scholar] [CrossRef]
  148. Tammi, R.H.; Passi, A.G.; Rilla, K.; Karousou, E.; Vigetti, D.; Makkonen, K.; Tammi, M.I. Transcriptional and Post-Translational Regulation of Hyaluronan Synthesis. FEBS J. 2011, 278, 1419–1428. [Google Scholar] [CrossRef]
  149. Bourguignon, L.Y.; Wong, G.; Earle, C.A.; Xia, W. Interaction of Low Molecular Weight Hyaluronan with Cd44 and Toll-Like Receptors Promotes the Actin Filament-Associated Protein 110-Actin Binding and Myd88-Nfκb Signaling Leading to Proinflammatory Cytokine/Chemokine Production and Breast Tumor Invasion. Cytoskeleton 2011, 68, 671–693. [Google Scholar] [CrossRef] [Green Version]
  150. Schwertfeger, K.L.; Cowman, M.K.; Telmer, P.G.; Turley, E.A.; McCarthy, J.B. Hyaluronan, Inflammation, and Breast Cancer Progression. Front. Immunol. 2015, 6, 236. [Google Scholar] [CrossRef]
  151. Misra, S.; Hascall, V.C.; Markwald, R.R.; Ghatak, S. Interactions between Hyaluronan and Its Receptors (Cd44, Rhamm) Regulate the Activities of Inflammation and Cancer. Front. Immunol. 2015, 6, 201. [Google Scholar] [CrossRef] [Green Version]
  152. Bourguignon, L.Y.; Singleton, P.A.; Zhu, H.; Diedrich, F. Hyaluronan-Mediated Cd44 Interaction with Rhogef and Rho Kinase Promotes Grb2-Associated Binder-1 Phosphorylation and Phosphatidylinositol 3-Kinase Signaling Leading to Cytokine (Macrophage-Colony Stimulating Factor) Production and Breast Tumor Progression. J. Biol. Chem. 2003, 278, 29420–29434. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Okamoto, I.; Tsuiki, H.; Kenyon, L.C.; Godwin, A.K.; Emlet, D.R.; Holgado-Madruga, M.; Lanham, I.S.; Joynes, C.J.; Vo, K.T.; Guha, A.; et al. Proteolytic Cleavage of the Cd44 Adhesion Molecule in Multiple Human Tumors. Am. J. Pathol. 2002, 160, 441–447. [Google Scholar] [CrossRef] [Green Version]
  154. Sugahara, K.N.; Murai, T.; Nishinakamura, H.; Kawashima, H.; Saya, H.; Miyasaka, M. Hyaluronan Oligosaccharides Induce Cd44 Cleavage and Promote Cell Migration in Cd44-Expressing Tumor Cells. J. Biol. Chem. 2003, 278, 32259–32265. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Kato, Y.; Hayatsu, N.; Kaneko, M.K.; Ogasawara, S.; Hamano, T.; Takahashi, S.; Nishikawa, R.; Matsutani, M.; Mishima, K.; Narimatsu, H. Increased Expression of Highly Sulfated Keratan Sulfate Synthesized in Malignant Astrocytic Tumors. Biochem. Biophys. Res. Commun. 2008, 369, 1041–1046. [Google Scholar] [CrossRef]
  156. Chhabra, M.; Doherty, G.G.; See, N.W.; Gandhi, N.S.; Ferro, V. From Cancer to COVID-19: A Perspective on Targeting Heparan Sulfate-Protein Interactions. Chem. Rec. 2021, 21, 1–16. [Google Scholar] [CrossRef]
  157. Cole, J.B.; Florez, J.C. Genetics of Diabetes Mellitus and Diabetes Complications. Nat. Rev. Nephrol. 2020, 16, 377–390. [Google Scholar] [CrossRef]
  158. Yang, Q.Q.; Sun, J.W.; Shao, D.; Zhang, H.H.; Bai, C.F.; Cao, F.L. The Association between Diabetes Complications, Diabetes Distress, and Depressive Symptoms in Patients with Type 2 Diabetes Mellitus. Clin. Nurs. Res. 2021, 30, 293–301. [Google Scholar] [CrossRef]
  159. Gowd, V.; Gurukar, A.; Chilkunda, N.D. Glycosaminoglycan Remodeling During Diabetes and the Role of Dietary Factors in Their Modulation. World J. Diabetes 2016, 7, 67–73. [Google Scholar] [CrossRef]
  160. Irving-Rodgers, H.F.; Ziolkowski, A.F.; Parish, C.R.; Sado, Y.; Ninomiya, Y.; Simeonovic, C.J.; Rodgers, R.J. Molecular Composition of the Peri-Islet Basement Membrane in Nod Mice: A Barrier against Destructive Insulitis. Diabetologia 2008, 51, 1680–1688. [Google Scholar] [CrossRef] [Green Version]
  161. Ziolkowski, A.F.; Popp, S.K.; Freeman, C.; Parish, C.R.; Simeonovic, C.J. Heparan Sulfate and Heparanase Play Key Roles in Mouse Beta Cell Survival and Autoimmune Diabetes. J. Clin. Investig. 2012, 122, 132–141. [Google Scholar] [CrossRef] [Green Version]
  162. Simeonovic, C.J.; Popp, S.K.; Starrs, L.M.; Brown, D.J.; Ziolkowski, A.F.; Ludwig, B.; Bornstein, S.R.; Wilson, J.D.; Pugliese, A.; Kay, T.W.H.; et al. Loss of Intra-Islet Heparan Sulfate Is a Highly Sensitive Marker of Type 1 Diabetes Progression in Humans. PLoS ONE 2018, 13, e0191360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Theodoraki, A.; Hu, Y.; Poopalasundaram, S.; Oosterhof, A.; Guimond, S.E.; Disterer, P.; Khoo, B.; Hauge-Evans, A.C.; Jones, P.M.; Turnbull, J.E.; et al. Distinct Patterns of Heparan Sulphate in Pancreatic Islets Suggest Novel Roles in Paracrine Islet Regulation. Mol. Cell. Endocrinol. 2015, 399, 296–310. [Google Scholar] [CrossRef] [PubMed]
  164. Simeonovic, C.J.; Ziolkowski, A.F.; Wu, Z.; Choong, F.J.; Freeman, C.; Parish, C.R. Heparanase and Autoimmune Diabetes. Front. Immunol. 2013, 4, 471. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Simeonovic, C.J.; Popp, S.K.; Brown, D.K.; Li, F.J.; Lafferty, A.R.A.; Freeman, C.; Parish, C.R. Heparanase and Type 1 Diabetes. Adv. Exp. Med. Biol. 2020, 1221, 607–630. [Google Scholar]
  166. Raats, C.J.; Bakker, M.A.; van den Born, J.; Berden, J.H. Hydroxyl Radicals Depolymerize Glomerular Heparan Sulfate in Vitro and in Experimental Nephrotic Syndrome. J. Biol. Chem. 1997, 272, 26734–26741. [Google Scholar] [CrossRef] [Green Version]
  167. Rota, C.; Liverani, L.; Spelta, F.; Mascellani, G.; Tomasi, A.; Iannone, A.; Vismara, E. Free Radical Generation During Chemical Depolymerization of Heparin. Anal. Biochem. 2005, 344, 193–203. [Google Scholar] [CrossRef]
