Review Open Access
Copyright ©The Author(s) 2016. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Diabetes. Feb 25, 2016; 7(4): 67-73
Published online Feb 25, 2016. doi: 10.4239/wjd.v7.i4.67
Glycosaminoglycan remodeling during diabetes and the role of dietary factors in their modulation
Vemana Gowd, Abhignan Gurukar, Nandini D Chilkunda, Department of Molecular Nutrition, CSIR-Central Food Technological Research Institute, Mysore 570 020, India
Author contributions: All the authors have contributed by searching for relevant literature and writing of the manuscript.
Conflict-of-interest statement: The authors hereby declare that they have no conflict-of -interest to state with respect to this paper.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Nandini D Chilkunda, PhD, Department of Molecular Nutrition, CSIR-Central Food Technological Research Institute, Mysore 570 020, India. cdnandini@cftri.res.in
Telephone: +91-821-2514192
Received: October 3, 2015
Peer-review started: October 9, 2015
First decision: November 10, 2015
Revised: November 23, 2015
Accepted: December 29, 2015
Article in press: January 4, 2016
Published online: February 25, 2016

Abstract

Glycosaminoglycans (GAGs) play a significant role in various aspects of cell physiology. These are complex polymeric molecules characterized by disaccharides comprising of uronic acid and amino sugar. Compounded to the heterogeneity, these are variously sulfated and epimerized depending on the class of GAG. Among the various classes of GAG, namely, chondroitin/dermatan sulfate, heparin/heparan sulfate, keratan sulfate and hyaluronic acid (HA), only HA is non-sulfated. GAGs are known to undergo remodeling in various tissues during various pathophysiological conditions, diabetes mellitus being one among them. These changes will likely affect their structure thereby impinging on their functionality. Till date, diabetes has been shown to affect GAGs in organs such as kidney, liver, aorta, skin, erythrocytes, etc. to name a few, with deleterious consequences. One of the mainstays in the treatment of diabetes is though dietary means. Various dietary factors are known to play a significant role in regulating glucose homeostasis. Furthermore, in recent years, there has been a keen interest to decipher the role of dietary factors on GAG metabolism. This review focuses on the remodeling of GAGs in various organs during diabetes and their modulation by dietary factors. While effect of diabetes on GAG metabolism has been worked out quite a bit, studies on the role of dietary factors in their modulation has been few and far between. We have tried our best to give the latest reports available on this subject.

Key Words: Glycosaminoglycans, Diabetes, Proteoglycans, Remodeling, Dietary factors

Core tip: Glycosaminoglycans/Proteoglycans are important polymeric molecules which play important roles in cell physiology. Under pathological conditions such as diabetes, they are known to undergo remodeling affecting their structure-function relationship. This review article deals with its remodeling in various tissues and their modulation by dietary factors.



INTRODUCTION

Diabetes mellitus, henceforth referred as diabetes, is a disorder characterized by sustained hyperglycemia. According to World Health Organization studies, diabetes could be one of the leading causes of death in world population by 2030[1]. Approximately 80% of the deaths due to diabetes are reported to have occurred in low- and middle-income countries[2]. Diabetes could occur as a result of either decreased insulin secretion by pancreatic beta cells (Type 1 diabetes) or as a result of insulin resistance (Type 2 diabetes)[3]. In both these instances, glucose uptake by the cells and their disposal is affected. Insulin is required for storing glucose as glycogen in the liver and skeletal muscles. Lack of insulin secretion/insulin sensitivity, alter the glycogenolysis and gluconeogenesis results in the subsequent increase in blood glucose[4].

Sustained hyperglycemia in the long run results in the manifestation of micro- and macro-vascular complications. Nephropathy, retinopathy, and neuropathy are some of the serious secondary complications of diabetes which are known to affect kidney, eye and nerves, respectively[5]. In both micro- and macro-vascular complications, extracellular matrix components (ECM) are affected. ECM is composed of proteoglycans (PGs) and glycoproteins. PGs are complex polymeric molecules consisting of core proteins to which are attached the glycosaminoglycan (GAG) chains. They play an important role in various aspects of cell growth and behavior not only by acting as a supporting structure but also as a depot for growth factors and other signaling molecules. Changes in the structure-function relationship of GAGs/PGs impinge on their biological activities and influence the functioning of organ and the organ systems[6].

Controlling blood sugar levels will have a positive impact on the health of the organism. It can be managed by drugs, diet and exercise with positive outcomes. Diet, in particular, has been demonstrated to play a vital role in the management of diabetes. In recent years, with increasing emphasis being placed on functional foods, a lot of research efforts are being directed towards elucidating and deciphering novel bioactive molecules from dietary sources that could be used in tailoring functional foods. These foods are expected to increase the general wellness as well as attenuate the disease condition by modulating various processes.

