Abstract
Chondroitin sulfate (CS), a glycosaminoglycan (GAG) member, is a partially sulfated linear polysaccharide. CS covalently binds to core proteins to form chondroitin sulfate proteoglycans (CSPGs), which are ubiquitously distributed on the cell surface and within the extracellular matrix. Over the past two decades, CS/CSPG products derived from natural sources, such as bovine, porcine, and salmon, have garnered increasing attention worldwide as promising agents for cosmetics, pharmaceuticals, and nutraceuticals. This review explores the characteristics of salmon cartilage proteoglycan (sPG) in pharmaceutical and nutraceutical oral administration compared with other CS/CSPGs. sPG can be easily extracted as a safe, functional anti-inflammatory and analgesic agent. sPG and other CS/CSPGs are translocated outside the gastrointestinal tract after oral intake. However, the translocated amount is minimal, and the underlying mechanism remains unclear. In addition to the conventional narrative that CSPGs exert their effects after absorption or translocation, this review discusses the potential behavior of CS/CSPGs that are not absorbed and remain in the gastrointestinal tract. The interaction activity of the CS chain and regulatory effects of sPG on intestinal bacterial flora suggest the potential luminacoid nature of CS/CSPGs. These insights improve our understanding of the functionality of exogenous CSPGs and expand their potential therapeutic applications.
1. Introduction
1.1 Structure and function of chondroitin sulfate (CS)/chondroitin sulfate proteoglycans (CSPG)
Glycosaminoglycans (GAGs) are linear polysaccharides comprising repeating disaccharide units. Each disaccharide unit consists of an amino sugar moiety (hexosamines, such as D-glucosamine and D-galactosamine) alternately paired with a uronic acid moiety (D-glucuronic or L-iduronic acid). GAGs are classified into four major groups based on their disaccharide components: hyaluronic acid (HA), CS, heparan sulfate (HS), and keratan sulfate (KS). Except for HA, all GAGs covalently bind to core proteins to form diverse proteoglycans (PGs). Endogenous PGs are widely distributed on the cell surface and within the extracellular matrix (ECM) of all cell types, where they cooperate with other glycoconjugates to form the glycocalyx. Many PGs play critical roles in various biological and pathophysiological processes.
CS is the most abundant GAG found in the human body. The disaccharide unit of CS consists of N-acetyl-D-galactosamine (GalNAc) as hexosamine and D-glucuronic acid (GlcA) or L-iduronic acid (IdoA) as uronic acid[1, 2]. IdoA is synthesized through the epimerization of GlcA, a reaction catalyzed by the dermatan sulfate epimerase enzyme family. When IdoA is present at a proportion substantially greater than that of GlcA, the compound is referred to as dermatan sulfate (DS). However, CS and DS regions often coexist within a single chain, referred to as CS/DS. Mizumoto and Yamada provided a detailed discussion on the relationship between CS and DS, and the biological significance of DS [3]. The sulfate groups are randomly esterified on the hydroxyl groups of the CS/DS chains, resulting in various classes, such as GlcA-GalNAc4S (CS-A), GlcA-GalNAc6S (CS-C), GlcA2S-GalNAc6S (CS-D), GlcA-GalNAc4/6S (CS-E), and non-sulfated GlcA-GalNAc (CS-O). The CS structure exhibits considerable heterogeneity owing to the diverse arrangements of these disaccharide units.
The biological functions of CS are strongly associated with its structure, particularly the sulfation pattern. In neurophysiology, a substantial amount of CS exists in the central nervous system as a covalently bound proteoglycan (CSPG) moiety. Although disulfated disaccharides, such as CS-D and CS-E, constitute only a minor proportion, they have notable functional significance [4, 5]. Sakamoto et al. [6] demonstrated that only CS-E residue-rich short stretches, rather than other CS domains, bind to receptor-type tyrosine phosphatase S (PTPRσ) in competition with HS. This binding disrupts autophagic flux, leading to the formation of dystrophic endoballs and the inhibition of axon regeneration. In contrast, Shida et al. [7] reported that CS-D residues facilitated neurite extension via integrin αVβ3-mediated signal transduction, whereas CS-C residues did not exhibit this effect. These findings indicate that the biological activity of CS is highly dependent on its sulfation pattern.
1.2 Exogenous CS/CSPG usage (cosmetic and oral agents)
In addition to the accumulating studies on endogenous CS and CSPG (CS/CSPG), the exogenous use of CS/CSPG has attracted increasing attention. Exogenous CS/CSPG use ranges from cosmetics to pharmaceuticals and supplements. As a cosmetic agent, CS/CSPG is believed to have moisturizing effects and control skin conditions. The precise biological mechanism underlying these effects remains unclear; however, the water-retention properties of CS may have moisturizing effects. The epidermal growth factor (EGF)-like region in the C-terminal G3 domain of aggrecan and versican has been suggested to act as an EGF ligand [8, 9], and skin maintenance effects, including wound healing, may be due to this activity. In addition, CS/CSPG may have an anti-ultraviolet (UV) effect [10, 11], which may be due to its antioxidant properties. As an orally administered agent, including pharmaceutical products and nutraceutical supplements, CS/CSPG is expected to have analgesic and anti-inflammatory effects against joint pain caused by conditions, such as osteoarthritis (OA). In Europe and the USA, CS is authorized as a pharmaceutical product and a symptomatic slow-acting drug for OA (SYSADOA) [12, 13, 14, 15, 16, 17, 18]. CS has been marketed as a nutraceutical supplement in other countries, particularly Japan, claiming similar effects.
However, the precise mechanisms and behaviors of CS/CSPG after oral administration remain unclear. At least some absorption or translocation of CS is expected to occur in the proximal small intestine [19]. CS chains generally have a high molecular weight, therefore, it is hypothesized that the paracellular, rather than the transcellular pathway, is dominant in their transport by epithelial cells [19, 20]. However, the extent of the translocation is limited. In addition, the contribution of intestinal bacterial flora is related to CS behavior.
1.3 Brief characteristics of salmon nasal cartilage proteoglycan (sPG) and overview of this review
sPG is the major CSPG product marketed in Japan. The CS used in laboratory studies is mainly derived from mammals, such as bovine, porcine, and whale. These materials have problems with religious restrictions and serious infectious diseases (e.g., bovine spongiform encephalopathy); however, using salmon can avoid these problems. Salmon has a long history of consumption worldwide, which ensures its safety for oral consumption. Salmon nasal cartilage is generally discarded. However, in Japan, it is referred to as hizu, a traditional food. Majima et al. developed a method for extracting proteoglycans from salmon (Oncorhyncus keta) nasal cartilage using acetic acid at a concentration similar to that of vinegar [21]. This method is extremely simple compared with previous proteoglycan extraction methods, which are complex and involve multiple steps. It was the first in the world to achieve the mass production of proteoglycans. Acetic acid extracts from salmon nasal cartilage are composed mainly of aggrecan, a representative CSPG [22]. Kakizaki et al. [23] obtained microscopic images of the molecules using atomic force microscopy (AFM), clearly showing the test-tube brush-like structural characteristics of aggrecan.
Notably, sPG is the only proteoglycan administered to animals and cells as a CSPG, rather than as a CS single chain [24]. As mentioned above, sPG has a typical proteoglycan-like conformation. This conformation and densely packed nature of CS may produce results different from those obtained when CS is administered alone. Anti-inflammatory, cell proliferation, anti-angiogenesis, anti-arthritic, anti-diabetic, and wound-healing effects have been reported [10, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35]. However, detailed molecular mechanisms underlying these effects remain unclear. When administered orally, the same absorption or translocation mechanisms as those of the other CS/CSPG products mentioned above remain unresolved.
In this review, the effects and mechanisms of sPG administration reported to date are summarized and compared with those of CS/CSPG obtained from non-salmon sources, and the mechanisms underlying these effects are discussed. In addition, the possible but uninvestigated effects of orally administered sPG are discussed based on information on the structure of sPG and the physiological activity of other CS/CSPG studies. In particular, based on the fact that CS/CSPG is a non-digestible component that reaches the distal section of the intestine without the influence of digestive enzymes, and interaction of CS/CSPG with membrane and ECM proteins known to exist in the intestinal lumen, the possibility that CS/CSPG exhibits luminacoid behavior.
