Abstract

Chemical structures derived from marine foods are highly diverse and pharmacologically promising. In particular, chitooligosaccharides (COS) present a safe pharmacokinetic profile and a great source of new bioactive polymers. This review describes the antioxidant, anti-inflammatory, and antidiabetic properties of COS from recent publications. Thus, COS constitute an effective agent against oxidative stress, cellular damage, and inflammatory pathogenesis. The mechanisms of action and targeted therapeutic pathways of COS are summarized and discussed. COS may act as antioxidants via their radical scavenging activity and by decreasing oxidative stress markers. The mechanism of COS antidiabetic effect is characterized by an acceleration of pancreatic islets proliferation, an increase in insulin secretion and sensitivity, a reduction of postprandial glucose, and an improvement of glucose uptake. COS upregulate the GLUT2 and inhibit digestive enzyme and glucose transporters. Furthermore, they resulted in reduction of gluconeogenesis and promotion of glucose conversion. On the other hand, the COS decrease inflammatory mediators, suppress the activation of NF-κB, increase the phosphorylation of kinase, and stimulate the proliferation of lymphocytes. Overall, this review brings evidence from experimental data about protective effect of COS.

1. Introduction

Marine sources constitute nearly the half of the nature biodiversity and are considered the largest remaining reservoir of natural structures of food functional interest. Among marine food molecules, chitooligosaccharides (COS) are derivatives of chitosan with a lower molecular weight (MWt) [1]. The bioactivity and physicochemical properties of COS is highly dependent on their chemical characteristics. COS are produced by degradation of either chitin or chitosan via physical hydrolysis, acid hydrolysis, and enzymatic hydrolysis [2]. These oligomers usually present a degree of polymerization (DP) of <50–55 and a molecular weight (MWt) of <10,000 Da [3], and also they are characterized by a degree of deacetylation (DD), which is related to GlcN molar fraction in the compound, MWt distribution (polydispersity), and N-acetylation pattern (PA) [4]. In contrast with chitosan, the COS have higher solubility in water and their viscosity is low, so they are partially soluble in dimethyl sulfoxide and methanol but insoluble in ethanol and acetone [3]. COS have been traditionally obtained by chemical digestion with strong acids. However, many difficulties are associated with the traditional processes. In this regard, the use of chemical, enzymatic, or microbial hydrolysis is commonly employed for COS production. Degradation of chitosan using enzymatic hydrolysis has more benefits than using chemical hydrolysis regarding controllability and predictability [57]. Chitosan may be particularly degraded by chitinases, chitosanases, and lysozymes [3]. Even though chitinases and chitosanases obtained from microbes produce a relatively higher proportion of COS, their usage is limited due to their elevated price. So, many nonspecific enzymes are used, including cellulases, pectinases, lipases, amylases, and deacetylases [2, 8]. Moreover, COS having certain PA pattern may be generated by bacterial deacetylases [9]. COS generated by these enzymes are mainly consisted of a combination of oligomers with different PA, DD, and DP. Recently, COS have received much attention in biomedical and food industries mainly because of their solubility, biocompatibility, biodegradability, and nontoxicity. COS had broad range of applications and proved diverse promising biological activities, and subsequently, they drew attention in drug therapy [6,10]. This review focuses on marine COS as a therapeutic strategy against diabetes, oxidative stress, and inflammation. The pharmacokinetic profile and the molecular and biochemical regulatory pathways of these molecules are discussed with updated bibliographic data.

2. Search Methodology

Electronic searches were performed in PubMed, Scopus, Google Scholar, and Web of Science databases, using the following keywords: chitooligosaccharides, chitosan-oligomers, and chitooligomers, to identify relevant articles. All published articles were evaluated, and a total of 103 papers of important contribution were selected to compose the present review.