  168. Dhounchak, S.; Popp, S.K.; Brown, D.J.; Laybutt, D.R.; Biden, T.J.; Bornstein, S.R.; Parish, C.R.; Simeonovic, C.J. Heparan Sulfate Proteoglycans in Beta Cells Provide a Critical Link between Endoplasmic Reticulum Stress, Oxidative Stress and Type 2 Diabetes. PLoS ONE 2021, 16, e0252607. [Google Scholar] [CrossRef]
  169. Liu, Z.; Lv, Y.; Zhao, N.; Guan, G.; Wang, J. Protein Kinase R-Like Er Kinase and Its Role in Endoplasmic Reticulum Stress-Decided Cell Fate. Cell Death Dis. 2015, 6, e1822. [Google Scholar] [CrossRef] [Green Version]
  170. Scheuner, D.; Kaufman, R. The Unfolded Protein Response: A Pathway That Links Insulin Demand with Beta-Cell Failure and Diabetes. Endocr. Rev. 2008, 29, 631. [Google Scholar] [CrossRef] [Green Version]
  171. Laybutt, D.R.; Preston, A.M.; Akerfeldt, M.C.; Kench, J.G.; Busch, A.K.; Biankin, A.V.; Biden, T.J. Endoplasmic Reticulum Stress Contributes to Beta Cell Apoptosis in Type 2 Diabetes. Diabetologia 2007, 50, 752–763. [Google Scholar] [CrossRef] [Green Version]
  172. Chan, J.Y.; Luzuriaga, J.; Bensellam, M.; Biden, T.J.; Laybutt, D.R. Failure of the Adaptive Unfolded Protein Response in Islets of Obese Mice Is Linked with Abnormalities in Β-Cell Gene Expression and Progression to Diabetes. Diabetes 2013, 62, 1557–1568. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Asthana, S.; Mallick, B.; Alexandrescu, A.T.; Jha, S. Iapp in Type Ii Diabetes: Basic Research on Structure, Molecular Interactions, and Disease Mechanisms Suggests Potential Intervention Strategies. Biochim. Biophys. Acta Biomembr. 2018, 1860, 1765–1782. [Google Scholar] [CrossRef] [PubMed]
  174. De Carufel, C.A.; Nguyen, P.T.; Sahnouni, S.; Bourgault, S. New Insights into the Roles of Sulfated Glycosaminoglycans in Islet Amyloid Polypeptide Amyloidogenesis and Cytotoxicity. Biopolymers 2013, 100, 645–655. [Google Scholar] [CrossRef] [PubMed]
  175. Castillo, G.M.; Cummings, J.A.; Yang, W.; Judge, M.E.; Sheardown, M.J.; Rimvall, K.; Hansen, J.B.; Snow, A.D. Sulfate Content and Specific Glycosaminoglycan Backbone of Perlecan Are Critical for Perlecan’s Enhancement of Islet Amyloid Polypeptide (Amylin) Fibril Formation. Diabetes 1998, 47, 612–620. [Google Scholar] [CrossRef]
  176. Quittot, N.; Fortier, M.; Babych, M.; Nguyen, P.T.; Sebastiao, M.; Bourgault, S. Cell Surface Glycosaminoglycans Exacerbate Plasma Membrane Perturbation Induced by the Islet Amyloid Polypeptide. FASEB J. 2021, 35, e21306. [Google Scholar] [CrossRef]
  177. Wang, H.; Raleigh, D.P. The Ability of Insulin to Inhibit the Formation of Amyloid by Pro-Islet Amyloid Polypeptide Processing Intermediates Is Significantly Reduced in the Presence of Sulfated Glycosaminoglycans. Biochemistry 2014, 53, 2605–2614. [Google Scholar] [CrossRef]
  178. Bogdani, M.; Johnson, P.Y.; Potter-Perigo, S.; Nagy, N.; Day, A.J.; Bollyky, P.L.; Wight, T.N. Ha-Hyaluronan and Hyaluronan-Binding Proteins Accumulate in Both Human Type 1 Diabetic Islets and Lymphoid Tissues and Associate with Inflammatory Cells in Insulitis. Diabetes 2014, 63, 2727–2743. [Google Scholar] [CrossRef]
  179. Bogdani, M.; Simeonovic, C.; Nagy, N.; Johnson, P.Y.; Chan, C.K.; Wight, T.N. The Detection of Glycosaminoglycans in Pancreatic Islets and Lymphoid Tissues. Methods Mol. Biol. 2015, 1229, 413–430. [Google Scholar]
  180. Poplawska-Kita, A.; Mierzejewska-Iwanowska, B.; Szelachowska, M.; Siewko, K.; Nikolajuk, A.; Kinalska, I.; Gorska, M. Glycosaminoglycans Urinary Excretion as a Marker of the Early Stages of Diabetic Nephropathy and the Disease Progression. Diabetes Metab. Res. Rev. 2008, 24, 310–317. [Google Scholar] [CrossRef]
  181. Parthasarathy, N.; Spiro, R.G. Effect of Diabetes on the Glycosaminoglycan Component of the Human Glomerular Basement Membrane. Diabetes 1982, 31, 738–741. [Google Scholar] [CrossRef]
  182. Wijnhoven, T.J.; van den Hoven, M.J.; Ding, H.; van Kuppevelt, T.H.; van der Vlag, J.; Berden, J.H.; Prinz, R.A.; Lewis, E.J.; Schwartz, M.; Xu, X. Heparanase Induces a Differential Loss of Heparan Sulphate Domains in Overt Diabetic Nephropathy. Diabetologia 2008, 51, 372–382. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Schumacher, V.A.; Schlotzer-Schrehardt, U.; Karumanchi, S.A.; Shi, X.; Zaia, J.; Jeruschke, S.; Zhang, D.; Pavenstadt, H.; Drenckhan, A.; Amann, K.; et al. Wt1-Dependent Sulfatase Expression Maintains the Normal Glomerular Filtration Barrier. J. Am. Soc. Nephrol. 2011, 22, 1286–1296. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Lauer, M.E.; Hascall, V.C.; Wang, A. Heparan Sulfate Analysis from Diabetic Rat Glomeruli. J. Biol. Chem. 2007, 282, 843–852. [Google Scholar] [CrossRef] [Green Version]
  185. Van den Born, J.; Pisa, B.; Bakker, M.A.; Celie, J.W.; Straatman, C.; Thomas, S.; Viberti, G.C.; Kjellen, L.; Berden, J.H. No Change in Glomerular Heparan Sulfate Structure in Early Human and Experimental Diabetic Nephropathy. J. Biol. Chem. 2006, 281, 29606–29613. [Google Scholar] [CrossRef] [Green Version]
  186. Reine, T.M.; Grondahl, F.; Jenssen, T.G.; Hadler-Olsen, E.; Prydz, K.; Kolset, S.O. Reduced Sulfation of Chondroitin Sulfate but Not Heparan Sulfate in Kidneys of Diabetic Db/Db Mice. J. Histochem. Cytochem. 2013, 61, 606–616. [Google Scholar] [CrossRef] [Green Version]