2. Structural characteristics of salmon nasal cartilage proteoglycan
Salmon nasal cartilage proteoglycans were derived by an extraction method using a low concentration of acetic acid [21]. The fractions extracted by this method contained aggrecan and epiphycan [23, 36]. Notably, sPG are often a mixture of proteoglycan species. Aggrecan is a member of the hyalectan family, and the N-terminal G1 domain of these proteoglycans has a common non-covalent hyaluronic acid-binding nature [37, 38, 39]. The binding of the HA and hyalectans is stabilized by link proteins, and lower pH conditions disrupt this binding because the electric dissociation of the carboxyl groups on HA is a cue for binding. Therefore, the acetic acid extraction method is considered to be a method to release proteoglycans into solution by inhibiting the association of HA, hyalectan, and a link protein.
The molecular weight of sPG was determined to be 344,000 using SDS-PAGE [22]. This value is small for cartilage proteoglycans. AFM images obtained by Kakizaki et al. [23] show a small test-tube-brush-like molecule. The disaccharide composition of the CS chain on sPG was CSA/C/D/O=28/58/7/8.6 [21, 22, 23]. This CS-C-rich and CS-D-containing composition is similar to that of shark-fin cartilage CS. In contrast to monosulfated disaccharide units, which are often having no notable insignificant, disulfated disaccharide units regulate the activity of the CS chain [7, 40]. The CS-D units in the CS chain of sPG are also crucial for proteoglycan function. In addition, aggrecan in sPG is free of KS [22]. KS contamination is a frequent quality control issue in CS products worldwide and [12, 18, 41 42, 43, 44, 45, 46, 47] may be avoided in sPG products. sPG have been studied in many fields. See Table 1 for a summary of previous sPG studies.
Table 1: Research on the effects of salmon nasal cartilage proteoglycan
Species |
Dosage |
Oral Admin-istration |
Results |
Ref. |
Anti-inflammatory effects |
Mice (RAW264.7 cell) |
50–500 µg/well |
- |
Heat-killed E. coli cells-induced TNF-α production was suppressed. STAT3 phosphorylation and expression were enhanced. |
[27] |
Rats (Wistar rats) |
1% PG, 1.5 mL |
Yes |
Clinical symptoms of experimental colitis and leukocyte number were improved. Total SCFA and n-butyrate in feces were enhanced. |
[48] |
Mice (C57BL/6) |
0.08–2 mg/mL 100 µL/day, 30 days |
Yes |
Clinical/histological severity of experimental autoimmune encephalomyelitis was improved. Foxp3 expression in CD4+CD25+ cells was enhanced. |
[30] |
Mice (C57BL/6) |
1 mg/day |
Yes |
Papain-induced allergic response was suppressed. |
[31] |
Anti-arthritis effect |
Human |
10 mg, 16 weeks |
Yes |
Clinical symptoms of knee joint discomfort in subjects were improved. |
[35] |
Human |
10 mg, 12 weeks |
Yes |
Mild knee pain in subjects was improved. |
[49] |
Wound healing/Cell proliferation promoting effect |
Mice (NIH3T3, TIG-118 and TIG-121 cells) |
0.1–1000 µg/mL |
- |
CD44-dependent wound healing was enhanced. |
[33] |
Mice (BALB/c) |
0.4–10 mg/mL 10 µL/day, 14 days |
No |
The early stage of wound healing was enhanced. |
[28] |
Human (NHDF cells) |
100–1600 µg/mL |
- |
ERK-driven Cell proliferation was enhanced. |
[29] |
Mice (C3H/HeN) |
Approx. 1mg/day |
Yes |
Hair growth after depilation was enhanced. |
[50] |
Intestinal bacterial flora regulatory effect |
Mice (C57BL/6) |
10 mg/mL 100 µL/day, 30 days |
Yes |
The numbers of probiotic and immunoregulation-related bacteria were enhanced. |
[51] |
Metabolism regulatory effects |
Rat (jejunum sacs) |
1.5–10 mg |
- |
SGLT1-dependent glucose transport was suppressed. |
[34] |
Mice (ICR) |
1 mg/day |
Yes |
High-fat diet-induced increased body weight, serum lipids, and SREBP-1 mRNA levels were suppressed. Leptin level in blood was regulated. |
[32] |
Anti-angiogenesis effect |
Human (EA.hy926 cells) |
0.02–1.0 mg/mL |
- |
The tube formation was suppressed. |
[25] |
Chick (fertilized eggs) |
10–80 µg |
- |
The formation of blood vessels in the chorioallantoic membrane was suppressed. |
Skin condition maintenance effect |
Human (HaCaT, NHDF cells) |
5–25 µg/mL |
- |
The skin barrier function was maintained even in UVB-induced damage conditions. |
[10] |
Mice (hairless SKH-1) |
1 or 5 mg/kg/day |
Yes |
3. The immunoregulatory activities of sPG
3.1 Direct effects of sPG
Immunoregulatory activity of sPG was previously demonstrated. In vitro, the administration of sPG decreased the stimulation of RAW 264.7 cells by heat-killed Escherichia coli cells [27]. TLR4 production was also reduced, suggesting the suppression of signal transduction by the lipopolysaccharide receptor. Pro-inflammatory factors, such as tumor necrosis factor (TNF-α) and inducible NO synthase (iNOS), were simultaneously suppressed. Another study with differentiated Caco-2 cells [52] also indicated the suppression of TNF-α induced interleukin-6 (IL-6) production by sPG, suggesting the inhibition of TNF-α driven inflammation. In vivo, sPG reduced the bacterial number and elevated anti-inflammatory cytokine, (TGF-β and IL-6) production in a Staphylococcus aureus-infected wound [28]. Oral administration of sPG reduces cytokine expression in lamina propria monocytes in an experimental mouse model of colitis [30]. Therefore, sPG exerts an immunoregulatory effect by modulating the expression and activity of cytokines. However, the detailed pathways involved have not yet been identified. Upregulation of Foxp3+ T cells and induction of the expression and phosphorylation of signal transducer and activator of transcription 3 (STAT3) [30] were shown by sPG administration, suggesting that sPG possibly has an anti-inflammatory effect by inducing the differentiation of regulatory T cells (Treg). Foxp3, forkhead box protein p3, is a differentiation marker of Treg. The mechanisms underlying Treg differentiation and CS administration are unclear. Only a few studies have shown that sea cucumber-derived CS (fucosylated CS) affects Treg differentiation [53]. The contribution of the CS chain structure is unclear; however, the effect of sPG on Treg cells may be due to the CS chain.
Regarding the immunomodulatory effects of CS, several in vitro studies have shown that an anti-inflammatory effect mediated by the promotion of nuclear translocation of NF-κB is becoming a common explanation [14, 16, 54, 55, 56, 57, 58]. Hsu et al. [58] reported the effects of CS on the proliferation and migration of chondrocytes (chon-001). CS administration decreases the phosphorylation of Akt, IκB kinase, and IκB. This leads to the inactivation of NF-κB, a downstream factor, and consequently, to the suppression of matrix metalloproteinase (MMP) family expression. MMP family proteins are IL-1β-induced collagen-degrading enzymes and are significant causes of the disruption of articular cartilage structures in OA lesions [59, 60, 61]. Increased expression of metalloproteinase inhibitor-1 and -2 (TIMP-1 and -2) also suggests the functional inhibition of MMP family proteins. Modulation of the NF-κB axis and expression of TIMPs alters the functional profile of the MMP family and interferes with the destruction of the chondrocyte ECM [58, 59, 60]. In addition, the expression of type II collagen increases, thereby improving the state of the ECM. IL-1β stimulation was downregulated in the presence of CS. The inhibition of IL-1β induced NLRP3 inflammasome activation has also been observed [54]. It is unclear whether this downregulation is mediated by blocking IL-1β receptors or the sequestration of cytokines through the interaction of CS and IL-1β. Potential interactions between sPG and hematopoietic cytokines have also been observed. HS, a member of the GAG family that shares a common structure with CS, is well-known for its ability to bind cytokines and morphogens [65, 66, 67, 68]. The CS chain, especially the CS-E units-abundant chain, exhibits activities similar to those of HS [69, 70, 71, 72, 73]. The structure-dependent binding of the CS chain to immunomodulatory factors, including interleukin, needs to be studied. Moreover, this explanation using NF-κB-related anti-inflammatory signaling is limited to systems where sPG can directly reach affected areas and cells. The systemic effects of orally administered CS/CSPG are discussed in the next section.