3. Results and Discussion

3.1. COS Antioxidant Properties

COS exhibit antioxidant properties in vitro and in vivo; the literature data reporting antioxidant COS are summarized in Tables 1 and 2. COS exhibited strong scavenging activity on the HO and O2 radicals [11]; however, their activity is relatively weak on alkyl and DPPH radicals. In this work, the activity was related to the concentration, MWt, and tested radicals. Haung et al. [12] modified COS into two derivatives by introducing carboxyl and quaternary amino groups to their amino positions with varying substitution levels. The aim of this modification was to change the ability of hydrogen atoms’ whole quantity to react with free radicals and make them more capable to chelate ion metals. COS were also previously prepared from different molecular weights using incomplete chitosan hydrolysis, and their free radical scavenging activity was estimated and correlated to their MWt and compared to aminoglucose [13]. The antioxidant action of COS, vitamin C, and aminoglucose was dose-dependent in a phenazine methosulfate-β-nicotinamide adenine dinucleotide system, though it was nonlinear. Yang et al. [13] suggested that COS amino groups may have an efficient task in their antioxidant action due to their short chains, regarding chitosan which forms well-built intramolecular and intermolecular bonds and reduces the reaction of the free radicals with the amino and hydroxyl groups of the polymers. Further investigation of COS with different MWt (<1000 Da) on intracellular radical (H₂O₂, OH, and ROO) using B16F1 and murine melanoma cell line [14] demonstrated marked decrease in those radical species supporting the concept that a lower MWt is essential for the radical scavenging activity in biological systems. Also, COS low MWt suppressed NF-κB gene promoter activity, proving their ability to avoid diseases associated with oxidative stress. Lee et al. [15] evaluated the protective effects of sulfated chitosan and COS by employing a fluorescence-activated cell sorter (FACS) on hydrogen peroxide and plasmid DNA strand breaks assay. The sulfated chitosan and sulfated COS (S-COS), having highest alkyl radical scavenging activity, were selected to investigate their protective liver cell effect against H2O2. Results proved that the S-COS III showed the highest protective activities on liver cells and exhibited a protective effect on H2O2-induced DNA damage. Fernandes et al. [16] studied the free radical scavenging action of two COS combinations and a low MWt LM-chitosan against two biological molecules, phages and erythrocytes, that underwent different oxidative stress situations. The potential inhibition of COS for each tested concentration was higher than that of LM-chitosan. Higher concentrations of COS potentiated the H2O2 action; in contrast, LM-chitosan potentiated H2O2 effect, at lower concentrations. In addition to the previous mechanisms, either COS or LM-chitosan had the capability to keep RBCs from oxidation of Hb by AAPH at conc. of 0.05–0.005 mg/mL. Fernandes et al. [16] concluded that COS may be utilized as antioxidants in vivo, at concentrations moderately low to stop more oxidative injury to cells, though this was not in agreement with chemical methodology. Ngo et al. [17] studied the effect of gallic acid conjugated to COS (gallate-COS) on cellular antioxidant activity. The mechanism of the antioxidant activity of gallate-COS was assessed by measuring the levels of gene expression of GSH and SOD antioxidant enzymes. Analysis of the results showed that gallate-COS reduced free radical-catalyzed activation of NF-κB protein (this is a transcription factor-mediated expression of several genes in osteoarthritis condition). Accordingly, gallate-COS may be utilized as a potent antioxidant to protect bone cells from oxidative stress. Lu et al. [18] investigated the antioxidative role of S-COS opposing H2O2-mediated injury in pancreatic MIN6 β-cell line. Results revealed that S-COS were safe at conc. of 0.1 to 0.5 mg/mL. Moreover, S-COS can augment the viability of cells, decreasing ROS generation and decreasing the LDH and MDA in oxidative damaged β-cells demonstrating its antioxidant effect. The suggested mechanisms are due to the S-COS improvement of activity of antioxidant enzymes, reduction of ROS generation, and inhibition of MIN6 β-cell apoptosis. Likewise, increasing the DS has an impact on the defense mechanisms against oxidative damage. Many studies estimated the different antioxidant action of LM-polysaccharides (LMPS) prepared by starch (LMST), agar (LMAG), and chitosan (LMCH), on fibroblasts of skin and correlated the activity regarding their amine, hydroxyl, and sulphur groups. The studies reported that the variation in antioxidant activity of LMPS might be because of LMPS functional groups, ability to chelate metal ions, ability to transfer electrons, and capacity to stabilize radicals. This high capacity to stabilize radicals and metal ion chelating property of LMCH resulted in a potent antioxidant action that is advanced to that of LMAG. So, LMCH showed a high ability to reduce ROS generation, reduced lipid peroxidation of the cells, and inhibited DNA damage due to oxidative stress. Accordingly, they reported that LMCH may have an efficient role in treating skin complications and can be used as sun protective agents [19]. Other reports indicated that low MW COS exhibited the highest antioxidant activities [20].