GAGS- STRUCTURE AND BIOSYNTHESIS

GAGs are polymeric molecules that are present in all cells. There are 4 classes of GAGs known till date namely; hyaluronic acid (HA), chondroitin sulfate/dermatan sulfate (CS/DS), heparan sulfate/heparin (HS/Hep) and keratan sulfate (KS). CS/DS and HS/Hep are synthesized on a core protein through the linkage region tetrasaccharide of Xylose-Galactose-Galactose-Glucuronic acid. They contain repeating disaccharide units of uronic acid and amino sugar which are variously sulfated[7]. Disaccharides of CS/DS are glucuronic acid/iduronic acid and N-acetyl-D galactosamine and disaccharides of HS/Hep are glucuronic acid/Iduronic acid and N-acetyl-D glucosamine. The composite structure comprising of GAGs with core proteins are known as PGs. There are a few exceptions: HA which is composed of glucuronic acid and N-acetyl-D-glucosamine is not attached to the core proteins, and KS does not contain an uronic acid but contains galactose instead. KS chains are attached to core proteins via aspargine (N-linked) or serine/threonine (O-linked)[8]. All the classes of GAGs except for HA are variously sulfated.

Biosynthesis of all classes of GAGs with the exception of HA is synthesized in endoplasmic reticulum and Golgi. HA, is however synthesized by transmembrane hyaluronan synthases (HAS1, HAS2, and HAS3)[9]. Biosynthesis of GAGs is an energy-intensive process. Interestingly, one of the disaccharides, N-Acetyl-D-glucosamine is synthesized through hexosamine biosynthetic pathway that is also known as nutrient sensor pathway[10].

GAGs play vital roles in various aspects of cell physiology which brings about cell growth and development[11]. They can bring about biological activities by forming binding domains on growth factors and other proteins. GAGs undergo remodeling under various pathological conditions bringing about changes in their structure and function. In conditions of diabetes, changes in GAG structure and function have been observed in different tissues.

DIABETIC NEPHROPATHY AND GAGS

Nephropathy is one of the major secondary complications of diabetes which, in the long run, leads to end-stage renal failure. It is characterized by increased deposition of ECM components. ECM components maintain the integrity of the cell and their interactions with GAGs are essential for maintaining the extracellular morphology and cell adhesion[12]. The increase in the ECM components in kidney leads to the thickening of basement membrane and expansion of glomerular mesangial matrix, thereby affecting the filtration process. PGs/GAGs are the part of ECM components, and it is experimentally proved that PGs/GAGs are altered during diabetic nephropathy[13]. DN is marked by albuminuria and overt proteinuria at later stages. Earlier studies have implicated loss of heparan sulfate on the basement membrane as one of the reasons for leakage of proteins[13]. It was further supported by biopsies from patients with diabetes, in which structural modifications in HS GAGs was observed[14]. GAGs in the basement membrane, especially HS are known to influence the permselectivity. This however, has been discounted by injecting heparinase, an enzyme which cleaves Hep/HS, intravenously which prevented albuminuria despite a decrease in HS chains[15]. Recent evidence further show that CS along with HA play equally important role in charge selectivity of the glomerular membrane[16]. Kidney GAGs comprises about 86% HS and 14% CS/DS among sulfated GAGs in adults whereas CS comprises 75% in embryonic kidney[17]. Studies on Streptozotocin-induced diabetic rats have shown that there is significant qualitative and quantitative changes in kidney GAGs. CS/DS from kidney was shown to be altered structurally which impinged on their functionality tested in terms of binding to laminin, fibronectin and type IV collagen[18]. Further, it has been observed that heparanase, an endosulfatase is involved in the pathogenesis of DN[19].

Multiple factors have been implicated in causation of diabetic nephropathy. Advanced glycation end products (AGEs) and oxidative stress has been considered as one of the major factors[20,21]. AGEs are the heterogeneous group of molecules which are formed via Maillard reactions from the non-enzymatic reaction of reducing sugars with free amino groups of proteins, lipids and nucleic acids[22]. Accumulation of AGEs in patients with diabetic nephropathy is due to enhanced formation and decreased the clearance of AGEs[23]. AGEs tend to alter properties of large matrix proteins like laminin, collagen, fibronectin and vitronectin, through AGE-AGE intermolecular covalent bonds or crosslinking with these proteins[23]. Formation of AGEs on laminin reduced polymer elongation as well as reduced binding of laminin to type IV collagen and HSPGs[24,25]. AGEs also play a significant role in thickening of GBM and mesangial expansions that are considered to be hallmarks of diabetic nephropathy because AGEs formation on ECM proteins dysregulates their degradation by matrix metalloproteinase[26,27]. AGEs are also implicated in the increased production of TGFβ, a cytokine which is responsible for increased synthesis of ECM components in the kidney[28-30]. However, the mechanism by which GAGs/PGs are modulated in the diabetic kidney has not been deciphered.