3.2 Systemic effects after oral sPG administration
Oral administration of sPG to mice with papain-induced experimental respiratory disease decreased the titer of serum IgE, infiltration of eosinophils, and epithelial or T helper type 2 cells-related cytokines [31]. In a collagen-induced allergic model of rheumatoid arthritis, the clinical and histological severity of the disease and pro-inflammatory cytokine production in the joints were reduced [26]. These experiments demonstrated the cytokine modulatory effects of orally administered sPG in affected areas, far from the gastrointestinal tract. In addition, clinical studies have revealed sustained improvement in diagnostic scores with sPG administration, especially in patients in poor conditions [35, 49, 74]. sPG has also improved collagen degradation and cartilage metabolism markers in the serum and urine, suggesting inhibition of cartilage degradation or promotion of synthesis. These results indicated that oral administration of sPG may exert anti-inflammatory and analgesic effects that are not solely dependent on its cytokine-modulatory effects. However, the amount of CS absorbed by oral administration is limited in many cases. Therefore, whether sufficient amounts of sPG reach the affected areas in these experiments, such as the respiratory organs and joints, is uncertain.
In addition to salmon, proteoglycans from sharks have also been investigated for their oral effects. A fraction of cartilage from the bramble shark (Echinorhinus brucus) was extracted by Ajeeshkumar et al. [75], which appeared to contain aggrecan and epiphycan. The bramble shark PG oral administration at doses of 50 mg/kg or higher improved the histopathological characteristics of the monosodium iodoacetate-induced OA rat model. The mRNA levels of pro-inflammatory cytokines TNF-α, IL-1β, MMP-13, NOS2, and COX-2 were reduced in the joints, while the anti-inflammatory cytokine, IL-10, was enhanced. Kitahashi et al. [76] prepared a CS/protein mixture from blue shark (Prionace glauca) and orally administered it to experimental pancreatic cancer mouse models. Although the mechanism is unknown, serum MMP-9 inhibitory activity was significantly enhanced after oral administration. The authors hypothesized the importance of the protein moiety, based on the lack of MMP-9 inhibitory activity of purified shark CS. However, according to the literature described above, this can be considered as an effect of the MMP inhibitor induced by the CS moiety. These results suggested that sPG and other CS/CSPG products exhibit systemic effects after oral administration. In Figure 1, the potential activities of CSPGs in the level of intracellular signaling that we have mentioned above are briefly summarized.

Figure 1: Potential effects of CSPG on cellular signal transductions. CSPG suppresses NF-κB axis-induced inflammation and ECM degradation. Conversely, CSPG enhances cell proliferation via ERK1/2 activation; however, the upper-stream receptors are not being identified. Solid and dotted arrows indicate activation and nuclear translocation, respectively. The numbers in the CS structure shown in green indicate the position of the sulfate group.
As mentioned above, many in vitro studies have shown that the direct exogenous administration of CS to chondrocytes can contribute to homeostasis by suppressing the inflammatory state of chondrocytes and inhibiting the destruction of aggrecan and collagen. These in vitro studies have been used as an explanation for the systemic clinical effects of oral administration. However, in interpreting the results of these studies, it must be noted that CS is an originally endogenous factor in the ECM of most somatic cells, including chondrocytes [2, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87]. CS synthesis and homeostatic alterations affect phenotype, proliferation, and migration of various cell species. Exogenous CS can substitute for some of the functions of endogenous CS. Therefore, experiments in which CS is administered directly to cells merely observe the results of altering the ECM of cultured cells, rather than reproducing the results of CS reaching the affected area after oral intake. Notably, many of them in vitro studies administered fairly large doses of CS, as described by Henrotin et al. [13]
CS is recognized as a slow-acting drug and some studies suggest that it may take up to two years of dosing to achieve benefits [19, 88]. Continuous use may lead to accumulation. However, accumulation has not yet been traced clinically or experimentally. Therefore, it is crucial to quantitatively study the distribution of CS in the body after oral administration.
4. Behavior of CS/CSPG after oral administration
Only Kudo et al. [52] reported experiments to trace the degradation of orally administered sPG in mice, showing that while the core protein underwent digestion, the CS moiety was not degraded in the digestive tract, at least up to the small intestine. Stellavato et al. [18] reported that CS might reach the intestine with little loss in digestion studies mimicking the stomach and intestine. To date, the only human extracellular enzyme capable of degrading CS chains is the hyaluronidase family, particularly hyaluronidase-4 (HYAL4) [89, 90, 91, 92]. Since HYAL4 expression is not ubiquitous and is spatiotemporally localized, it does not contribute to CS degradation in the digestive tract.
Barthe et al. [93] performed in vitro transit and digestion studies on 14C-labeled CS, and found that degradation occurred only when it was mixed with the contents of the colon and cecum. Absorption occurred in small amounts in the small intestine and, to a greater extent, in the colon and cecum. However, the only component absorbed was the disaccharide. Another study reported that CS did not undergo any degradation in the gastrointestinal tract other than by intestinal bacteria and absorption of CS is thought to occur in the proximal portion of the intestine [18].
Several radioisotope studies [94, 95, 96] in healthy adults have shown that orally administered CS is distributed in the liver, kidneys, and urine. Volpi [97, 98] revealed that the concentration of high-molecular-weight CS (leaving at least 10 residues) in the blood of healthy adults increases after its oral administration. These results suggest that orally administered CS can translocate from the gastrointestinal tract into blood vessels. However, the amount of CS absorbed or translocated is extremely limited. In Volpi’s experiment [97], in which 4 g of Chondrosulf®, derived from bovine, was administered to healthy volunteers, the amount of CS in the blood was 263% of baseline with endogenous CS. In another study by Volpi [98], in which 4 g of fish-derived CS was administered, blood CS levels increased by 126%. In contrast, Jackson et al. [99] could not detect CS transfer into the blood. This is because the amount of endogenous CS in the blood is very low (around 4 µg/mL), making detection and comparison difficult. Therefore, the amount absorbed by Volpi in these experiments was also insignificant.
Adebowale et al. [46] used monolayer-cultured differentiated Caco-2 cells as a model of the human intestinal epithelium and investigated the export of CS from the intestinal lumen via the paracellular pathway of the intestinal tract. Explanations based on the paracellular pathway have garnered attention. However, the size of the CS that can be transported via the paracellular pathway is extremely limited, even in vitro [20]. CSPG is much larger than a single CS chain, therefore, translocation through the gastrointestinal epithelium is more difficult for CSPG than for CS.
Preliminary ex vivo experiments using everted sacs have also been reported [100], in which dose-dependent sPG absorption was detected in the jejunum, ileum, and colon of the rat intestine. Addition of an inhibitor of clathrin-mediated endocytosis inhibited sPG absorption. In this preliminary study, although the setting of the control group in the inhibitor experiment was inappropriate, the decrease in detection from the proximal to distal supported CS/CSPG absorption in the proximal small intestine.
Therefore, the amount and molecular weight of CS that can be absorbed after oral administration are limited. CSPG, such as sPG, are even more limited because they are larger than a single CS chain. In addition, because the CS chains are rarely digested in the proximal small intestine, which is a possible site of CS absorption, the low-molecular-weight fraction originally present in the product would be absorbed rather than the depolymerized CS derived from digestion. Therefore, most CS/CSPGs would appear to pass through the gastrointestinal tract as passengers.
5. What can CS/CSPG do without being absorbed
The behavior of orally administered CS is not straightforward [15, 99]. As mentioned previously, CS intrinsically contributes to several physiological processes in various cell types. This suggests that CS interacts with signal transduction-associated proteins. Integrins, receptor type kinases, and N-cadherins are candidates [7, 29, 40, 101, 102]. When sPG was administered to normal human dermal fibroblast (NHDF) cells, a cell proliferation-promoting effect with increased ERK1/2 phosphorylation was observed, suggesting the contribution of integrin signaling [29]. These interactions indicate that part of CS/CSPG, which is not absorbed after oral administration and is considered a passenger, can still act as a messenger, potentially interacting with cells that form the lumen of the gastrointestinal tract.
Galectin-3 (Gal-3), a multifunctional lectin, is present in healthy intestinal epithelium [103]. An interaction between the CS chain and Gal-3 has been previously reported despite its physiological significance remains unclear [104]. The importance of Gal-3 and CS, particularly in the cancer microenvironment, has been independently reported [105, 106, 107, 108]. However, the relationship between Gal-3 and CS in healthy tissue requires further investigation. Furthermore, integrin αVβ3 is a candidate receptor protein of the CS-D units-rich CS chain in the central nervous system. Integrin αV is expressed in macrophages [109], suggesting that macrophages in intestinal lymph nodes may potentially receive CS chains. sPG contains approximately 7% CS-D, therefore, it is expected to be trapped by integrin αV.