  187. Joladarashi, D.; Salimath, P.V.; Chilkunda, N.D. Diabetes Results in Structural Alteration of Chondroitin Sulfate/Dermatan Sulfate in the Rat Kidney:Effects on the Binding to Extracellular Matrix Components. Glycobiology 2011, 21, 960–972. [Google Scholar] [CrossRef] [Green Version]
  188. Jura-Poltorak, A.; Olczyk, P.; Chalas-Lipka, A.; Komosinska-Vassev, K.; Kuznik-Trocha, K.; Winsz-Szczotka, K.; Ivanova, D.; Kiselova-Kaneva, Y.; Krysik, K.; Telega, A.; et al. Urinary Sulphated Glycosaminoglycans Excretion in Obese Patients with Type 2 Diabetes Mellitus Treated with Metformin. Arch. Physiol. Biochem. 2022, 128, 507–513. [Google Scholar] [CrossRef] [PubMed]
  189. Zhao, T.; Lu, X.; Davies, N.; Gong, Y.; Guo, J.; Zhang, H.; Li, Z.; Hong, J.; Fu, G.; Li, P. Diabetes Results in Structural Alteration of Chondroitin Sulfate in the Urine. J. Pharm. Sci. 2013, 16, 486–493. [Google Scholar] [CrossRef] [PubMed]
  190. Srikanth, C.B.; Salimath, P.V.; Nandini, C.D. Erythrocytes Express Chondroitin Sulphate/Dermatan Sulphate, Which Undergoes Quantitative Changes During Diabetes and Mediate Erythrocyte Adhesion to Extracellular Matrix Components. Biochimie 2012, 94, 1347–1355. [Google Scholar] [CrossRef]
  191. Morofuji, Y.; Nakagawa, S. Drug Development for Central Nervous System Diseases Using in Vitro Blood-Brain Barrier Models and Drug Repositioning. Curr. Pharm. Des. 2020, 26, 1466–1485. [Google Scholar] [CrossRef]
  192. Cenini, G.; Voos, W. Mitochondria as Potential Targets in Alzheimer Disease Therapy: An Update. Front. Pharmacol. 2019, 10, 902. [Google Scholar] [CrossRef] [PubMed]
  193. Livingston, G.; Sommerlad, A.; Orgeta, V.; Costafreda, S.G.; Huntley, J.; Ames, D.; Ballard, C.; Banerjee, S.; Burns, A.; Cohen-Mansfield, J.; et al. Dementia Prevention, Intervention, and Care. Lancet 2017, 390, 2673–2734. [Google Scholar] [CrossRef] [Green Version]
  194. Hardy, J.A.; Higgins, G.A. Alzheimer’s Disease the Amyloid Cascade Hypothesis. Science 1992, 256, 184–185. [Google Scholar] [CrossRef] [PubMed]
  195. Krishnaswamy, V.R.; Benbenishty, A.; Blinder, P.; Sagi, I. Demystifying the Extracellular Matrix and Its Proteolytic Remodeling in the Brain: Structural and Functional Insights. Cell. Mol. Life Sci. 2019, 76, 3229–3248. [Google Scholar] [CrossRef]
  196. Bonneh-Barkay, D.; Wiley, C.A. Brain Extracellular Matrix in Neurodegeneration. Brain Pathol. 2009, 19, 573–585. [Google Scholar] [CrossRef]
  197. Bandtlow, C.E.; Zimmermann, D.R. Proteoglycans in the Developing Brain New Conceptual Insights for Old Proteins. Physiol. Rev. 2000, 80, 1267–1290. [Google Scholar] [CrossRef] [Green Version]
  198. Smith, P.D.; Coulson-Thomas, V.J.; Foscarin, S.; Kwok, J.C.; Fawcett, J.W. “Gag-Ing with the Neuron”: The Role of Glycosaminoglycan Patterning in the Central Nervous System. Exp. Neurol. 2015, 274, 100–114. [Google Scholar] [CrossRef]
  199. Soleman, S.; Filippov, M.A.; Dityatev, A.; Fawcett, J.W. Targeting the Neural Extracellular Matrix in Neurological Disorders. Neuroscience 2013, 253, 194–213. [Google Scholar] [CrossRef]
  200. Carulli, D.; Laabs, T.; Geller, H.M.; Fawcett, J.W. Chondroitin Sulfate Proteoglycans in Neural Development and Regeneration. Curr. Opin. Neurobiol. 2005, 15, 116–120. [Google Scholar] [CrossRef]
  201. Logsdon, A.F.; Francis, K.L.; Richardson, N.E.; Hu, S.J.; Faber, C.L.; Phan, B.A.; Nguyen, V.; Setthavongsack, N.; Banks, W.A.; Woltjer, R.L.; et al. Decoding Perineuronal Net Glycan Sulfation Patterns in the Alzheimer’s Disease Brain. Alzheimers Dement. 2022, 18, 942–954. [Google Scholar] [CrossRef]
  202. Bertolotto, A.; Rocca, G.; Schiffer, D. Chondroitin-4-Sulfate Proteoglycan Forms an Extracellular Network in Human and Rat Central Nervous System. J. Neurol. Sci. 1990, 100, 113–123. [Google Scholar] [CrossRef]
  203. Zhao, J.; Zhu, Y.; Song, X.; Xiao, Y.; Su, G.; Liu, X.; Wang, Z.; Xu, Y.; Liu, J.; Eliezer, D.; et al. 3-O-Sulfation of Heparan Sulfate Enhances Tau Interaction and Cellular Uptake. Angew. Chem. Int. Ed. Engl. 2020, 59, 1818–1827. [Google Scholar] [CrossRef] [PubMed]
  204. Wang, H.; Katagiri, Y.; McCann, T.E.; Unsworth, E.; Goldsmith, P.; Yu, Z.X.; Tan, F.; Santiago, L.; Mills, E.M.; Wang, Y.; et al. Chondroitin-4-Sulfation Negatively Regulates Axonal Guidance and Growth. J. Cell Sci. 2008, 121, 3083–3091. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. Shioi, J.; Anderson, J.P.; Ripellino, J.A.; Robakis, N.K. Chondroitin Sulfate Proteoglycan Form of the Alzheimer’s Beta-Amyloid Precursor. J. Biol. Chem. 1992, 267, 13819–13822. [Google Scholar] [CrossRef]
  206. Mehra, S.; Ghosh, D.; Kumar, R.; Mondal, M.; Gadhe, L.G.; Das, S.; Anoop, A.; Jha, N.N.; Jacob, R.S.; Chatterjee, D.; et al. Glycosaminoglycans Have Variable Effects on Alpha-Synuclein Aggregation and Differentially Affect the Activities of the Resulting Amyloid Fibrils. J. Biol. Chem. 2018, 293, 12975–12991. [Google Scholar] [CrossRef] [Green Version]
  207. DeWitt, D.A.; Silver, J.; Canning, D.R.; Perry, G. Chondroitin Sulfate Proteoglycans Are Associated with the Lesions of Alzheimer’s Disease. Exp. Neurol. 1993, 121, 149–152. [Google Scholar] [CrossRef] [Green Version]
  208. Sobel, R.A.; Ahmed, A.S. White Matter Extracellular Matrix Chondroitin Sulfate/Dermatan Sulfate Proteoglycans in Multiple Sclerosis. J. Neuropathol. Exp. Neurol. 2001, 60, 1198–1207. [Google Scholar] [CrossRef] [Green Version]