LIVER GAGS IN DIABETES

The liver is an organ of great metabolic importance. It is rich in GAGs and harbors CS/DS, HS, HA and KS[31,32]. Changes in liver GAGs have been observed during various physiological and pathological conditions such as diabetes[33], hypercholesterolemia[34], liver cirrhosis[35], and cholestasis[36], etc., to name a few. Diabetes is known to deregulate lipid metabolism and GAGs, in particular the HS class. It plays an important role in lipoprotein metabolism in liver by acting as a receptor or a co-receptor along with LDL-receptor, LRP and ApoE[37,38]. HSPGs such as syndecan-1 and perlecan, in particular, have been implicated in lipoprotein metabolism, and it is evidenced by impairment in the clearance of remnant lipoproteins in syndecan-1 knockout mice[39]. HS-GAGs are also known to be involved in hepatic clearance of apoB-48-containing lipoproteins[40]. During experimental diabetic conditions reduced N-sulfation has been observed in liver HS GAG as compared to control[33]. This was determined to be due to decreased glucosaminyl N-deacetylase activity and N-sulfotransferase activities in hepatocytes[41]. Decreased content of liver HS GAGs as a result of decreased HSPGs was associated with the decreased postprandial clearance of apoB-48-containing lipoproteins. It has been determined that decrease in HSPG perlecan was associated with the delayed clearance of apoB-48-containing lipoproteins[40]. However, Bishop et al[42] have shown that decrease in lipoprotein clearance during diabetes is not due to changes in HS as no differences were observed between normal and diabetic littermates in liver heparan sulfate content, sulfation and syndecan-1 protein levels. Some of the degradative enzymes of HS have been observed to affect the metabolism of lipoproteins. Noted amongst them is Sulf 2 which encodes heparan sulfate glucosamine- 6-O-endosulfatase 2 which is responsible for the degradation of HSPGs by removing 6-O sulfate groups. Involvement of HSPGs in hepatic clearance was further evidenced by the deletion of SULF2 in cultures hepatocytes. Knockdown of SULF2 showcased the increased HSPG-mediated catabolism of remnant lipoproteins in cultured cells[43].

Matrix PGs in the liver are affected by insulin and fatty acids. In a study conducted by Olsson et al[44], it was observed that insulin and non-esterified fatty acids modulate PG synthesis in hepatic cells so much so that the changes in PG composition affected their binding to remnant B-VLDL particles contributing to dyslipidemia of insulin resistance.

EFFECT OF DIABETES ON AORTIC GAGS

Cardio-vascular disease is one of the major complications of diabetes. Arterial walls are rich in PGs and are implicated in the pathogenesis of atherosclerosis by virtue of their ability to bind and trap LDL. In a study carried out in diabetic monkeys, it was observed that diabetes resulted in increased DS class of GAGs in arteries which was positively correlated with tissue cholesterol promoting atherosclerosis[45]. Three major CS/DSPGs present in the arterial wall are versican, decorin and biglycan[46]. In aortic endothelial cells, high glucose condition resulted in decreased perlecan level indicating remodeling of PGs[47]. Studies on BAEC suggested that reduced sulfate incorporation in the HSPGs[48].

Factors affecting the synthesis and degradation of PGs in the aorta as a result of diabetes have not been critically studied. TGFβ has been one of the factors implicated in changes in PG synthesis. It is known to be produced in hyperglycemic conditions and induces changes in PGs secreted by vascular smooth muscle cells increasing their propensity to retain and bind lipoproteins in the vascular wall[49].

EFFECT OF DIABETES ON ERYTHROCYTE GAGS

Diabetes is known to affect erythrocytes by increasing their aggregation binding to endothelial cells and decreasing the deformability[50]. Diabetes is also associated with the increase in membrane lipids and changes in its fluidity. Reports on erythrocyte GAGs are scarce. HS expression was observed in human erythrocytes infected with the malarial parasite which helped in the rosette formation with uninfected erythrocytes[51]. Furthermore, HS was found to be a mediator for the binding of Plasmodium falciparum-infected erythrocytes to endothelial cells via the DBL1α domain of PfEMP[52]. Recent findings from our laboratory revealed the presence of CS/DS class of GAGs in erythrocytes of experimentally-induced diabetic rats. Erythrocytes from diabetic rats had increased levels of CS/DS when compared to age-matched non-diabetic control rats. They appeared to mediate the binding of erythrocytes to ECM[53]. Erythrocytes isolated from rats that were diabetic as well as hypercholesterolaemic showed higher binding to ECM components than that isolated from diabetic rats[54]. Further work on synthesis and regulation of GAGs in erythrocytes should be able to throw light on the function of these important molecules.