Pectin is an example of a polysaccharide that interacts directly with proteins in the intestinal tract. Pectin is a polysaccharide obtained from land plants and a typical dietary fiber [110, 111]. Pectin influences the behavior of animal cells, including proliferation and differentiation [112, 113, 114, 115, 116, 117]. Pectin has direct nonprebiotic and prebiotic effects on the intestinal tract [112, 118, 119]. Pectin is associated with fibronectin, and the association of fibronectin with integrin β1 is also disrupted by the presence of pectin [120]. These properties may explain the physiological effects of pectin. Notably, the interaction between CS and fibronectin is classically studied well [39, 121, 122, 123]. CSPG binds to fibronectin and collagen in a CS chain-dependent manner and HA in a core protein-dependent in joints, forming a network structure that makes joints more resistant to mechanical impact. This similarity between the two carboxy group-rich acidic polysaccharides implies the potential interaction of CS in the gastrointestinal tract.
Finally, CS is utilized by the intestinal bacteria. CS is degraded by intestinal bacteria to produce hydrogen sulfide (H2S). Oral administration of CS increases levels of H2S and its producing bacteria; glucagon-like peptide 1 (GLP-1) and insulin secretion; and glucose tolerance [124, 125, 126]. These alterations may have been caused by H2S. Presumably, the sulfate groups of CS are a good source of sulfur for intestinal bacteria. Oral administration of sPG decreases mRNA levels of SREBP-1 in the liver [32]. H2S regulates the expression of SREBP-1, therefore, oral sPG administration is considered to influence SREBP-1 via H2S production. Oral administration of sPG has also been reported to modulate the intestinal microbiota and increase the amount of short-chain fatty acids [48, 51]. These results indicate that CS is a prebiotic agent.
Thus, the amount of CS/CSPG absorbed and distributed throughout the body is insignificant. CS/CSPG distributed in the body may not only act directly on the affected area but also the whole body. It is possible that unabsorbed CS/CSPG interacts with proteins in the gastrointestinal tract and acts prebiotically. The function of CS/CSPG may be exerted not only by absorption, but also by other pathways in the gastrointestinal tract. The state of CS/CSPG in the gastrointestinal tract remains unexplored, but it potentially exhibits dietary fiber or luminacoid behavior, similar to pectin. sPG is larger than CS alone, and is expected to have poorer absorption. Studies focusing on luminacoid-like behavioral properties and absorption are needed to elucidate the effects of oral administration of sPG.
6. Conclusions
sPG is a safe proteoglycan with no concerns regarding religious restrictions or serious infectious diseases. The anti-inflammatory and cell-proliferation-promoting activities of sPG are supported by studies using CS derived from other sources. The physiological significance of the molecular weight and sulfation pattern of sPG can be better understood by comparing it with those of CS derived from other raw materials. However, the precise mechanisms of action of sPG and other CS/CSPGs, particularly after oral administration, remain unknown. No absorption or translocation pathways were identified. Due to the poor absorption of CS/CSPG and the uncertainty of its mechanism, the conventional narrative that CS/CSPG is absorbed from the intestinal tract and reaches the affected area may not be sufficient. However, studies on the role of CS/CSPG in the intestinal tract are lacking. The possibility of prebiotic or luminacoid behavior in CS/CSPG has not yet received much attention. Their elucidation may shed light on the mechanisms of the oral administration of CS/CSPG and its benefits.
References
- [1] Mikami T and Kitagawa H (2013) Biosynthesis and function of chondroitin sulfate. Biochim. Biophys. Acta, 1830: 4719–4733. https://doi.org/10.1016/J.BBAGEN.2013.06.006
- [2] Mikami T and Kitagawa H (2023) Chondroitin sulfate glycosaminoglycans function as extra/pericellular ligands for cell surface receptors. J. Biochem., 173: 329–332. https://doi.org/10.1093/JB/MVAC110
- [3] Mizumoto S and Yamada S (2023) Histories of Dermatan Sulfate Epimerase and Dermatan 4-O-Sulfotransferase from Discovery of Their Enzymes and Genes to Musculocontractural Ehlers-Danlos Syndrome. Genes (Basel), 14: 509. https://doi.org/10.3390/GENES14020509
- [4] Mitsunaga C, Mikami T, Mizumoto S, Fukuda J and Sugahara K (2006) Chondroitin sulfate/dermatan sulfate hybrid chains in the development of cerebellum: Spatiotemporal regulation of the expression of critical disulfated disaccharides by specific sulfotransferases. J. Biol. Chem., 281: 18942–18952. https://doi.org/10.1074/JBC.M510870200
- [5] Properzi F, Carulli D, Asher RA, Muir E, Camargo LM, Van Kuppevelt TH et al. (2005) Chondroitin 6-sulphate synthesis is up-regulated in injured CNS, induced by injury-related cytokines and enhanced in axon-growth inhibitory glia. Eur. J. Neurosci., 21: 378–390. https://doi.org/10.1111/J.1460-9568.2005.03876.X
- [6] Sakamoto K, Ozaki T, Ko YC, Tsai CF, Gong Y, Morozumi M et al. (2019) Glycan sulfation patterns define autophagy flux at axon tip via PTPRσ-cortactin axis. Nat. Chem. Biol., 15: 699–709. https://doi.org/10.1038/S41589-019-0274-X
- [7] Shida M, Mikami T, Tamura J ichi and Kitagawa H (2019) Chondroitin sulfate-D promotes neurite outgrowth by acting as an extracellular ligand for neuronal integrin αVβ3. Biochim. Biophys. Acta Gen. Subj., 1863: 1319–1331. https://doi.org/10.1016/J.BBAGEN.2019.06.004
- [8] Watanabe H (2022) Aggrecan and versican: two brothers close or apart. Am. J. Physiol. Cell Physiol., 322: C967–C976. https://doi.org/10.1152/AJPCELL.00081.2022/ASSET/IMAGES/LARGE/AJPCELL.00081.2022_F001.JPEG
- [9] Zhang Y, Cao L, Kiani CG, Yang BL and Yang BB (1998) The G3 domain of versican inhibits mesenchymal chondrogenesis via the epidermal growth factor-like motifs. J. Biol. Chem., 273: 33054–33063. https://doi.org/10.1074/JBC.273.49.33054
- [10] Lee HR, Hong SM, Cho K, Kim SH, Ko E, Lee E et al. (2024) Potential role of dietary salmon nasal cartilage proteoglycan on UVB-Induced photoaged skin. Biomol. Ther., 32: 249–260. https://doi.org/10.4062/BIOMOLTHER.2024.010
- [11] Phipps KR, Lee HY, Kim H and Jeon B (2020) Oral administration of a novel hydrolyzed chicken sternal cartilage extract (BioCell Collagen®) reduces UVB-induced photoaging in mice. J. Funct. Foods., 68: 103870. https://doi.org/10.1016/J.JFF.2020.103870
- [12] Volpi N (2019) Chondroitin sulfate safety and quality. Molecules, 24: 1447. https://doi.org/10.3390/MOLECULES24081447
- [13] Henrotin Y, Mathy M, Sanchez C and Lambert C (2010) Chondroitin sulfate in the treatment of osteoarthritis: From in vitro studies to clinical recommendations. Ther. Adv. Musculoskelet. Dis., 2: 335–348. https://doi.org/10.1177/1759720X10383076