  209. Stephenson, E.L.; Zhang, P.; Ghorbani, S.; Wang, A.; Gu, J.; Keough, M.B.; Rawji, K.S.; Silva, C.; Yong, V.W.; Ling, C.C. Targeting the Chondroitin Sulfate Proteoglycans: Evaluating Fluorinated Glucosamines and Xylosides in Screens Pertinent to Multiple Sclerosis. ACS Cent. Sci. 2019, 5, 1223–1234. [Google Scholar] [CrossRef]
  210. Keough, M.B.; Rogers, J.A.; Zhang, P.; Jensen, S.K.; Stephenson, E.L.; Chen, T.; Hurlbert, M.G.; Lau, L.W.; Rawji, K.S.; Plemel, J.R.; et al. An Inhibitor of Chondroitin Sulfate Proteoglycan Synthesis Promotes Central Nervous System Remyelination. Nat. Commun. 2016, 7, 11312. [Google Scholar] [CrossRef] [Green Version]
  211. Warford, J.R.; Lamport, A.C.; Clements, D.R.; Malone, A.; Kennedy, B.E.; Kim, Y.; Gujar, S.A.; Hoskin, D.W.; Easton, A.S. Surfen, a Proteoglycan Binding Agent, Reduces Inflammation but Inhibits Remyelination in Murine Models of Multiple Sclerosis. Acta Neuropathol. Commun. 2018, 6, 4. [Google Scholar] [CrossRef] [Green Version]
  212. Lau, L.W.; Cua, R.; Keough, M.B.; Haylock-Jacobs, S.; Yong, V.W. Pathophysiology of the Brain Extracellular Matrix: A New Target for Remyelination. Nat. Rev. Neurosci. 2013, 14, 722–729. [Google Scholar] [CrossRef] [PubMed]
  213. Pendleton, J.C.; Shamblott, M.J.; Gary, D.S.; Belegu, V.; Hurtado, A.; Malone, M.L.; McDonald, J.W. Chondroitin Sulfate Proteoglycans Inhibit Oligodendrocyte Myelination through Ptpsigma. Exp. Neurol. 2013, 247, 113–121. [Google Scholar] [CrossRef] [PubMed]
  214. Gilbert, R.J.; McKeon, R.J.; Darr, A.; Calabro, A.; Hascall, V.C.; Bellamkonda, R.V. Cs-4,6 Is Differentiallyupregulated in Glial Scar and Is a Potent Inhibitor of Neurite Extension. Mol. Cell. Neurosci. 2005, 29, 545–558. [Google Scholar] [CrossRef]
  215. Maiza, A.; Chantepie, S.; Vera, C.; Fifre, A.; Huynh, M.B.; Stettler, O.; Ouidja, M.O.; Papy-Garcia, D. The Role of Heparan Sulfates in Protein Aggregation and Their Potential Impact on Neurodegeneration. FEBS Lett. 2018, 592, 3806–3818. [Google Scholar] [CrossRef] [Green Version]
  216. Yamazaki, T.; Iharass, Y. The Molecular Pathology of Alzheimer’s Disease. Neuropathology 1997, 17, 263–269. [Google Scholar] [CrossRef]
  217. Shimizu, H.; Ghazizadeh, M.; Sato, S.; Oguro, T.; Kawanami, O. Interaction between Beta-Amyloid Protein and Heparan Sulfate Proteoglycans from the Cerebral Capillary Basement Membrane in Alzheimer’s Disease. J. Clin. Neurosci. 2009, 16, 277–2782. [Google Scholar] [CrossRef] [PubMed]
  218. Bruinsma, I.B.; Te Riet, L.; Gevers, T.; ten Dam, G.B.; van Kuppevelt, T.H.; David, G.; Kusters, B.; de Waal, R.M.; Verbeek, M.M. Sulfation of Heparan Sulfate Associated with Amyloid-Beta Plaques in Patients with Alzheimer’s Disease. Acta Neuropathol. 2010, 119, 211–220. [Google Scholar] [CrossRef]
  219. Lindahl, B.; Westling, C.; Gimenez-Gallego, G.; Lindahl, U.; Salmivirta, M. Common Binding Sites for Beta-Amyloid Fibrils and Fibroblast Growth Factor-2 in Heparan Sulfate from Human Cerebral Cortex. J. Biol. Chem. 1999, 274, 30631–30635. [Google Scholar] [CrossRef]
  220. Zhou, X.; Wang, Y.; Zheng, W.; Deng, G.; Wang, F.; Jin, L. Characterizing Heparin Tetrasaccharides Binding to Amyloid-Beta Peptide. Front. Mol. Biosci. 2022, 9, 824146. [Google Scholar] [CrossRef]
  221. Konno, T.; Oiki, S.; Hasegawa, K.; Naiki, H. Anionic Contribution for Fibrous Maturation of Protofibrillar Assemblies of the Human Tau Repeat Domain in a Fluoroalcohol Solution. Biochemistry 2004, 43, 13613–13620. [Google Scholar] [CrossRef]
  222. Liu, I.H.; Uversky, V.N.; Munishkina, L.A.; Fink, A.L.; Halfter, W.; Cole, G.J. Agrin Binds Alpha-Synuclein and Modulates Alpha-Synuclein Fibrillation. Glycobiology 2005, 15, 1320–1331. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  223. Tao, Y.; Sun, Y.; Lv, S.; Xia, W.; Zhao, K.; Xu, Q.; Zhao, Q.; He, L.; Le, W.; Wang, Y.; et al. Heparin Induces Alpha-Synuclein to Form New Fibril Polymorphs with Attenuated Neuropathology. Nat. Commun. 2022, 13, 4226. [Google Scholar] [CrossRef] [PubMed]
  224. Ihse, E.; Yamakado, H.; van Wijk, X.M.; Lawrence, R.; Esko, J.D.; Masliah, E. Cellular Internalization of Alpha-Synuclein Aggregates by Cell Surface Heparan Sulfate Depends on Aggregate Conformation and Cell Type. Sci. Rep. 2017, 7, 9008. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  225. DeWitt, D.A.; Richey, P.L.; Praprotnik, D.; Silver, J.; Perry, G. Chondroitin Sulfate Proteoglycans Are a Common Component of Neuronal Inclusions and Astrocytic Reaction in Neurodegenerative Diseases. Brain Res. 1994, 656, 205–209. [Google Scholar] [CrossRef] [Green Version]
  226. Ling, J.; Li, J.; Khan, A.; Lundkvist, A.; Li, J.P. Is Heparan Sulfate a Target for Inhibition of Rna Virus Infection? Am. J. Physiol. Cell Physiol. 2022, 322, C605–C613. [Google Scholar] [CrossRef]
  227. Shajahan, A.; Pepi, L.E.; Rouhani, D.S.; Heiss, C.; Azadi, P. Glycosylation of Sars-Cov-2: Structural and Functional Insights. Anal. Bioanal. Chem. 2021, 413, 7179–7193. [Google Scholar] [CrossRef]
  228. Sorin, M.N.; Kuhn, J.; Stasiak, A.C.; Stehle, T. Structural Insight into Non-Enveloped Virus Binding to Glycosaminoglycan Receptors: A Review. Viruses 2021, 13, 800. [Google Scholar] [CrossRef]
  229. Clausen, T.M.; Sandoval, D.R.; Spliid, C.B.; Pihl, J.; Perrett, H.R.; Painter, C.D.; Narayanan, A.; Majowicz, S.A.; Kwong, E.M.; McVicar, R.N.; et al. SARS-CoV-2 Infection Depends on Cellular Heparan Sulfate and Ace2. Cell 2020, 183, 1043–1057.e15. [Google Scholar] [CrossRef] [PubMed]