Recently, Qu and Han [39] investigated COS antioxidant action in vivo using high-fat diet (HFD) mice model. Mice groups were administered normal diet, HFD, or HFD + COS (0.5%) for a period of 6 weeks. Group fed with HFD +0.5% COS significantly restored the action of GPx, CAT, and SOD in liver, stomach, and blood when comparing it with the HFD-administered group. Morphologically, there was a significant decrease in the measured parameters: decreased villus height compared with the group fed with HFD. However, those fed with HFD + COS (0.5%) were the same in the height of villus compared with the control. This result supports the improvement in intestinal integrity when COS was added to their diet. Furthermore, results showed that gallate-COS had a defensive potential regarding H2O2-mediated DNA oxidative harm [21]. Gallate-COS reduced the generation of reactive oxygen species inside H2O2-mediated A549 cells. On the other hand, Xie et al. [40] proved that supplementary maternal diet with COS enhanced blood SOD level, which resulted in decreasing MDA content. Also, expression of some antioxidative genes mRNA was enhanced in the mothers’ placenta. Jiang et al. [41] also reported the N-acetyl chitooligosaccharides can attenuate amyloid β-induced damage by reducing the oxidative stress in animal and cell models of Alzheimer’s disease.

3.2. COS Anti-Inflammatory Effect

COS are promising anti-inflammatory agents (Table 3). LM-COS comprising of glucosamine (GlcN) n, where n = 3–5, were fit for restraining both antigen-invigorated degranulation and cytokine production in rodent basophilic leukemia cells (RBL-2H3) [53]. Moreover, the defensive impact of LM-COS regarding ovalbumin-incited lung irritation in asthma mouse model (16 mg/kg/day) was also established [53]. Dung et al. [54] demonstrated that COS upregulated the declarations of CD86 and MHCII on SDCs and advanced the emission of TNF-α. Additionally, nonsoluble COS invigorated the differentiation of the CD4+T in dendritic cells. COS were examined for their consequences for epithelial cells as well as tissues [55]. Chitooligomers fundamentally invigorated the mitochondrial action of in vitro keratinocytes’ cultures. Supplementation with COS significantly reduces the intestinal provocative reaction, which is corresponding with the enactment of CaSR and the suppression of NF-κB signaling pathway under inflammatory conditions [54].