- [14] Kubo M, Ando K, Mimura T, Matsusue Y and Mori K (2009) Chondroitin sulfate for the treatment of hip and knee osteoarthritis: current status and future trends. Life Sci., 85: 477–483. https://doi.org/10.1016/J.LFS.2009.08.005
- [15] Bishnoi M, Jain A, Hurkat P and Jain SK (2016) Chondroitin sulphate: a focus on osteoarthritis. Glycoconj. J., 2016 33:5, 33: 693–705. https://doi.org/10.1007/S10719-016-9665-3
- [16] Vasiliadis HS and Tsikopoulos K (2017) Glucosamine and chondroitin for the treatment of osteoarthritis. World J. Orthop., 8: 1–11. https://doi.org/10.5312/wjo.v8.i1.1
- [17] Bottegoni C, Muzzarelli RAA, Giovannini F, Busilacchi A and Gigante A (2014) Oral chondroprotection with nutraceuticals made of chondroitin sulphate plus glucosamine sulphate in osteoarthritis. Carbohydr. Polym., 109: 126–138. https://doi.org/10.1016/J.CARBPOL.2014.03.033
- [18] Restaino OF, Finamore R, Stellavato A, Diana P, Bedini E, Trifuoggi M et al. (2019) European chondroitin sulfate and glucosamine food supplements: A systematic quality and quantity assessment compared to pharmaceuticals. Carbohydr. Polym., 222: 114984. https://doi.org/10.1016/J.CARBPOL.2019.114984
- [19] Du Souich P (2014) Absorption, distribution and mechanism of action of SYSADOAS. Pharmacol. Ther., 142: 362–374. https://doi.org/10.1016/J.PHARMTHERA.2014.01.002
- [20] Jin M, Satsu H, Yamada K, Hisada N, Totsuka M and Shimizu M (2010) Permeation of disaccharides derived from chondroitin sulfate through human intestinal Caco-2 cell monolayers via the paracellular pathway. Biosci. Biotechnol. Biochem., 74: 1243–1249. https://doi.org/10.1271/BBB.100075
- [21] Majima M, Takagaki K, Sudo Shin-ichiro, Yoshihara S, Kudo Y and Yamagishi S (2001) Effect of proteoglycan on experimental colitis. Int. Congr. Ser., 1223: 221–224. https://doi.org/10.1016/S0531-5131(01)00479-4
- [22] Kakizaki I, Tatara Y, Majima M, Kato Y and Endo M (2011) Identification of proteoglycan from salmon nasal cartilage. Arch. Biochem. Biophys., 506: 58–65. https://doi.org/10.1016/J.ABB.2010.10.025
- [23] Kakizaki I, Mineta T, Sasaki M, Tatara Y, Makino E and Kato Y (2014) Biochemical and atomic force microscopic characterization of salmon nasal cartilage proteoglycan. Carbohydr. Polym., 103: 538–549. https://doi.org/10.1016/J.CARBPOL.2013.12.083
- [24] Li W, Ura K and Takagi Y (2022) Industrial application of fish cartilaginous tissues. Curr. Res. Food Sci., 5: 698–709. https://doi.org/10.1016/J.CRFS.2022.04.001
- [25] Kobayashi T, Kakizaki I, Nozaka H and Nakamura T (2017) Chondroitin sulfate proteoglycans from salmon nasal cartilage inhibit angiogenesis. Biochem. Biophys. Rep., 9: 72–78. https://doi.org/10.1016/J.BBREP.2016.11.009
- [26] Yoshimura S, Asano K and Nakane A (2014) Attenuation of Collagen-Induced Arthritis in Mice by Salmon Proteoglycan. Biomed Res. Int., 2014: 406453. https://doi.org/10.1155/2014/406453
- [27] Sashinami H, Takagaki K and Nakane A (2006) Salmon cartilage proteoglycan modulates cytokine responses to Escherichia coli in mouse macrophages. Biochem. Biophys. Res. Commun., 351: 1005–1010. https://doi.org/10.1016/J.BBRC.2006.10.146
- [28] Hirose S, Narita K, Asano K and Nakane A (2018) Salmon cartilage proteoglycan promotes the healing process of Staphylococcus aureus-infected wound. Heliyon, 4: e00587. https://doi.org/10.1016/J.HELIYON.2018.E00587
- [29] Sano M, Shang Y, Nakane A and Saito T (2017) Salmon nasal cartilage proteoglycan enhances growth of normal human dermal fibroblast through Erk1/2 phosphorylation. Biosci. Biotechnol. Biochem., 81: 1379–1385 https://doi.org/10.1080/09168451.2017.1318695.
- [30] Mitsui T, Sashinami H, Sato F, Kijima H, Ishiguro Y, Fukuda S et al. (2010) Salmon cartilage proteoglycan suppresses mouse experimental colitis through induction of Foxp3+ regulatory T cells. Biochem. Biophys. Res. Commun., 402: 209–215. https://doi.org/10.1016/J.BBRC.2010.09.123
- [31] Ono HK, Yoshimura S, Hirose S, Narita K, Tsuboi M, Asano K et al. (2018) Salmon cartilage proteoglycan attenuates allergic responses in mouse model of papain-induced respiratory inflammation. Mol. Med. Rep., 18: 4058–4064. https://doi.org/10.3892/MMR.2018.9364/HTML
- [32] Hirose S, Asano K, Harada S, Takahashi T, Kondou E, Ito K et al. (2022) Effects of salmon cartilage proteoglycan on obesity in mice fed with a high-fat diet. Food Sci. Nutr., 10: 577–583. https://doi.org/10.1002/FSN3.2685
- [33] Ito G, Kobayashi T, Takeda Y and Sokabe M (2015) Proteoglycan from salmon nasal cartridge promotes in vitro wound healing of fibroblast monolayers via the CD44 receptor. Biochem. Biophys. Res. Commun., 456: 792–798. https://doi.org/10.1016/J.BBRC.2014.12.037
- [34] Tsuchiya Y, Kawamata K, Tomita M, Tsuboi M, Takahashi T and Yonezuka M (2015) Effects of salmon nasal cartilage proteoglycan on plasma glucose concentration and active glucose transport in the small intestine. J. Nutr. Sci. Vitaminol. (Tokyo), 61: 502–505. https://doi.org/10.3177/JNSV.61.502
- [35] Tomonaga A, Takahashi T, Tanaka YT, Tsuboi M, Ito K and Nagaoka I (2017) Evaluation of the effect of salmon nasal proteoglycan on biomarkers for cartilage metabolism in individuals with knee joint discomfort: A randomized double-blind placebo-controlled clinical study. Exp. Ther. Med., 14: 115. https://doi.org/10.3892/ETM.2017.4454
- [36] Tatara Y, Kakizaki I, Kuroda Y, Suto S, Ishioka H and Endo M (2013) Epiphycan from salmon nasal cartilage is a novel type of large leucine-rich proteoglycan. Glycobiology, 23: 993–1003. https://doi.org/10.1093/GLYCOB/CWT038
- [37] Watanabe H, Yamada Y and Kimata K (1998) Roles of aggrecan, a large chondroitin sulfate proteoglycan, in cartilage structure and function. J. Biochem., 124: 687–693. https://doi.org/10.1093/OXFORDJOURNALS.JBCHEM.A022166
- [38] Watanabe H (2022) Aggrecan and versican: two brothers close or apart. Am. J. Physiol. Cell Physiol., 322: C967–C976. https://doi.org/10.1152/AJPCELL.00081.2022/ASSET/IMAGES/LARGE/AJPCELL.00081.2022_F001.JPEG
- [39] Yamagata M, Yamada KM, Yoneda M, Suzuki S and Kimata K (1986) Chondroitin sulfate proteoglycan (PG-M-like proteoglycan) is involved in the binding of hyaluronic acid to cellular fibronectin. J. Biol. Chem., 261: 13526–13535. https://doi.org/10.1016/S0021-9258(18)67050-4
- [40] Satoh F, Sugiura A, Tashiro J, Hosaka YZ and Warita K (2022) Chondroitin sulfate E downregulates N-cadherin and suppresses myotube formation. J. Vet. Med. Sci., 84: 494. https://doi.org/10.1292/JVMS.21-0662
- [41] Volpi N (2009) Quality of different chondroitin sulfate preparations in relation to their therapeutic activity. J. Pharm. Pharmacol., 61: 1271–1280. https://doi.org/10.1211/JPP.61.10.0002
- [42] Zhang W, Giancaspro G, Adams KM, Neal-Kababick J, Hildreth J, Li A et al. (2014) Electrophoretic separation of alginic sodium diester and sodium hexametaphosphate in chondroitin sulfate that interfere with the cetylpyridinium chloride titration assay. J. AOAC Int., 97: 1503–1513. https://doi.org/10.5740/JAOACINT.14-167