  230. Kim, S.Y.; Jin, W.; Sood, A.; Montgomery, D.W.; Grant, O.C.; Fuster, M.M.; Fu, L.; Dordick, J.S.; Woods, R.J.; Zhang, F.; et al. Glycosaminoglycan Binding Motif at S1/S2 Proteolytic Cleavage Site on Spike Glycoprotein May Facilitate Novel Coronavirus (SARS-CoV-2) Host Cell Entry. Mol. Biol. 2020. [Google Scholar] [CrossRef] [Green Version]
  231. Kearns, F.L.; Sandoval, D.R.; Casalino, L.; Clausen, T.M.; Rosenfeld, M.A.; Spliid, C.B.; Amaro, R.E.; Esko, J.D. Spike-Heparan Sulfate Interactions in SARS-CoV-2 Infection. Curr. Opin. Struc. Biol. 2022, 76, 102439. [Google Scholar] [CrossRef]
  232. Avirutnan, P.; Zhang, L.; Punyadee, N.; Manuyakorn, A.; Puttikhunt, C.; Kasinrerk, W.; Malasit, P.; Atkinson, J.P.; Diamond, M.S. Secreted Ns1 of Dengue Virus Attaches to the Surface of Cells Via Interactions with Heparan Sulfate and Chondroitin Sulfate E. PLoS Pathog. 2007, 3, e183. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  233. Dalrymple, N.; Mackow, E.R. Productive Dengue Virusinfection of Human Endothelial Cells Is Directed by Heparan Sulfate-Containing Proteoglycan Receptors. J. Virol. 2011, 85, 9478–9485. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  234. Barth, H.; Schafer, C.; Adah, M.I.; Zhang, F.; Linhardt, R.J.; Toyoda, H.; Kinoshita-Toyoda, A.; Toida, T.; van Kuppevelt, T.H.; Depla, E.; et al. Cellular Binding of Hepatitis C Virus Envelope Glycoprotein E2 Requires Cell Surface Heparan Sulfate. J. Biol. Chem. 2003, 278, 41003–41012. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  235. Uyama, T.; Ishida, M.; Izumikawa, T.; Trybala, E.; Tufaro, F.; Bergstrom, T.; Sugahara, K.; Kitagawa, H. Chondroitin 4-O-Sulfotransferase-1 Regulates E Disaccharide Expression of Chondroitin Sulfate Required for Herpes Simplex Virus Infectivity. J. Biol. Chem. 2006, 281, 38668–38674. [Google Scholar] [CrossRef]
  236. Abidine, Y.; Liu, L.; Wallen, O.; Trybala, E.; Olofsson, S.; Bergstrom, T.; Bally, M. Cellular Chondroitin Sulfate and the Mucin-Like Domain of Viral Glycoprotein C Promote Diffusion of Herpes Simplex Virus 1 While Heparan Sulfate Restricts Mobility. Viruses 2022, 14, 1836. [Google Scholar] [CrossRef] [PubMed]
  237. Kines, R.C.; Thompson, C.D.; Lowy, D.R.; Schiller, J.T.; Day, P.M. The Initial Steps Leading to Papillomavirus Infection Occur on the Basement Membrane Prior to Cell Surface Binding. Proc. Natl. Acad. Sci. USA 2009, 106, 20458–20463. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  238. Alcorn, M.D.H.; Klimstra, W.B. Glycosaminoglycan Binding by Arboviruses: A Cautionary Tale. J. Gen. Virol. 2022, 103. [Google Scholar] [CrossRef]
  239. Klenk, K.; Roberts, S.R. Use of a Vesicular Stomatitis Virus Complementation System to Analyze Respiratory Syncytial Virus Binding. Virus Res. 2002, 90, 327–335. [Google Scholar] [CrossRef]
  240. Shi, D.; He, P.; Song, Y.; Cheng, S.; Linhardt, R.J.; Dordick, J.S.; Chi, L.; Zhang, F. Kinetic and Structural Aspects of Glycosaminoglycan-Monkeypox Virus Protein A29 Interactions Using Surface Plasmon Resonance. Molecules 2022, 27, 5898. [Google Scholar] [CrossRef]
  241. Kim, S.Y.; Jin, W.; Sood, A.; Montgomery, D.W.; Grant, O.C.; Fuster, M.M.; Fu, L.; Dordick, J.S.; Woods, R.J.; Zhang, F.; et al. Characterization of Heparin and Severe Acute Respiratory Syndrome-Related Coronavirus 2 (SARS-CoV-2) Spike Glycoprotein Binding Interactions. Antivir. Res. 2020, 181, 104873. [Google Scholar] [CrossRef]
  242. Tiwari, V.; Tandon, R.; Sankaranarayanan, N.V.; Beer, J.C.; Kohlmeir, E.K.; Swanson-Mungerson, M.; Desai, U.R. Preferential Recognition and Antagonism of Sars-Cov-2 Spike Glycoprotein Binding to 3-O-Sulfated Heparan Sulfate. bioRxiv 2020. [Google Scholar] [CrossRef]
  243. Du Preez, H.N.; Aldous, C.; Hayden, M.R.; Kruger, H.G.; Lin, J. Pathogenesis of Covid-19 Described through the Lens of an Undersulfated and Degraded Epithelial and Endothelial Glycocalyx. FASEB J. 2022, 36, e22052. [Google Scholar] [CrossRef] [PubMed]
  244. Andonegui-Elguera, S.; Taniguchi-Ponciano, K.; Gonzalez-Bonilla, C.R.; Torres, J.; Mayani, H.; Herrera, L.A.; Pena-Martinez, E.; Silva-Roman, G.; Vela-Patino, S.; Ferreira-Hermosillo, A.; et al. Molecular Alterations Prompted by SARS-CoV-2 Infection: Induction of Hyaluronan, Glycosaminoglycan and Mucopolysaccharide Metabolism. Arch. Med. Res. 2020, 51, 645–653. [Google Scholar] [CrossRef] [PubMed]
  245. Burgess, J.K.; Harmsen, M.C. Chronic Lung Diseases: Entangled in Extracellular Matrix. Eur. Respir. Rev. 2022, 31, 210202. [Google Scholar] [CrossRef]
  246. Ennemoser, M.; Pum, A.; Kungl, A. Disease-Specifc Glycosaminoglycan Patterns in the Extracellular Matrix of Human Lung and Brain. Carbohyd. Res. 2022, 511, 108480. [Google Scholar] [CrossRef]
  247. Westergren-Thorsson, G.; Hedstrom, U.; Nybom, A.; Tykesson, E.; Ahrman, E.; Hornfelt, M.; Maccarana, M.; van Kuppevelt, T.H.; Dellgren, G.; Wildt, M.; et al. Increased Deposition of Glycosaminoglycans and Altered Structure of Heparan Sulfate in Idiopathic Pulmonary Fibrosis. Int. J. Biochem. Cell Biol. 2017, 83, 27–38. [Google Scholar] [CrossRef] [PubMed]
  248. Huang, J.; Olivenstein, R.; Taha, R.; Hamid, Q.; Ludwig, M. Enhanced Proteoglycan Deposition in the Airway Wall of Atopic Asthmatics. Am. J. Respir. Crit. Care Med. 1999, 160, 725–729. [Google Scholar] [CrossRef]