COS produce an anti-inflammatory impact via Nrf2/MAPK-stimulated HO-1 production [56]. Kim et al. [57] showed that LM-S-COS suppressed the generation of nitric oxide and some inflammatory mediators like TNF-α as well as IL-6 in LPS-induced RAW 264.7 cells. Kunanusornchai et al. [58] indicated that COS may have in vitro as well as in vivo synovial anti-inflammatory activity through AMPK phosphorylation. Moreover, COS showed suppressor action on LPS-stimulated reduction in the Bcl-2/Bax fraction and higher activity of caspase-3 and BKCa [42]. One of the techniques that COS can perform to suppress apoptosis of cells is by controlling BKCa channel. Then again, COS can hinder the activation of LPS-prompted p38 as well as quicken the activation of O-GlcNAc glycosyltransferase enzyme. Li et al. [59] found that five chitooligomers from dimer to hexamer (chitobiose, chitotriose, chitotetraose, chitopentaose, and chitohexaose) separated from COS enhanced NF-κB-related luciferase gene expression as well as inhibited NF-κB gene transcription. COS reduced OGT-related NF-κB O-GlcNAcylation and so inhibited LPS-mediated inflammation of the vascular endothelia [60]. Polar chitosan formulation based on nanoparticles CMC-COS NP and SC-COS NP was produced by the development of complexes of polyelectrolyte [61]. Injection of CMC-COS NP and SC-COS NP changed Th cytokine content in blood and induced the differentiation of lymphocytes in spleen in vivo, approving their ability to control cell-induced immune reactions. COS efficiently suppressed TNF-α-mediated activation of ICAM-1 and VCAM-1 at transcription as well as translation levels [62]. Liu et al. [62] found that COS suppressed LPS-mediated IL-8 activation in HUVECs via blocking p38 MAPK as well as PI3K/Akt pathways. COS decreased the generation of proinflammatory mediators such as IL-1β and nitric oxide in LPS-mediated RAW 264.7 cells [63]. COS may also defend inflammation and oxidative stress in LPS-mediated complications in vivo, which may present valuable properties for septic patients [63]. Results showed that COS decreased the contents of nitric oxide and PGE2 generation by inhibiting COX-2 and iNOS activation without considerable cytotoxicity in BV-2 microglia [64]. Notably, COS exerted anti-inflammatory effect through blockade of IκB-a decomposition, NF-κB translocation, and MAPK phosphorylation (dose dependent). Xu et al. [65] demonstrated that COS suppressed EGF-mediated cell migration via blocking EGFR/MAPK pathway. COS suppressed the proliferation of epithelia cells of the breast via deactivation of GnT-V as well as its products [66]. Nutritional supplements containing COS or GMOS increased IL-1β gene activation of mucosa in jejuna as well as lymph nodes, when compared to lincomycin supplements [67]. Yoon et al. [68] showed that COS can act as an anti-inflammatory agent through stimulating TNF-α in LPS-mediated inflammatory complications of RAW 264.7 cells. COS may also be efficient in treating inflammatory bowel disease via inhibiting NF-κB pathway as well as apoptosis of cells of epithelia in intestine [69]. Zhang et al. [70] found that COS may possess potential stimulating incidence in the immune system by enhancing the production of TLR4 on macrophage while Zhang et al. [71] found that they efficiently suppresses the IL-1β-mediated chondrocytes’ apoptosis through activation of p38 MAPK pathway. Dai et al. reported protective effect of COS against neuroinflammation via its inhibitory effect on BACE1 [22]. Moreover, COS have been shown to exhibit immunostimulatory effect via the MAPK and PI3K/Akt signaling pathways [72]. Another study demonstrated that fully deacetylated and acetylated COS exhibit an ability to reduce the level of TNF-α in murine macrophages after LPS stimulation [73].

3.3. COS Antidiabetic Mechanisms

COS exert its antidiabetic activity through different mechanisms (Table 4). In a study made by Liu et al. [82], the highly deacetylated COS (90%) of molecular weight 1200 Da obtained by chitosan enzymatic degradation using chitosanase enzyme. COS incidence on pancreatic islet cell viability as well as β-cell line using MTT colorimetric test was established. The COS sufficient entrance into cells of pancreatic islet was approved by COS-induced insulin secretion assays through utilizing cultures of rat monolayer islet cells, where COS at a concentration (100 mg/L) enhanced the differentiation of islets and induced recovery of damaged pancreatic β-cells increasing insulin release to 14 days, when comparing the results to the group of normal control. COS also promoted β-cell proliferation in pancreas (INS-1) and upregulated GLUT2 mRNA gene expression, which could stimulate insulin release. COS also can defend INS-1 cell against apoptosis [83] which occurs as a result of increasing the circulating glucose, saturated FA [84, 85], inflammatory markers [86], and oxidative stress [8789].