- [43] Martel-Pelletier J, Farran A, Montell E, Vergés J and Pelletier JP (2015) Discrepancies in composition and biological effects of different formulations of chondroitin sulfate. Molecules, 20: 4277. https://doi.org/10.3390/MOLECULES20034277
- [44] Restaino OF and Schiraldi C (2022) Chondroitin sulfate: are the purity and the structural features well assessed? A review on the analytical challenges. Carbohydr. Polym., 292: 119690. https://doi.org/10.1016/J.CARBPOL.2022.119690
- [45] Da Cunha AL, De Oliveira LG, Maia LF, De Oliveira LFC, Michelacci YM and De Aguiar JAK (2015) Pharmaceutical grade chondroitin sulfate: Structural analysis and identification of contaminants in different commercial preparations. Carbohydr. Polym., 134: 300–308. https://doi.org/10.1016/J.CARBPOL.2015.08.006
- [46] Adebowale AO, Cox DS, Liang Z, and Eddington ND (2000) Analysis of glucosamine and chondroitin sulfate content in marketed products and the Caco-2 permeability of chondroitin sulfate raw materials. J. Am. Nutraceutical Assoc., Accessed: Nov. 14, 2024. [Online]. Available: http://hospitalprivileges.net/documents/9ChondroitinGlucosamineEddingtonStudy.pdf
- [47] Restaino OF, Finamore R, Diana P, Marseglia M, Vitiello M, Casillo A et al. (2017) A multi-analytical approach to better assess the keratan sulfate contamination in animal origin chondroitin sulfate. Anal. Chim. Acta, 958: 59–70. https://doi.org/10.1016/J.ACA.2016.12.005
- [48] Ota S, Yoshihara S, Ishido K, Tanaka M, Takagaki K and Sasaki M (2008) Effects of proteoglycan on dextran sulfate sodium-induced experimental colitis in rats. Dig. Dis. Sci., 53: 3176–3183. https://doi.org/10.1007/S10620-008-0304-0
- [49] Takahashi T et al. (2018) Evaluation of the efficacy and safety of long-term intake of a dietary supplement containing salmon nasal cartilage—derived proteoglycan on subjects with subjective knee symptoms ―An open study―. Yakuri to Chiryo, 46: 1693–1706. Accessed: Jan. 28, 2025. [Online]. Available: https://www.pieronline.jp/content/article/0386-3603/46100/1693
- [50] Nakane A, Hirose S, Kawai N, Fujimoto N, Kondo E and Asano K (2023) Salmon nasal cartilage proteoglycan stimulates hair growth. Biosci. Biotechnol. Biochem., 88: 107–110. https://doi.org/10.1093/BBB/ZBAD149
- [51] Asano K, Yoshimura S and Nakane A (2013) Alteration of intestinal microbiota in mice orally administered with salmon cartilage proteoglycan, a prophylactic agent. PLoS One, 8: e75008. https://doi.org/10.1371/JOURNAL.PONE.0075008
- [52] Kudo K, Kobayashi T, Kasai K, Nozaka H and Nakamura T (2023) Chondroitin sulfate is not digested at all in the mouse small intestine but may suppress interleukin 6 expression induced by tumor necrosis factor-α. Biochem. Biophys. Res. Commun., 642: 185–191. https://doi.org/10.1016/J.BBRC.2022.12.051
- [53] Li C, Tian Y, Pei J, Zhang Y, Hao D, Han T et al. (2023) Sea cucumber chondroitin sulfate polysaccharides attenuate OVA-induced food allergy in BALB/c mice associated with gut microbiota metabolism and Treg cell differentiation. Food Funct., 14: 7375–7386. https://doi.org/10.1039/D3FO00146F
- [54] Chang YT, Huang KC, Pranata R, Chen YL, Chen SN, Cheng YH et al. (2024) Evaluation of the protective effects of chondroitin sulfate oligosaccharide against osteoarthritis via inactivation of NLRP3 inflammasome by in vivo and in vitro studies. Int. Immunopharmacol., 142: 113148. https://doi.org/10.1016/J.INTIMP.2024.113148
- [55] Legendre F, Baugé C, Roche R, Saurel AS and Pujol JP (2008) Chondroitin sulfate modulation of matrix and inflammatory gene expression in IL-1β-stimulated chondrocytes – study in hypoxic alginate bead cultures. Osteoarthr. Cartil., 16: 105–114. https://doi.org/10.1016/J.JOCA.2007.05.020
- [56] Jomphe C, Gabriac M, Hale TM, Héroux L, Trudeau LÉ, Deblois D et al. (2008) Chondroitin sulfate inhibits the nuclear translocation of nuclear factor-kappaB in interleukin-1beta-stimulated chondrocytes. Basic Clin. Pharmacol. Toxicol., 102: 59–65. https://doi.org/10.1111/J.1742-7843.2007.00158.X
- [57] Du Souich P, García AG, Vergés J and Montell E (2009) Immunomodulatory and anti-inflammatory effects of chondroitin sulphate. J. Cell. Mol. Med., 13: 1451–1463. https://doi.org/10.1111/J.1582-4934.2009.00826.X
- [58] Hsu HC, Ke YL, Lai YH, Hsieh WC, Lin CH, Huang SS et al. (2022) Chondroitin sulfate enhances proliferation and migration via inducing β-Catenin and intracellular ROS as well as suppressing metalloproteinases through Akt/NF-ϰB pathway inhibition in human chondrocytes. J. Nutr. Health Aging, 26: 307–313. https://doi.org/10.1007/S12603-022-1752-5
- [59] Shlopov B V., Gumanovskaya ML and Hasty KA (2000) Autocrine regulation of collagenase 3 (matrix metalloproteinase 13) during osteoarthritis. Arthritis Rheum., 43: https://doi.org/10.1002/1529-0131(200001)43:1<195::AID-ANR24>3.0.CO;2-G
- [60] Forsyth CB, Cole A, Murphy G, Bienias JL, Im HJ and Loeser RF (2005) Increased matrix metalloproteinase-13 production with aging by human articular chondrocytes in response to catabolic stimuli. J. Gerontol. A Biol. Sci. Med. Sci., 60: 1118–1124. https://doi.org/10.1093/gerona/60.9.1118
- [61] Goldring MB, Otero M, Tsuchimochi K, Ijiri K and Li Y (2008) Defining the roles of inflammatory and anabolic cytokines in cartilage metabolism. Ann. Rheum. Dis., 2008. https://doi.org/10.1136/ard.2008.098764
- [62] Cui N, Hu M and Khalil RA (2017) Biochemical and biological attributes of matrix metalloproteinases. Prog. Mol. Biol. Transl. Sci., 147: 1–73. https://doi.org/10.1016/BS.PMBTS.2017.02.005
- [63] Nagase H, Visse R and Murphy G (2006) Structure and function of matrix metalloproteinases and TIMPs. Cardiovasc. Res., 69: 562–573. https://doi.org/10.1016/J.CARDIORES.2005.12.002/2/69-3-562-FIG3.GIF
- [64] Grillet B, Pereira RVS, Van Damme J, Abu El-Asrar A, Proost P and Opdenakker G (2023) Matrix metalloproteinases in arthritis: towards precision medicine. Nat. Rev. Rheumatol., 2023 19:6, 19: 363–377. https://doi.org/10.1038/s41584-023-00966-w
- [65] Tumova S, Woods A and Couchman JR (2000) Heparan sulfate proteoglycans on the cell surface: Versatile coordinators of cellular functions. Int. J. Biochem. Cell Biol., 32: 269–288. https://doi.org/10.1016/S1357-2725(99)00116-8
- [66] Dreyfuss JL, Regatieri C V., Jarrouge TR, Cavalheiro RP, Sampaio LO and Nader HB (2009) Heparan sulfate proteoglycans: Structure, protein interactions and cell signaling. An. Acad. Bras. Cienc., 81: 409–429. https://doi.org/10.1590/S0001-37652009000300007
- [67] Sarrazin S, Lamanna WC and Esko JD (2011) Heparan sulfate proteoglycans. Cold Spring Harb. Perspect. Biol., 3: 1–33. https://doi.org/10.1101/CSHPERSPECT.A004952
- [68] Chua CC, Rahimi N, Forsten-Williams K and Nugent MA (2004) Heparan sulfate proteoglycans function as receptors for fibroblast growth factor-2 activation of extracellular signal-regulated kinases 1 and 2. Circ. Res., 94: 316–323. https://doi.org/10.1161/01.RES.0000112965.70691.AC