  249. Hallgren, O.; Alsafadi, H.N.; Nybom, A.; Zhou, X.; Bjermer, L.H.; Thorsson, G.W. Copd-Specific Chondroitin Sulfate Modifications Are Linked to Tgf-B1. Am. J. Respir. Crit. Care Med. 2019, 199, A5782. [Google Scholar]
  250. Reeves, E.P.; Williamson, M.; O’Neill, S.J.; Greally, P.; McElvaney, N.G. Nebulized Hypertonic Saline Decreases Il-8 in Sputum of Patients with Cystic Fibrosis. Am. J. Respir. Crit. Care Med. 2011, 183, 1517–1523. [Google Scholar] [CrossRef]
  251. Cheng, P.W.; Boat, T.F.; Cranfill, K.; Yankaskas, J.R.; BoucherR, C. Increased Sulfation of Glycoconjugates by Cultured Nasal Epithelial Cells from Patients with Cystic Fibrosis. J. Clin. Investig. 1989, 84, 68–72. [Google Scholar] [CrossRef] [Green Version]
  252. Kim, J.S.; Werth, V.P. Identification of Specific Chondroitin Sulfate Species in Cutaneous Autoimmune Disease. J. Histochem. Cytochem. 2011, 59, 780–790. [Google Scholar] [CrossRef] [PubMed]
  253. Nikkilä, M.T. Urinary Glycosaminoglycan Excretion in Normal and Stone-Forming Subjects: Significant Disturbance in Recurrent Stone Formers. Urol. Int. 1989, 44, 157–159. [Google Scholar] [CrossRef]
  254. Verkoelen, C.F.; Verhulst, A. Proposed Mechanisms in Renal Tubular Crystal Retention. Kidney Int. 2007, 72, 13–18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  255. Ustundağ, Y.; Huysal, K.; Guzelsoy, M.; Genim, C.E.; Yavuz, A. Urine and Serum Glycosaminoglycan Levels in the Diagnosis of Urological Diseases and Conditions: A Narrative Review of the Literature. Urol. J. 2021, 88, 103–109. [Google Scholar] [CrossRef] [PubMed]
  256. Shirane, Y.; Kurokawa, Y.; Miyashita, S.; Komatsu, H.; Kagawa, S. Study of Inhibition Mechanisms of Glycosaminoglycans on Calcium Oxalate Monohydrate Crystals by Atomic Force Microscopy. Urol. Res. 1999, 27, 426–431. [Google Scholar] [CrossRef]
  257. Suzuki, K.; Ryall, R.L. The Effect of Heparan Sulphate on the Crystallization of Calcium Oxalate in Undiluted, Ultrafiltered Human Urine. Br. J. Urol. 1996, 78, 15–21. [Google Scholar] [CrossRef]
  258. Gohel, M.D.I.; Shum, D.K.Y.; Tam, P.C. Electrophoretic Separation and Characterization of Urinary Glycosaminoglycans and Their Roles in Urolithiasis. Carbohyd. Res. 2007, 342, 79–86. [Google Scholar] [CrossRef]
  259. Dissayabutra, T.; Kalpongnukul, N.; Chindaphan, K.; Srisa-art, M.; Ungjaroenwathana, W.; Kaewwongse, M.; Lampenkhae, K.; Tosukhowong, P. Urinary Sulfated Glycosaminoglycan Insufficiency and Chondroitin Sulfate Supplement in Urolithiasis. PLoS ONE 2019, 14, e0213180. [Google Scholar] [CrossRef]
  260. Jappie, D.; Rodgers, A.; Ravenscroft, N.; Webber, D.; Gohel, M.D.I. Composition and Inhibitory Properties of Endogenous Urinary GAGs are Different in Subjects from Two Race Groups with Different Occurrence Rates of Kidney Stones: Pilot Studies Provide Unique Evidence in Support of an Inhibitory Role for This Group of Compounds. Clin. Chim. Acta 2022, 525, 84–90. [Google Scholar]
  261. Jappie, D.; Rodgers, A.; Webber, D.; Gohel, M.D.I. Seeking Consistency for the Role of Urinary Macromolecules and Glycosaminoglycans in Calcium Oxalate Crystallization Processes Pertaining to the Risk of Renal Stone Formation Using a Multi-Faceted Basic Science Approach. Clin. Chim. Acta 2021, 521, 76–84. [Google Scholar] [CrossRef]
  262. Pessentheiner, A.R.; Ducasa, G.M.; Gordts, P. Proteoglycans in Obesity-Associated Metabolic Dysfunction and Meta-Inflammation. Front. Immunol. 2020, 11, 769. [Google Scholar] [CrossRef] [PubMed]
  263. Hatano, S.; Watanabe, H. Regulation of Macrophage and Dendritic Cell Function by Chondroitin Sulfate in Innate to Antigen-Specific Adaptive Immunity. Front. Immunol. 2020, 11, 232. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  264. Reeves, S.R.; Barrow, K.A.; Rich, L.M.; White, M.P.; Shubin, N.J.; Chan, C.K.; Kang, I.; Ziegler, S.F.; Piliponsky, A.M.; Wight, T.N.; et al. Respiratory Syncytial Virus Infection of Human Lung Fibroblasts Induces a Hyaluronan-Enriched Extracellular Matrix That Binds Mast Cells and Enhances Expression of Mast Cell Proteases. Front. Immunol. 2019, 10, 3159. [Google Scholar] [CrossRef] [PubMed]
  265. Dong, Y.; Arif, A.A.; Guo, J.; Ha, Z.; Lee-Sayer, S.S.M.; Poon, G.F.T.; Dosanjh, M.; Roskelley, C.D.; Huan, T.; Johnson, P. Cd44 Loss Disrupts Lung Lipid Surfactant Homeostasis and Exacerbates Oxidized Lipid-Induced Lung Inflammation. Front. Immunol. 2020, 11, 29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  266. Zhang, B.; Chi, L. Chondroitin Sulfate/Dermatan Sulfate-Protein Interactions and Their Biological Functions in Human Diseases: Implications and Analytical Tools. Front. Cell Dev. Biol. 2021, 9, 693563. [Google Scholar] [CrossRef]
Figure 1. Distribution and structures (repeating disaccharide units) of GAGs in the extracellular matrix. CS/DS, HS, and KS are linked to core proteins, whereas HA is free. “Polymers 14 05014 i001” and “Polymers 14 05014 i002” indicate that the hydroxyl can be substituted by a sulfate group and acetyl, respectively. It is worth noting that the structural formula represents the maximum degree of sulfation for each GAG type. “Polymers 14 05014 i003” only represents the possibility of being substituted by a sulfate group. Such as CS, they can be classified as CS-A (GlcA-GalNAc4S), CS-C (GlcA-GalNAc6S), CS-D (GlcA2S-GalNAc6S), CS-E (GlcA4S-GalNAc6S), etc. (Used with permission of Royal Society of Chemistry from ref. [1]; permission conveyed through Copyright Clearance Center, Inc).