Streptozotocin (STZ) is a molecule that may act as selective cytotoxic agent for pancreatic cells as well as generating ROS that results in DNA degradation [90, 91]. COS showed a protective effect on INS-1 cell in opposition to apoptosis mediated by STZ, through raising the activity of reduced SOD and reduction of elevated levels of malondialdehyde (MDA) content in pancreas homogenate [82, 83, 92]. On the other hand, COS may not be able to defend INS-1 cell against harm mediated by some proinflammatory cytokines, such as IFN-γ, TNF-α, or IL-1β that can enhance β-cell apoptosis via the intrinsic mitochondrial apoptotic pathway [93].

In vivo experiments in STZ-induced diabetes mellitus (DM) in mice showed that COS could decrease 2 h plasma glucose in 60 days at a dose of 500 mg/kg, to 16.14 mmol/L, when comparing the results to the diseased DM group at . This study also proved that the dose 500 mg/kg of COS also showed the best incidence in the oral glucose tolerance test (OGTT) through improving the sensitivity to insulin. Area under the curve (AUC) for the DM group was regarded as 100%, and AUC of normal control group was only calculated as 17.69% of the DM group AUC, so treatment with 500 mg/kg of chitooligosaccharides caused a decrease to 68.69% of the DM group AUC [82]. STZ-mediated type 2 DM results in insulin resistance as well as β-cell disordering [94]. In a study designed by Ju et al. [83], STZ-diabetic rats showed metabolic disorders including hyperglycemia, hyperlipidemia, insulin resistance, and ruptured islet. COS showed in vivo hypoglycemic effect at a dose of 1000 mg/kg, and it exerts its effect by reducing fasting serum glucose as well as insulin levels, increasing index of insulin sensitivity and improving OGT, protecting pancreatic islet, and reducing insulin resistance. COS also caused a considerable increase in glycogen level in liver, and this might be through increasing the transfer of blood glucose into liver glycogen by increasing glucokinase.

Chronic hyperglycemia may be acquired as a result of the failure of the body peripheral tissues to utilize glucose properly. GLUT-4 is a vital glucose transporter and regulator of its metabolism found in skeletal muscles and adipocytes [95, 96]. In this study, the expression of GLUT-4 was decreased in adipose and skeletal tissues in diabetic animal model. Treatment of the diabetic animals with COS caused a significant increase in GLUT-4; this effect may be attributed to COS which can upregulate GLUT-4 mRNA expression, thus improving insulin resistance. In vivo administration of COS in STZ-diabetic rats could also repair the injury of pancreatic tissues and restore the BWt ratio of pancreas. Liu et al. [82] suggested that COS being an alkaline help in decreasing the plasma glucose through increasing the pH of the body fluids and thus raise insulin sensitization. COS may also maintain normal metabolism of plasma glucose by regulating the endocrine system as well as reducing insulin secretion to standard levels. Administration of COS to alloxan-induced type II diabetes mellitus in mice at two doses 5 and 10 mg/kg decreased plasma glucose levels by 54.1% on day 21st of the experiment, significantly increased hepatic glycogen content, and gradually increased the body weight of the treated mice, while the diabetic mice continued to lose weight [97]. SGOT and SGPT activities were increased significantly in alloxan-induced diabetic mice [97]. This increased activity of transaminases occurs in the absence of insulin due to the increase of amino acids levels in the blood causing increased gluconeogenesis and ketogenesis [97]. SGOT and SGPT which act as indicators of liver function were come towards control level after treatment with COS (10 mg/kg); this indicates the normal liver function [97] COS (10 mg/kg) have also caused alteration in lipid metabolism and this was evidenced by decreasing the levels of serum LDL, VLDL, triglycerides, and total cholesterol levels which were elevated due to diabetes and increased HDL-c level [97]. Interestingly, these results from Ha et al. [98] study suggest that deep-sea water having COS showed a higher inducible result on glucose uptake more than either alone. Also, this consequence is accomplished by activating different signaling pathways associated with GSV trafficking. Yu et al. [99] showed that COS may have dual mechanism of actions as antidiabetic in intestinal cell lines, either through suppressing α-glucosidase and SGLT1 as well as GLUT2, or through improving adipocyte proliferation and activation of PPARγ as well as its targeted genes, like FABP4 and adiponectin, where these properties were enhanced through cotreating with BADGE (PPARγ antagonist). Also, COS considerably enhanced glucose uptake. In vivo COS administration did not demonstrate any antidiabetic/hypocholesterolemic effects, since glycemia and cholesterol levels in GK rats were not altered [100]. In a recent study [101], the consumption of COS with complexes to resistant starch produced a higher capability for insulin sensitivity revival. Gene expression of liver tissues obtained from COS-injected rats approved that this effect may be due to increase in the liver glucose alteration through activation of GS2 as well as GYG1, decrease in gluconeogenesis through activation of G6PC1, and change in glucolipid metabolism through activation of Insig-2. Also GlcNAc 2 decreased lipid peroxidation as well as inflammatory markers in pancreatic cells with enhancement of SOD activity and decreased IL-1b, NF-κB, MDA, and TNF-α contents [102]. GlcNAc 2 may also appreciably deactivate MAPK pathway particularly through IL-1b-Erk/p38-histone H3 pathway in type 2 DM in vivo.