- [69] Maruyama T, Toida T, Imanari T, Yu G and Linhardt RJ (1998) Conformational changes and anticoagulant activity of chondroitin sulfate following its O-sulfonation. Carbohydr. Res., 306: 35–43. https://doi.org/10.1016/S0008-6215(97)10060-X
- [70] Higashi N, Maeda R, Sesoko N, Isono M, Ishikawa S, Tani Y et al. (2019) Chondroitin sulfate E blocks enzymatic action of heparanase and heparanase-induced cellular responses. Biochem. Biophys. Res. Commun., 520: 152–158. https://doi.org/10.1016/J.BBRC.2019.09.126
- [71] Habuchi O (2022) Functions of chondroitin/dermatan sulfate containing GalNAc4,6-disulfate. Glycobiology, 32: 664–678. https://doi.org/10.1093/GLYCOB/CWAC030
- [72] Nadanaka S, Ishida M, Ikegami M and Kitagawa H (2008) Chondroitin 4-O-sulfotransferase-1 modulates Wnt-3a signaling through control of E disaccharide expression of chondroitin sulfate. J. Biol. Chem., 283: 27333–27343. https://doi.org/10.1074/JBC.M802997200
- [73] Nadanaka S, Kinouchi H, Taniguchi-Morita K, Tamura JI and Kitagawa H (2011) Down-regulation of chondroitin 4-O-sulfotransferase-1 by Wnt signaling triggers diffusion of Wnt-3a. J. Biol. Chem., 286: 4199–4208. https://doi.org/10.1074/jbc.M110.155093
- [74] Thanh Vu HT, Thu Nguyen HT, Hoai Nguyen TT, Nguyen TX, Nguyen TN, Nguyen AL et al. (2024) The efficacy and tolerability of proteoglycan F in the treatment of knee osteoarthritis: A prospective, randomized, double-blind controlled trial. Osteoarthr. Cartil. Open, 6: https://doi.org/10.1016/J.OCARTO.2024.100483
- [75] Ajeeshkumar KK, Vishnu KV, Navaneethan R, Raj K, Remyakumari KR, Swaminathan TR et al. (2019) Proteoglycans isolated from the bramble shark cartilage show potential anti-osteoarthritic properties. Inflammopharmacology, 27: 175–187. https://doi.org/10.1007/S10787-018-00554-5/TABLES/1
- [76] Kitahashi T, Ikawa S, Sakamoto A, Nomura Y, Tsujiuchi T, Shimizu K et al. (2012) Ingestion of proteoglycan fraction from shark cartilage increases serum inhibitory activity against matrix metalloproteinase-9 and suppresses development of N-nitrosobis(2-oxopropyl)amine-induced pancreatic duct carcinogenesis in hamster. J. Agric. Food Chem., 60: 940–945. https://doi.org/10.1021/JF203613B
- [77] Macke EL, Henningsen E, Jessen E, Zumwalde NA, Landowski M, Western DE et al. (2020) Loss of chondroitin sulfate modification causes inflammation and neurodegeneration in skt mice. Genetics, 214: 121–134. https://doi.org/10.1534/GENETICS.119.302834
- [78] Liu CH, Lan CT, Chou JF, Tseng TJ and Liao WC (2017) CHSY1 promotes aggressive phenotypes of hepatocellular carcinoma cells via activation of the hedgehog signaling pathway. Cancer Lett., 403: 280–288. https://doi.org/10.1016/J.CANLET.2017.06.023
- [79] Kwok JCF, Sela-Donenfeld D, Cai C, Wang Z, Chi L and Zhang B (2021) Chondroitin sulfate/dermatan sulfate-protein interactions and their biological functions in human diseases: implications and analytical tools. Front. Cell Dev. Biol., 9: 693563. https://doi.org/10.3389/FCELL.2021.693563
- [80] Adhikara IM, Yagi K, Mayasari DS, Ikeda K, Kitagawa H, Miyata O et al. (2019) Chondroitin sulfate N-acetylgalactosaminyltransferase-2 deletion alleviates lipoprotein retention in early atherosclerosis and attenuates aortic smooth muscle cell migration. Biochem. Biophys. Res. Commun., 509: 89–95. https://doi.org/10.1016/J.BBRC.2018.12.068
- [81] Adhikara IM, Yagi K, Mayasari DS, Suzuki Y, Ikeda K, Ryanto GRT et al. (2021) Chondroitin sulfate N-acetylgalactosaminyltransferase-2 impacts foam cell formation and Atherosclerosis by Altering Macrophage Glycosaminoglycan Chain. Arterioscler. Thromb. Vasc. Biol., 41: 1076–1091. https://doi.org/10.1161/ATVBAHA.120.315789
- [82] Schwartz NB and Domowicz MS (2022) Roles of Chondroitin Sulfate Proteoglycans as Regulators of Skeletal Development. Front. Cell Dev. Biol., 10: 745372. https://doi.org/10.3389/FCELL.2022.745372.
- [83] Kitazawa K, Nadanaka S, Kadomatsu K and Kitagawa H (2021) Chondroitin 6-sulfate represses keratinocyte proliferation in mouse skin, which is associated with psoriasis. Commun. Biol., 2021 4:1, 4: 1–15. https://doi.org/10.1038/s42003-020-01618-5
- [84] Mizumoto S and Yamada S (2021) An overview of in vivo functions of chondroitin sulfate and dermatan sulfate revealed by their deficient mice. Front. Cell Dev. Biol., 9: https://doi.org/10.3389/FCELL.2021.764781
- [85] Mishra S and Ganguli M (2021) Functions of, and replenishment strategies for, chondroitin sulfate in the human body. Drug Discov. Today, 26: 1185–1199. https://doi.org/10.1016/J.DRUDIS.2021.01.029
- [86] Maeda N (2010) Structural variation of chondroitin sulfate and its roles in the central nervous system. CNS Agents Med. Chem., 10: 22–31. https://doi.org/10.2174/187152410790780136
- [87] Maeda N, Ishii M, Nishimura K and Kamimura K (2011) Functions of chondroitin sulfate and heparan sulfate in the developing brain. Neurochem. Res., 36: 1228–1240. https://doi.org/10.1007/S11064-010-0324-Y/FIGURES/4
- [88] Singh JA, Noorbaloochi S, Macdonald R and Maxwell LJ (2015) Chondroitin for osteoarthritis. Cochrane Database Syst. Rev., 1: https://doi.org/10.1002/14651858.CD005614.PUB2
- [89] Kaneiwa T, Mizumoto S, Sugahara K and Yamada S (2010) Identification of human hyaluronidase-4 as a novel chondroitin sulfate hydrolase that preferentially cleaves the galactosaminidic linkage in the trisulfated tetrasaccharide sequence. Glycobiology, 20: 300–309. https://doi.org/10.1093/GLYCOB/CWP174
- [90] Kaneiwa T, Miyazaki A, Kogawa R, Mizumoto S, Sugahara K and Yamada S (2012) Identification of amino acid residues required for the substrate specificity of human and mouse chondroitin sulfate hydrolase (conventional hyaluronidase-4). J. Biol. Chem., 287: 42119–42128. https://doi.org/10.1074/JBC.M112.360693
- [91] Yamada S and Mizumoto S (2022) Characterization of hyaluronidase 4 involved in the catabolism of chondroitin sulfate. Molecules, 27: 6103. https://doi.org/10.3390/MOLECULES27186103
- [92] Gushulak L, Hemming R, Martin D, Seyrantepe V, Pshezhetsky A and Triggs-Raine B (2012) Hyaluronidase 1 and β-hexosaminidase have redundant functions in hyaluronan and chondroitin sulfate degradation. J. Biol. Chem., 287: 16689–16697. https://doi.org/10.1074/JBC.M112.350447
- [93] Barthe L, Woodley J, Lavit M, Przybylski C, Philibert C and Houin G (2004) In vitro intestinal degradation and absorption of chondroitin sulfate, a glycosaminoglycan drug. Arzneimittelforschung, 54: 286–292. https://doi.org/10.1055/S-0031-1296972
- [94] Conte A, Palmieri L, Segnini D and Ronca G (1991) Metabolic fate of partially depolymerized chondroitin sulfate administered to the rat. Drugs Exp. Clin. Res., 17: 27–33.
- [95] Ronca G and Conte A (1993) Metabolic fate of partially depolymerized shark chondroitin sulfate in man. Int. J. Clin. Pharmacol. Res., 13: 27–34.