GAGS IN DIABETIC RETINOPATHY

Retinopathy is one of the secondary complications of diabetes. Diabetic retinopathy leads to vision loss with their associated abnormalities in vascular permeability[55]. Changes in the vascular permeability are associated with combination of abnormalities namely: Thickening of basement membrane, leakage of various compounds, capillary occlusion, and formation of new vessels along with fibrous tissue[56-58]. Fluorescent microscopic studies have found the presence of HS, CS/DS, HA throughout the retina but the presence of KS is found to be limited to the sclera[59]. Studies on the incorporation of [35S]- and [14C] glucosamine into GAGs that were isolated from retinal vessel basement membrane suggested that HS GAG is the major GAG present in basement membrane[60]. Various studies have been conducted to decipher the role of HS GAG in retinopathy. A study on the metabolism of GAGs in retina in streptozotocin-induced diabetic rats found decreased synthesis of HSPGs and was found to be associated with the decreased expression of perlecan[61]. It has also been demonstrated that quantitatively more GAGs is found in tears of patients with diabetic retinopathy than in non-diabetic people[62]. Furthermore, diabetic retinopathy was associated with the reduced production of HS GAG in the vitreous and increased expression of surface binding exogenous VEGF[63]. It is well known that HS GAGs are recognized as a co-receptor for fibroblast growth factor. FGF is a potent endothelial cell mitogen that has proposed to be involved in the development of proliferative diabetic retinopathy. Changes in the distribution of FGF during diabetes are associated with the development of retinopathy and retinal neovascularization. These studies reveal the possible role of FGF in the development of neovascularization and contribution of HSPGs in it[64]. In a study conducted on diabetic subjects, it was observed that there was a correlation between diabetic retinopathy, erythrocyte anionic charge and urinary GAG excretion[65].

DIABETES AND SKIN GAGS

Despite the fact that skin is rich in GAGs, not much work has been carried out to determine the effect of diabetes on its remodeling. The skin of STZ-induced diabetic rats showed decreased GAG content as a result of decreased GAG biosynthesis[66]. The decrease of GAG content in the skin of diabetic rats was earlier reported to be as a result of decreased circulating IGF-I level, increased plasma content of LMW-BPs and increased proteolytic activity of the skin[66].

ROLE OF DIETARY FACTORS IN MODULATION OF GAGs

Diet plays major roles in the management of diabetes. The information with respect to the role of diet on GAG metabolism is scanty. In a study conducted by Taylor et al[67], it was observed that extracts from some of the plants present in Amazon rain forest stimulated GAG assembly in both wild type and mutant cell line defect in one of the key biosynthetic enzymes-xylosyltransferase. These findings suggest the importance of plant products in modulation of GAGs in animal cells. Various dietary factors have been implicated in attenuating diabetic nephropathy per se, but there are very few reports on their effect on GAG metabolism. Fiber-rich sources such as wheat bran and Guar gum altered decreased GAG synthesis in STZ-induced diabetic rats[68]. In another study, feeding of bitter gourd (Momordica charantia LINN) resulted in amelioration of decreased synthesis of GAGs in the kidney of diabetic rats[69]. In a similar vein, dietary feeding of Tinospora cordifolia resulted in attenuation of decreased CS/DS in STZ-induced diabetic rat kidney[70]. Not only in diabetes, even in normal conditions are some of the dietary factors known to affect GAGs. Noted among them, Genistein has been determined to decrease synthesis of GAGs[71]. Dietary manganese has been found to affect aortic GAGs in rats by altering composition and sulfation pattern of heparan sulfate GAG[72]. Similarly, wild blue berry consumption altered the GAG composition in the aorta[73]. In a study conducted, Annona squamosa showed beneficial effect on wound healing by increasing the synthesis of GAGs and collagen in STZ-induced diabetic rats[74].

GAG consumption can also occur by consuming foods of animal origin[75]. However, not much work has been carried out with respect to its metabolic effects in the body. The mixture of GAGs, called sulodexide has been demonstrated to contain proteinuria and ameliorate markers of diabetic nephropathy in clinical studies[76]. It has also been observed that oral administration of high molecular weight hyaluronan controls immune system via Toll-like receptor 4 in the intestinal epithelium[77].

FUTURE PERSPECTIVES

Despite the fact that rapid strides have been made with respect to deciphering the importance of GAGs in health and disease of the organism, more needs to be done especially with relation to their regulation under various conditions both normal and pathological. Also, evaluating various dietary molecules which could influence GAG metabolism will go a long way in therapeutic applications and development of functional foods.