4. Current Evidence and Perspectives

Marine compounds with interesting in vivo antioxidant properties are excellent drug candidates against inflammation, diabetes, and oxidative stress. COS especially show interesting properties in experimental data. COS are generated from chitin deacetylation and hydrolysis reactions, through enzymatic or chemical methods. The COS are normally absorbed via the epithelia of the intestine and mainly dispersed to the hepatocytes, splenocytes, and renal cells. COS are also decomposed by lysozymes before being excreted via urine. Due to their high polarity, low viscosity, safety, and good pharmacokinetic profile, COS have already been integrated in many functional formulations, such as COS capsules. It is noteworthy that, among the several publications that focuses on COS bioactivity, many recent works detailed the molecular mechanisms and targeted signaling pathways of COS action. However, many mechanisms are not evidenced, mainly because of poor reproducibility, divergence in physicochemical properties, and dose effect. Among the proposed mechanisms are, e.g., the pancreatic islet proliferation, the increase in insulin secretion and sensitivity, the reduced postprandial glucose and improved glucose uptake, the upregulatory GLUT2 expression and inhibition of digestive enzymes and glucose transporters, the reduction of gluconeogenesis and promotion of glucose conversion, the decrease in inflammatory mediators and enzymes, and the suppression of NF-κB pathway. Moreover, COS stimulate the proliferation of lymphocytes and radical scavenging activity and decrease ROS and reduce MDA in oxidative damaged cells. COS have been shown to increase intracellular antioxidant enzymes, prevent oxidative DNA damage, and inhibit or suppress superoxide anion and NO production. Interestingly, the bioactivity of COS is highly dependent on its physicochemical properties, which may result in new findings and bioactive compounds according to extraction and purification method and/or molecular weights. Therefore, optimization and controllable methods of extraction and polymer synthesis will contribute to reproduce relevant therapeutic effects. For example, decrease in COS MWt is related to their high antioxidative effects. Similarly, increases in DD and positive charges are related to their high anti-inflammatory as well as antioxidative activities. Chemical modifications of COS present a chance for enhancing different biological activities of COS. Future research should be directed towards the understanding of mechanism of action of standardized and chemically characterized COS. All together experimental data and processing technology provide an evidence of defensive potential of COS against glucose-mediated oxidative complications through being in vivo antioxidant, anti-inflammatory, and antidiabetic agents; COS can also regulate many signaling pathways involved in glucose metabolism and homeostasis. The safe profile of COS, diverse mechanism, and MWt-dependent bioactivity lead us to expect a wide range of applications for COS technological formulations.

Conflicts of Interest

The authors declare that there are no conflicts of interest regarding the publication of this paper.

Acknowledgments

This research was made possible as part of Ministry of Agriculture Malaysia initiative under the New Key Economic Areas Entry Point Project on High Value Herbals awarded to Natural Wellness Biotech (M) Sdn Bhd. The authors express their gratitude and appreciation for the trust and opportunity given.