- [96] Ronca F, Palmieri L, Panicucci P and Ronca G (1998) Anti-inflammatory activity of chondroitin sulfate. Osteoarthr. Cartil., 6: 14–21. https://doi.org/10.1016/S1063-4584(98)80006-X
- [97] Volpi N (2002) Oral bioavailability of chondroitin sulfate (Condrosulf®) and its constituents in healthy male volunteers. Osteoarthr. Cartil., 10: 768–777. https://doi.org/10.1053/JOCA.2002.0824
- [98] Volpi N (2003) Oral absorption and bioavailability of ichthyic origin chondroitin sulfate in healthy male volunteers. Osteoarthr. Cartil., 11: 433–441. https://doi.org/10.1016/S1063-4584(03)00051-7
- [99] Jackson CG, Plaas AH, Sandy JD, Hua C, Kim-Rolands S, Barnhill JG et al. (2010) The human pharmacokinetics of oral ingestion of glucosamine and chondroitin sulfate taken separately or in combination. Osteoarthr. Cartil., 18: 297–302. https://doi.org/10.1016/J.JOCA.2009.10.013
- [100] Tsuchiya Y, Tomita M, Tsuboi M, Takahashi T, Yonezuka M, Kikuchi S et al. (2013) Absorption of proteoglycan via Clathrin-mediated endocytosis in the small intestine of rats. Biosci. Biotechnol. Biochem., 77: 654–656. https://doi.org/10.1271/BBB.120773
- [101] Tan CL, Kwok JCF, Patani R, Ffrench-Constant C, Chandran S and Fawcett JW (2011) Integrin activation promotes axon growth on inhibitory chondroitin sulfate proteoglycans by enhancing integrin signaling. J. Neurosci., 31: 6289. https://doi.org/10.1523/JNEUROSCI.0008-11.2011
- [102] Nadanaka S, Kinouchi H and Kitagawa H (2018) Chondroitin sulfate–mediated N-cadherin/β-catenin signaling is associated with basal-like breast cancer cell invasion. J. Biol. Chem., 293: 444–465. https://doi.org/10.1074/JBC.M117.814509
- [103] Müller S, Schaffer T, Flogerzi B, Fleetwood A, Weimann R, Schoepfer AM et al. (2006) Galectin-3 modulates T cell activity and is reduced in the inflamed intestinal epithelium in IBD. Inflamm. Bowel Dis., 12: 588–597. https://doi.org/10.1097/01.MIB.0000225341.37226.7C
- [104] Talaga ML, Fan N, Fueri AL, Brown RK, Bandyopadhyay P and Dam TK (2016) Multitasking human lectin galectin-3 interacts with sulfated glycosaminoglycans and chondroitin sulfate proteoglycans. Biochemistry, 55: 4541–4551. https://doi.org/10.1021/ACS.BIOCHEM.6B00504/ASSET/IMAGES/MEDIUM/BI-2016-005044_0006.GIF
- [105] Nangia-Makker P, Hogan V and Raz A (2018) Galectin-3 and cancer stemness. Glycobiology, 28: 172–181. https://doi.org/10.1093/GLYCOB/CWY001
- [106] Lima T, Perpétuo L, Henrique R, Fardilha M, Leite-Moreira A, Bastos J et al. (2023) Galectin-3 in prostate cancer and heart diseases: a biomarker for these two frightening pathologies?. Mol. Biol. Rep., 50: 2763–2778. https://doi.org/10.1007/S11033-022-08207-1
- [107] Pudełko A, Wisowski G, Olczyk K and Koźma EM (2019) The dual role of the glycosaminoglycan chondroitin-6-sulfate in the development, progression and metastasis of cancer. FEBS J., 286: 1815–1837. https://doi.org/10.1111/FEBS.14748
- [108] Silver DJ and Silver J (2014) Contributions of chondroitin sulfate proteoglycans to neurodevelopment, injury, and cancer. Curr. Opin. Neurobiol., 27: 171–178. https://doi.org/10.1016/J.CONB.2014.03.016
- [109] Lin X, Sun Y, Yang S, Yu M, Pan L, Yang J et al. (2021) Omentin-1 modulates macrophage function via integrin receptors αvβ3 and αvβ5 and reverses plaque vulnerability in animal models of Atherosclerosis. Front. Cardiovasc. Med., 8: 757926. https://doi.org/10.3389/FCVM.2021.757926
- [110] Cui J, Zhao C, Feng L, Han Y, Du H, Xiao H et al. (2021) Pectins from fruits: Relationships between extraction methods, structural characteristics, and functional properties. Trends Food Sci. Technol., 110: 39–54. https://doi.org/10.1016/J.TIFS.2021.01.077
- [111] Mohnen D (2008) Pectin structure and biosynthesis. Curr. Opin. Plant Biol., 11: 266–277. https://doi.org/10.1016/J.PBI.2008.03.006
- [112] Zhang W, Xu P and Zhang H (2015) Pectin in cancer therapy: A review. Trends Food Sci. Technol., 44: 258–271. https://doi.org/10.1016/J.TIFS.2015.04.001
- [113] Prado SBR Do, Ferreira GF, Harazono Y, Shiga TM, Raz A, Carpita NC et al. (2017) Ripening-induced chemical modifications of papaya pectin inhibit cancer cell proliferation. Sci. Rep. 2017 7:1, 7: 1–17. https://doi.org/10.1038/s41598-017-16709-3.
- [114] Leclere L, Cutsem P Van and Michiels C (2013) Anti-cancer activities of pH- or heat-modified pectin. Front. Pharmacol., 4: https://doi.org/10.3389/FPHAR.2013.00128
- [115] do Prado SBR, Shiga TM, Harazono Y, Hogan VA, Raz A, Carpita NC et al. (2019) Migration and proliferation of cancer cells in culture are differentially affected by molecular size of modified citrus pectin. Carbohydr. Polym., 211: 141–151. https://doi.org/10.1016/J.CARBPOL.2019.02.010
- [116] Nangia-Makker P, Hogan V, Honjo Y, Baccarini S, Tait L, Bresalier R et al. (2002) Inhibition of human cancer cell growth and metastasis in nude mice by oral intake of modified citrus pectin. J. Natl. Cancer. Inst., 94: 1854–1862. https://doi.org/10.1093/JNCI/94.24.1854
- [117] Zhang T, Lan Y, Zheng Y, Liu F, Zhao D, Mayo KH et al. (2016) Identification of the bioactive components from pH-modified citrus pectin and their inhibitory effects on galectin-3 function. Food Hydrocoll., 58: 113–119. https://doi.org/10.1016/J.FOODHYD.2016.02.020
- [118] Beukema M, Faas MM and de Vos P (2020) The effects of different dietary fiber pectin structures on the gastrointestinal immune barrier: impact via gut microbiota and direct effects on immune cells. Exp. Mol. Med., 52: 1364–1376. https://doi.org/10.1038/S12276-020-0449-2
- [119] Cui J, Lian Y, Zhao C, Du H, Han Y, Gao W et al. (2019) Dietary fibers from fruits and vegetables and their health benefits via modulation of gut microbiota. Compr. Rev. Food Sci. Food Saf., 18: 1514–1532. https://doi.org/10.1111/1541-4337.12489
- [120] Masuda R, Ohira N, Kitaguchi K and Yabe T (2024) Novel role of homogalacturonan region of pectin in disrupting the interaction between fibronectin and integrin β1. Carbohydr. Polym., 336: https://doi.org/10.1016/J.CARBPOL.2024.122122
- [121] Barkalow FJ and Schwarzbauer JE (1994) Interactions between fibronectin and chondroitin sulfate are modulated by molecular context. J. Biol. Chem., 269: 3957–3962. https://doi.org/10.1016/S0021-9258(17)41727-3
- [122] Oldberg A and Ruoslahti E (1982) Interactions between chondroitin sulfate proteoglycan, fibronectin, and collagen. J. Biol. Chem., 257: 4859–4863. https://doi.org/10.1016/S0021-9258(18)34604-0
- [123] Hedman K, Christner J, Julkunen I and Vaheri A (1983) Chondroitin sulfate at the plasma membranes of cultured fibroblasts. J. Cell Biol., 97: 1288–1293. https://doi.org/10.1083/JCB.97.4.1288
- [124] Rey FE, Gonzalez MD, Cheng J, Wu M, Ahern PP and Gordon JI (2013) Metabolic niche of a prominent sulfate-reducing human gut bacterium. Proc. Natl. Acad. Sci. U. S. A., 110: 13582–13587. https://doi.org/10.1073/PNAS.1312524110/-/DCSUPPLEMENTAL
- [125] Pichette J, Fynn-Sackey N and Gagnon J (2017) Hydrogen sulfide and sulfate prebiotic stimulates the secretion of GLP-1 and improves glycemia in male mice. Endocrinology, 158: 3416–3425. https://doi.org/10.1210/EN.2017-00391
- [126] Chen L, Gao Y, Zhao Y, Yang G, Wang C, Zhao Z et al. (2022) Chondroitin sulfate stimulates the secretion of H2S by Desulfovibrio to improve insulin sensitivity in NAFLD mice. Int. J. Biol. Macromol., 213: 631–638. https://doi.org/10.1016/J.IJBIOMAC.2022.05.195