Footnotes

P- Reviewer: Gómez-Sáez J S- Editor: Wang JL L- Editor: A E- Editor: Jiao XK

References
1.  Mathers CD, Loncar D. Projections of global mortality and burden of disease from 2002 to 2030. PLoS Med. 2006;3:e442.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Klafke A, Duncan BB, Stevens A, Rosa Rdos S, de Moura L, Malta D, Schmidt MI. The decline in mortality due to acute complications of diabetes mellitus in Brazil, 1991-2010. BMC Public Health. 2015;15:772.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Farag YM, Gaballa MR. Diabesity: an overview of a rising epidemic. Nephrol Dial Transplant. 2011;26:28-35.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Edgerton DS, Cardin S, Emshwiller M, Neal D, Chandramouli V, Schumann WC, Landau BR, Rossetti L, Cherrington AD. Small increases in insulin inhibit hepatic glucose production solely caused by an effect on glycogen metabolism. Diabetes. 2001;50:1872-1882.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  King P, Peacock I, Donnelly R. The UK prospective diabetes study (UKPDS): clinical and therapeutic implications for type 2 diabetes. Br J Clin Pharmacol. 1999;48:643-648.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Bülow HE, Hobert O. The molecular diversity of glycosaminoglycans shapes animal development. Annu Rev Cell Dev Biol. 2006;22:375-407.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Sugahara K, Kitagawa H. Recent advances in the study of the biosynthesis and functions of sulfated glycosaminoglycans. Curr Opin Struct Biol. 2000;10:518-527.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Funderburgh JL. Keratan sulfate: structure, biosynthesis, and function. Glycobiology. 2000;10:951-958.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Lee JY, Spicer AP. Hyaluronan: a multifunctional, megaDalton, stealth molecule. Curr Opin Cell Biol. 2000;12:581-586.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Zachara NE, Hart GW. O-GlcNAc a sensor of cellular state: the role of nucleocytoplasmic glycosylation in modulating cellular function in response to nutrition and stress. Biochim Biophys Acta. 2004;1673:13-28.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Bishop JR, Schuksz M, Esko JD. Heparan sulphate proteoglycans fine-tune mammalian physiology. Nature. 2007;446:1030-1037.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Barkalow FJ, Schwarzbauer JE. Interactions between fibronectin and chondroitin sulfate are modulated by molecular context. J Biol Chem. 1994;269:3957-3962.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Lewis EJ, Xu X. Abnormal glomerular permeability characteristics in diabetic nephropathy: implications for the therapeutic use of low-molecular weight heparin. Diabetes Care. 2008;31 Suppl 2:S202-S207.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Yard BA, Kahlert S, Engelleiter R, Resch S, Waldherr R, Groffen AJ, van den Heuvel LP, van der Born J, Berden JH, Kröger S. Decreased glomerular expression of agrin in diabetic nephropathy and podocytes, cultured in high glucose medium. Exp Nephrol. 2001;9:214-222.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Wijnhoven TJ, Lensen JF, Wismans RG, Lefeber DJ, Rops AL, van der Vlag J, Berden JH, van den Heuvel LP, van Kuppevelt TH. Removal of heparan sulfate from the glomerular basement membrane blocks protein passage. J Am Soc Nephrol. 2007;18:3119-3127.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Jeansson M, Haraldsson B. Glomerular size and charge selectivity in the mouse after exposure to glucosaminoglycan-degrading enzymes. J Am Soc Nephrol. 2003;14:1756-1765.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Steer DL, Shah MM, Bush KT, Stuart RO, Sampogna RV, Meyer TN, Schwesinger C, Bai X, Esko JD, Nigam SK. Regulation of ureteric bud branching morphogenesis by sulfated proteoglycans in the developing kidney. Dev Biol. 2004;272:310-327.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Joladarashi D, Salimath PV, Chilkunda ND. 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.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Maxhimer JB, Somenek M, Rao G, Pesce CE, Baldwin D, Gattuso P, Schwartz MM, Lewis EJ, Prinz RA, Xu X. Heparanase-1 gene expression and regulation by high glucose in renal epithelial cells: a potential role in the pathogenesis of proteinuria in diabetic patients. Diabetes. 2005;54:2172-2178.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Yamagishi S, Matsui T. Advanced glycation end products, oxidative stress and diabetic nephropathy. Oxid Med Cell Longev. 2010;3:101-108.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Thomas MC, Forbes JM, Cooper ME. Advanced glycation end products and diabetic nephropathy. Am J Ther. 2005;12:562-572.