Figure 1. Distribution and structures (repeating disaccharide units) of GAGs in the extracellular matrix. CS/DS, HS, and KS are linked to core proteins, whereas HA is free. “Polymers 14 05014 i001” and “Polymers 14 05014 i002” indicate that the hydroxyl can be substituted by a sulfate group and acetyl, respectively. It is worth noting that the structural formula represents the maximum degree of sulfation for each GAG type. “Polymers 14 05014 i003” only represents the possibility of being substituted by a sulfate group. Such as CS, they can be classified as CS-A (GlcA-GalNAc4S), CS-C (GlcA-GalNAc6S), CS-D (GlcA2S-GalNAc6S), CS-E (GlcA4S-GalNAc6S), etc. (Used with permission of Royal Society of Chemistry from ref. [1]; permission conveyed through Copyright Clearance Center, Inc).
Polymers 14 05014 g001
Figure 2. Schematic diagram of the ternary complex of FGF/HS/FGFR and the key sites for HS binding. (a) FGFR comprises extracellular Ig-like domains, intracellular tyrosine kinase domains, and transmembrane domain. Ig-like domains bind FGF with the assistance of HS to form ternary complex; (b) HS binding to FGF2 requires 2-O-S of IdoA and N-S of GlcN; HS binding to FGF1 and FGFR requires 2-O-S of IdoA and 6-O-S, N-S of GlcN; the N-S of the nonreducing terminal residue is also necessary for HS binding to FGF/FGFR. The increased expression of HS in most tumor cells enhances FGF/FGFR signal transduction, which is beneficial to tumor cell growth and angiogenesis. (The data of the HS sequence is cited from reference [96,97,98,99]).
Figure 2. Schematic diagram of the ternary complex of FGF/HS/FGFR and the key sites for HS binding. (a) FGFR comprises extracellular Ig-like domains, intracellular tyrosine kinase domains, and transmembrane domain. Ig-like domains bind FGF with the assistance of HS to form ternary complex; (b) HS binding to FGF2 requires 2-O-S of IdoA and N-S of GlcN; HS binding to FGF1 and FGFR requires 2-O-S of IdoA and 6-O-S, N-S of GlcN; the N-S of the nonreducing terminal residue is also necessary for HS binding to FGF/FGFR. The increased expression of HS in most tumor cells enhances FGF/FGFR signal transduction, which is beneficial to tumor cell growth and angiogenesis. (The data of the HS sequence is cited from reference [96,97,98,99]).
Polymers 14 05014 g002
Table 1. Alterations or roles of GAGs/PGs in different diseases.
Table 1. Alterations or roles of GAGs/PGs in different diseases.
Pathology TypesStudy Objects (Cells/Tissues)GAGs/PGs AlterationsReferences
Atherosclerotic type IIAortaBoth the total CS content and the ratio of 6-O-S/4-O-S disaccharides in type II atherosclerosis arteries were increased[50]
AtherosclerosisArterial smooth muscle cellsTGF-β prolongs CS chains in arterial smooth muscle cells and increases versican binding to LDL.[51]
Atherosclerosis (symptomatic carotid stenosis)Iliac arteriesThe expression of perlecan gene decreased while versican gene remained unchanged.[53]
Atherosclerosis and vascularrestenosisMacrophagesSyndecan-1 protein level in macrophages was significantly decreased under hypoxia condition, and mRNA expression of key enzymes involved in HS biosynthesis in hypoxia cells was decreased. In addition, hypoxia also reduced the relative content of HS.[58]
Atherosclerosis and vascularrestenosisAortic smooth muscle cellsHA and HA synthase are increased in senescent cells. HA accumulation promotes SMC metastasis via ERK1/2 modulation of the CD44 signaling pathway, resulting in intimal hyperplasia.[61]
AtherosclerosisMacrophagesLMW-HA induces macrophage/foam cell production and promotes atherosclerosis via the PKC pathway. LMW-HA also amplifies the migration of monocytes to inflammatory atherosclerotic plaques.[65]
Breast cancerTumor tissue and plasmaPerlecan is absent in epithelial cell basement membrane while markedly upregulated in stroma. Furthermore, plasma perlecan level was significantly higher in estrogen receptor (ER)+ patients than ER- patients.[78]
Prostate cancer Perlecan expression is increased, which can the regulate sonic hedgehog signaling path.[79]
Neuroendocrine tumors (NETs)Tumor tissueSyndecan-2 is significantly increased in well-differentiated NETs and significantly decreased in poorly differentiated NETs. Glypican-5 was overexpressed in high-grade tumors with epithelial differentiation, but not in tumors with neuroendocrine phenotype.[80]
Breast cancerTumor tissueThe GAG length increased by approximately 15% in tumor tissue compared to normal tissue. Both the 6-O-S CS and the total sulfation of HS increased. Compared to nonfatal breast cancers, the sulfation degree of HS, particularly 6-O-S, was decreased in fatal breast cancers, whereas the proportion of non-sulfated disaccharides was increased.[87]
Glioblastoma multiforme (GBM)Tumor tissue60–65% of GBM tumor samples showed increased levels of CS. A 1.5-fold increase in decorin, a 3-fold increase in biglycan and a 2-fold increase in serglycin. Only decorin levels were negatively associated with overall survival in GBM patients.[115]
Pancreatic carcinomaTumor tissue The total GAG level was increased by 4 times, HA increased 12 times, CS increased 22 times, DS increased 1.5 times. A significant increase in non-sulfate and 6-sulfate disaccharides of CS.[127]
Pancreatic carcinomaTumor tissueThere are 27-fold and 7-fold increases in versican and decorin, respectively, compared with normal pancreases. The expression of 6-O-S and non-sulfated disaccharide units are enhanced.[130]
Prostate cancerTumor tissueProstate cancer has an elevated 4-O-S CS content in the ECM, which may be due to inhibited androgen receptor (AR) signaling, thus resulting in increased 4-O-sulfotransferase CHST11 expression.[131]
Gastric cancerTumor tissueCS has a 10-fold increase in 6-O-S and non-disaccharide units, while 4-O-S disaccharides were correspondingly decreased.[132]
Ovarian adenocarcinomaTumor tissueHigh expression of CS-E in the ECM of ovarian adenocarcinoma enhances vascular endothelial growth factor (VEGF) mediation.[133]