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Vlassara H, Palace MR. Diabetes and advanced glycation endproducts. J Intern Med. 2002;251:87-101.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Oleniuc M, Secara I, Onofriescu M, Hogas S, Voroneanu L, Siriopol D, Covic A. Consequences of Advanced Glycation End Products Accumulation in Chronic Kidney Disease and Clinical Usefulness of Their Assessment Using a Non-invasive Technique - Skin Autofluorescence. Maedica (Buchar). 2011;6:298-307.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Charonis AS, Reger LA, Dege JE, Kouzi-Koliakos K, Furcht LT, Wohlhueter RM, Tsilibary EC. Laminin alterations after in vitro nonenzymatic glycosylation. Diabetes. 1990;39:807-814.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Charonis AS, Tsilbary EC. Structural and functional changes of laminin and type IV collagen after nonenzymatic glycation. Diabetes. 1992;41 Suppl 2:49-51.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Krishnamurti U, Rondeau E, Sraer JD, Michael AF, Tsilibary EC. Alterations in human glomerular epithelial cells interacting with nonenzymatically glycosylated matrix. J Biol Chem. 1997;272:27966-27970.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Ishibashi Y, Yamagishi S, Matsui T, Ohta K, Tanoue R, Takeuchi M, Ueda S, Nakamura K, Okuda S. Pravastatin inhibits advanced glycation end products (AGEs)-induced proximal tubular cell apoptosis and injury by reducing receptor for AGEs (RAGE) level. Metabolism. 2012;61:1067-1072.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Border WA, Noble NA. Transforming growth factor beta in tissue fibrosis. N Engl J Med. 1994;331:1286-1292.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Ling H, Vamvakas S, Busch G, Dämmrich J, Schramm L, Lang F, Heidland A. Suppressing role of transforming growth factor-beta 1 on cathepsin activity in cultured kidney tubule cells. Am J Physiol. 1995;269:F911-F917.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Xiang G, Schinzel R, Simm A, Münch G, Sebekova K, Kasper M, Niwa T, Schmitz C, Heidland A. Advanced glycation end products (AGEs)-induced expression of TGF-beta 1 is suppressed by a protease in the tubule cell line LLC-PK1. Nephrol Dial Transplant. 2001;16:1562-1569.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Minami R, Ikeno T, Igarashi C, Tsugawa S, Nakao T. Characterization of keratan sulfate isolated from liver affected by Morquio syndrome. Tohoku J Exp Med. 1983;139:321-326.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Gressner AM, Vasel A. Developmental changes of proteoglycan synthesis in rat liver and isolated hepatocytes. Mech Ageing Dev. 1985;31:307-327.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Kjellén L, Bielefeld D, Hook M. Reduced sulfation of liver heparan sulfate in experimentally diabetic rats. Diabetes. 1983;32:337-342.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  MacArthur JM, Bishop JR, Stanford KI, Wang L, Bensadoun A, Witztum JL, Esko JD. Liver heparan sulfate proteoglycans mediate clearance of triglyceride-rich lipoproteins independently of LDL receptor family members. J Clin Invest. 2007;117:153-164.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Tátrai P, Egedi K, Somorácz A, van Kuppevelt TH, Ten Dam G, Lyon M, Deakin JA, Kiss A, Schaff Z, Kovalszky I. Quantitative and qualitative alterations of heparan sulfate in fibrogenic liver diseases and hepatocellular cancer. J Histochem Cytochem. 2010;58:429-441.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Guedes PL, Castañon MC, Nagaoka MR, Aguiar JA. Increase of glycosaminoglycans and metalloproteinases 2 and 9 in liver extracellular matrix on early stages of extrahepatic cholestasis. Arq Gastroenterol. 2014;51:309-315.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  de Beer F, Hendriks WL, van Vark LC, Kamerling SW, van Dijk KW, Hofker MH, Smelt AH, Havekes LM. Binding of beta-VLDL to heparan sulfate proteoglycans requires lipoprotein lipase, whereas ApoE only modulates binding affinity. Arterioscler Thromb Vasc Biol. 1999;19:633-637.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Williams KJ, Fless GM, Petrie KA, Snyder ML, Brocia RW, Swenson TL. Mechanisms by which lipoprotein lipase alters cellular metabolism of lipoprotein(a), low density lipoprotein, and nascent lipoproteins. Roles for low density lipoprotein receptors and heparan sulfate proteoglycans. J Biol Chem. 1992;267:13284-13292.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Stanford KI, Bishop JR, Foley EM, Gonzales JC, Niesman IR, Witztum JL, Esko JD. Syndecan-1 is the primary heparan sulfate proteoglycan mediating hepatic clearance of triglyceride-rich lipoproteins in mice. J Clin Invest. 2009;119:3236-3245.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Ebara T, Conde K, Kako Y, Liu Y, Xu Y, Ramakrishnan R, Goldberg IJ, Shachter NS. Delayed catabolism of apoB-48 lipoproteins due to decreased heparan sulfate proteoglycan production in diabetic mice. J Clin Invest. 2000;105:1807-1818.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Unger E, Pettersson I, Eriksson UJ, Lindahl U, Kjellén L. Decreased activity of the heparan sulfate-modifying enzyme glucosaminyl N-deacetylase in hepatocytes from streptozotocin-diabetic rats. J Biol Chem. 1991;266:8671-8674.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Bishop JR, Foley E, Lawrence R, Esko JD. Insulin-dependent diabetes mellitus in mice does not alter liver heparan sulfate. J Biol Chem. 2010;285:14658-14662.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Chen K, Liu ML, Schaffer L, Li M, Boden G, Wu X, Williams KJ. Type 2 diabetes in mice induces hepatic overexpression of sulfatase 2, a novel factor that suppresses uptake of remnant lipoproteins. Hepatology. 2010;52:1957-1967.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Olsson U, Egnell AC, Lee MR, Lundén GO, Lorentzon M, Salmivirta M, Bondjers G, Camejo G. Changes in matrix proteoglycans induced by insulin and fatty acids in hepatic cells may contribute to dyslipidemia of insulin resistance. Diabetes. 2001;50:2126-2132.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Edwards IJ, Wagner JD, Vogl-Willis CA, Litwak KN, Cefalu WT. Arterial heparan sulfate is negatively associated with hyperglycemia and atherosclerosis in diabetic monkeys. Cardiovasc Diabetol. 2004;3:6.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Williams KJ. Arterial wall chondroitin sulfate proteoglycans: diverse molecules with distinct roles in lipoprotein retention and atherogenesis. Curr Opin Lipidol. 2001;12:477-487.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Vogl-Willis CA, Edwards IJ. High-glucose-induced structural changes in the heparan sulfate proteoglycan, perlecan, of cultured human aortic endothelial cells. Biochim Biophys Acta. 2004;1672:36-45.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Humphries DE, Silbert CK, Silbert JE. Glycosaminoglycan production by bovine aortic endothelial cells cultured in sulfate-depleted medium. J Biol Chem. 1986;261:9122-9127.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Yang SN, Burch ML, Tannock LR, Evanko S, Osman N, Little PJ. Transforming growth factor-β regulation of proteoglycan synthesis in vascular smooth muscle: contribution to lipid binding and accelerated atherosclerosis in diabetes. J Diabetes. 2010;2:233-242.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Yedgar S, Koshkaryev A, Barshtein G. The red blood cell in vascular occlusion. Pathophysiol Haemost Thromb. 2002;32:263-268.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Vogt AM, Winter G, Wahlgren M, Spillmann D. Heparan sulphate identified on human erythrocytes: a Plasmodium falciparum receptor. Biochem J. 2004;381:593-597.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Vogt AM, Barragan A, Chen Q, Kironde F, Spillmann D, Wahlgren M. Heparan sulfate on endothelial cells mediates the binding of Plasmodium falciparum-infected erythrocytes via the DBL1alpha domain of PfEMP1. Blood. 2003;101:2405-2411.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Srikanth CB, Salimath PV, Nandini CD. 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.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Gowd V, Nandini CD. Erythrocytes in the combined milieu of high glucose and high cholesterol shows glycosaminoglycan-dependent cytoadherence to extracellular matrix components. Int J Biol Macromol. 2015;73:182-188.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Scheppke L, Aguilar E, Gariano RF, Jacobson R, Hood J, Doukas J, Cao J, Noronha G, Yee S, Weis S. Retinal vascular permeability suppression by topical application of a novel VEGFR2/Src kinase inhibitor in mice and rabbits. J Clin Invest. 2008;118:2337-2346.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Ljubimov AV, Burgeson RE, Butkowski RJ, Couchman JR, Zardi L, Ninomiya Y, Sado Y, Huang ZS, Nesburn AB, Kenney MC. Basement membrane abnormalities in human eyes with diabetic retinopathy. J Histochem Cytochem. 1996;44:1469-1479.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Conde-Knape K. Heparan sulfate proteoglycans in experimental models of diabetes: a role for perlecan in diabetes complications. Diabetes Metab Res Rev. 2001;17:412-421.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Engerman RL, Kern TS. Experimental galactosemia produces diabetic-like retinopathy. Diabetes. 1984;33:97-100.  [PubMed]  [DOI]  [Cited in This Article: ]