Lung cancerLewis lung carcinoma cellsThe proportion of Δ4.5HexA-GalNAc-4, 6-O-disulfate was higher in highly metastatic lung cancer cell lines than in low metastatic cell lines.[134]
Breast cancerTumor tissueHA was significantly increased in 143 tumor tissue samples, indicating that HA is directly involved in breast cancer metastasis.[143]
Breast cancerBreast tumor cells (MDA-MB-231 cells)LMW-HA activates actin filament-associated protein (AFAP-110) to bind to F-actin, resulting in nuclear translocation of myeloiddifferentiation factor (MyD88)/NF-xB and enhanced expression of pro-inflammatory cytokines IL-1β and IL-8. AFAP-110 binding with F-actin also promoted tumor cell metastasis.[149]
Diabetes mellitus 1 type (T1D)PancreasHSPGs such as Col18 and syndecan-1 showed significant loss in T1D human islets.[162]
Diabetes mellitus GAG (including perlecan) sulfation patterns affected amyloid fibril formation. The order of impact was heparin > N-desulfated acetylated heparin > fully desulfated N-sulfated heparin > fully desulfated N-acetylated heparin.[175]
Diabetic nephropathyGlomerulusHeparin sulfate 6-O-S plays an important role in extracellular matrix remodeling. Regulation of VEGFA and FGF2 signaling was achieved by increasing the expression of 6-O-endosulfatases Sulf1 and Sulf2 by the transcription factor Wilms’ Tumor 1 (WT1).[183]
Diabetic nephropathyKidneys of ratsHS has less N-S in the GBM of diabetic rats compared to the normal group.[184]
Diabetic nephropathyKidneys of ratsRenal CS/DS content decreased, accompanied by a decrease in the degree of sulfation, particularly 4, 6-O-sulfated GalN content.[187]
Diabetic nephropathyRenal cortex of diabetic db/db mice4-O-disaccharide sulfate significantly decreased from 65% to 40%, whereas 6-O-S disaccharide decreased from 11% to 6% and non-sulfated disaccharide increased from 21.5% to 51% in the renal cortex of diabetic db/db mice.[186]
T2DUrineThe contents of total GAGs, CS/DS, and HS in urine were significantly higher than those in healthy subjects.[188]
Diabetes mellitusUrine6-O-S and the 6-O-S/4-O-S ratio in the urine of diabetic patients with microalbuminuria were significantly increased compared with the healthy group.[189]
Alzheimer’s disease (AD)Brain4-O-S and 6-O-S CS are found in neurofibrillary tangles (NFTs) of AD patients, while only 4-O-S CS is found in senile plaques (SPs).[207]
Multiple sclerosisWhite matterCSPGs (versican, neurocan, and aggrecan) and DSPGs were mainly located at the edge of active plaques, while the content of CSPGs in the active center of MS plaques was significantly decreased, possibly due to the internalization of PGs in PNNs by their foam macrophages together with myelin.[208]
ADBrainPGs were more abundant in areas with amyloid plaques and neurofibrillary tangles. For example, relative to healthy individuals, the total PGs increased 1.6-fold in the AD hippocampus and 3.4-fold in the superior frontal gyrus (superior gyrus frontalis). Among them, HSPGs increased the most.[217]
ADOccipital neocortical and hippocampal tissueFibrillar Aβ plaques and nonfibrillar Aβ plaques contained high levels of N-sulfated HS, while N-sulfation was very low in nonfibrillar Aβ plaques.[218]
ADCerebral cortexThe critical sites for binding of heparin sulfate to β-amyloid (Aβ) fibrils contain 2-O-S IdoA and N-S from the human cerebral cortex, whereas binding of Aβ monomers requires 6-O-S on GlcN residues.[219]
Parkinson’s disease (PD)Neuronal cellsThe internalization of α-syn aggregates strongly depends on the cell surface HS and is associated with their total sulfation level. [224]
COVID-19 infection N-S, 2-O-S, and 6-O-S in HS and 6-O-S, 2-O-S, 3-O-S, and N-S in HP were critical for competitive binding to SARS-CoV-2 spike protein.[241]
COVID-19 infection The presence of 3-O-S in HS contributes to the recognition and binding of SARS-CoV-2 spikes in vitro.[242]
COVID-19 infection 3-O-sulfotransferase 3B overexpression and glycocalyx sulfation degree were too low to promote SARS-CoV-2 infection under pathological conditions.[243]
Idiopathic pulmonary fibrosisLungCS/DS, HA, and the CS/DS ratio increased significantly. CS/DS increases in 4-O-S, 6-O-S, and 2-O-S disaccharide units and decreases in non-sulfated disaccharides, resulting in significant increases in sulfated levels. Similarly, significant increases were observed in NS, 2-O-S, and 6-O-S disaccharides of HS, particularly the UA2S–GlcNS6S unit.[247]
AsthmaEndobronchial biopsy specimensSignificant increases in PG (versican, biglycan, and decorin) content at the lesion were observed in biopsy specimens from asthma cases.[248]
Chronic obstructive pulmonary diseaseLungHS increased significantly, and its sulfation pattern was related to the COPD stage, while CS/DS did not change significantly. The 2-O-S and NS of HS increased during the fourth phase of COPD, while 6-O-S did not change.[249]
Cystic fibrosisLungGAG expression is elevated in cystic fibrosis and abnormal sulfation of GAG can be found in bronchial epithelial cells of patients.[250,251]
Familial urolithiasisUrineThe total sulfated GAG, CS, and HS contents in urine all decreased, while the HA content was increased, and the proportion of HS in total sulfated GAGs was increased. [259]
Kidney stonesUrineBlack South Africans were found to have higher CS levels in urine than whites (kidney stones were significantly more prevalent in whites than blacks in South Africans), suggesting that higher CS levels may inhibit kidney stone formation.[260]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Wang, Q.; Chi, L. The Alterations and Roles of Glycosaminoglycans in Human Diseases. Polymers 2022, 14, 5014. https://doi.org/10.3390/polym14225014

AMA Style

Wang Q, Chi L. The Alterations and Roles of Glycosaminoglycans in Human Diseases. Polymers. 2022; 14(22):5014. https://doi.org/10.3390/polym14225014

Chicago/Turabian Style

Wang, Qingchi, and Lianli Chi. 2022. "The Alterations and Roles of Glycosaminoglycans in Human Diseases" Polymers 14, no. 22: 5014. https://doi.org/10.3390/polym14225014

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