59.  Clark SJ, Keenan TD, Fielder HL, Collinson LJ, Holley RJ, Merry CL, van Kuppevelt TH, Day AJ, Bishop PN. Mapping the differential distribution of glycosaminoglycans in the adult human retina, choroid, and sclera. Invest Ophthalmol Vis Sci. 2011;52:6511-6521.  [PubMed]  [DOI]  [Cited in This Article: ]
60.  Cohen MP, Ciborowski CJ. Presence of glycosaminoglycans in retinal capillary basement membrane. Biochim Biophys Acta. 1981;674:400-406.  [PubMed]  [DOI]  [Cited in This Article: ]
61.  Bollineni JS, Alluru I, Reddi AS. Heparan sulfate proteoglycan synthesis and its expression are decreased in the retina of diabetic rats. Curr Eye Res. 1997;16:127-130.  [PubMed]  [DOI]  [Cited in This Article: ]
62.  Moschos MM, Rouvas AA, Papadimitriou S, Kotsolis A, Sitaras N, Apostolopoulos M. Quantitative determination of glycosaminoglycans in tears of diabetic patients. Clin Ophthalmol. 2008;2:581-584.  [PubMed]  [DOI]  [Cited in This Article: ]
63.  Nishiguchi KM, Kataoka K, Kachi S, Komeima K, Terasaki H. Regulation of pathologic retinal angiogenesis in mice and inhibition of VEGF-VEGFR2 binding by soluble heparan sulfate. PLoS One. 2010;5:e13493.  [PubMed]  [DOI]  [Cited in This Article: ]
64.  Murakami M, Simons M. Fibroblast growth factor regulation of neovascularization. Curr Opin Hematol. 2008;15:215-220.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  Yenice O, Kazokoğlu H, Ozcan E, Yüksel M, Adigüzel G, Haklar G, Yavuz DG. Erythrocyte Membrane Anionic Content and Urinary Glycosaminoglycan Excretion in Type 1 Diabetes: Association with Retinopathy. Curr Eye Res. 2006;31:975-981.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Cechowska-Pasko M, Pałka J, Bańkowski E. Decrease in the glycosaminoglycan content in the skin of diabetic rats. The role of IGF-I, IGF-binding proteins and proteolytic activity. Mol Cell Biochem. 1996;154:1-8.  [PubMed]  [DOI]  [Cited in This Article: ]
67.  Taylor WH, Sinha A, Khan IA, McDaniel ST, Esko JD. Primers of glycosaminoglycan biosynthesis from Peruvian rain forest plants. J Biol Chem. 1998;273:22260-22266.  [PubMed]  [DOI]  [Cited in This Article: ]
68.  Nandini CD, Sambaiah K, Salimath PV. Dietary fibres ameliorate decreased synthesis of heparan sulphate in streptozotocin induced diabetic rats. J Nutr Biochem. 2003;14:203-210.  [PubMed]  [DOI]  [Cited in This Article: ]
69.  Kumar GS, Shetty AK, Salimath PV. Modulatory effect of bitter gourd (Momordica charantia LINN.) on alterations in kidney heparan sulfate in streptozotocin-induced diabetic rats. J Ethnopharmacol. 2008;115:276-283.  [PubMed]  [DOI]  [Cited in This Article: ]
70.  Joladarashi D, Chilkunda ND, Salimath PV. Tinospora cordifolia consumption ameliorates changes in kidney chondroitin sulphate/dermatan sulphate in diabetic rats. J Nutr Sci. 2012;1:e7.  [PubMed]  [DOI]  [Cited in This Article: ]
71.  Piotrowska E, Jakóbkiewicz-Banecka J, Barańska S, Tylki-Szymańska A, Czartoryska B, Wegrzyn A, Wegrzyn G. Genistein-mediated inhibition of glycosaminoglycan synthesis as a basis for gene expression-targeted isoflavone therapy for mucopolysaccharidoses. Eur J Hum Genet. 2006;14:846-852.  [PubMed]  [DOI]  [Cited in This Article: ]
72.  Kalea AZ, Lamari FN, Theocharis AD, Schuschke DA, Karamanos NK, Klimis-Zacas DJ. Dietary manganese affects the concentration, composition and sulfation pattern of heparan sulfate glycosaminoglycans in Sprague-Dawley rat aorta. Biometals. 2006;19:535-546.  [PubMed]  [DOI]  [Cited in This Article: ]
73.  Kalea AZ, Lamari FN, Theocharis AD, Cordopatis P, Schuschke DA, Karamanos NK, Klimis-Zacas DJ. Wild blueberry (Vaccinium angustifolium) consumption affects the composition and structure of glycosaminoglycans in Sprague-Dawley rat aorta. J Nutr Biochem. 2006;17:109-116.  [PubMed]  [DOI]  [Cited in This Article: ]
74.  Ponrasu T, Suguna L. Efficacy of Annona squamosa L in the synthesis of glycosaminoglycans and collagen during wound repair in streptozotocin induced diabetic rats. Biomed Res Int. 2014;2014:124352.  [PubMed]  [DOI]  [Cited in This Article: ]
75.  Cilla A, Olivares M, Laparra JM. Glycosaminoglycans from Animal Tissue Foods and Gut Health. Food Rev Int. 2013;29:192-200.  [PubMed]  [DOI]  [Cited in This Article: ]
76.  Lewis EJ, Lewis JB, Greene T, Hunsicker LG, Berl T, Pohl MA, de Zeeuw D, Heerspink HL, Rohde RD, Atkins RC. Sulodexide for kidney protection in type 2 diabetes patients with microalbuminuria: a randomized controlled trial. Am J Kidney Dis. 2011;58:729-736.  [PubMed]  [DOI]  [Cited in This Article: ]
77.  Asari A, Kanemitsu T, Kurihara H. Oral administration of high molecular weight hyaluronan (900 kDa) controls immune system via Toll-like receptor 4 in the intestinal epithelium. J Biol Chem. 2010;285:24751-24758.  [PubMed]  [DOI]  [Cited in This Article: ]