Next Article in Journal
Extended PRF: Impact of Heat on Gene Expression in Gingival Fibroblasts
Previous Article in Journal
Protective Effects of Methanolic Extract of Micromeria frivaldszkyana (Degen) Velen Against Acetaminophen-Induced Liver Toxicity in Male Wistar Rats
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Potential of Natural Products in Hangeshashinto Water Extract on the Direct Suppression of Stomatitis Induced by Intra-/Extracellular Advanced Glycation End-Products

1
Division of Molecular and Genetic Biology, Department of Life Science, Medical Research Institute, Kanazawa Medical University, Uchinada 920-0293, Ishikawa, Japan
2
Department of Pharmacy, Kanazawa Medical University Hospital, Uchinada 920-0293, Ishikawa, Japan
3
Department of General Internal Medicine, Kanazawa Medical University, Uchinada 920-0293, Ishikawa, Japan
4
General Medical Center, Kanazawa Medical University Hospital, Uchinada 920-0293, Ishikawa, Japan
5
Department of Urology, Kanazawa Medical University Hospital, Uchinada 920-0393, Ishikawa, Japan
6
Inoue Iin Clinic, Kusatsu 525-0034, Shiga, Japan
7
Department of Pathology and Laboratory Medicine, Kanazawa Medical University, Uchinada 920-0293, Ishikawa, Japan
8
Department of Pathology, Kanazawa Medical University Hospital, Uchinada 920-0293, Ishikawa, Japan
9
Department of Spleen and Stomach Diseases, First Affiliated Hospital of Hebei University of Chinese Medicine, Shijiazhuang 050011, China
10
Hebei Key Laboratory of Turbidity Toxin Syndrome, Shijiazhuang 050011, China
11
Research Center, First Affiliated Hospital of Hebei University of Chinese Medicine, Shijiazhuang 050011, China
12
Department of Immunology, Kanazawa Medical University, Uchinada 920-0293, Ishikawa, Japan
13
Department of Oral and Maxillofacial Surgery, Kanazawa Medical University, Uchinada 920-0293, Ishikawa, Japan
14
Department of Internal Medicine, Fukui Saiseikai Hospital, Wadanaka 918-8503, Fukui, Japan
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2025, 26(18), 9118; https://doi.org/10.3390/ijms26189118
Submission received: 20 August 2025 / Revised: 9 September 2025 / Accepted: 16 September 2025 / Published: 18 September 2025
(This article belongs to the Section Bioactives and Nutraceuticals)

Abstract

Oral mucositis (stomatitis) is a painful condition that affects the mouth lining. Kampo medicines (e.g., Hangeshashinto [Chinese name, Ban-Xia-Xie-Xin-Tang], Orento, and Orengedokuto) have been widely used to treat stomatitis, such as gargling with Hangeshashinto. However, the mechanisms by which Kampo medicines work are not widely understood due to their oral administration and the subsequent digestion, absorption, and metabolization of their components. Stomatitis is associated with advanced glycation end-products (AGEs) in patients with lifestyle diseases, and can be induced by both intra- and extracellular AGEs (blood and dietary AGEs). Various natural products inhibit intracellular AGE generation and suppress cytotoxicity, such as inflammation caused by extracellular AGEs. This review summarizes 19 natural products identified in the Hangeshashinto water extract and 16 natural products identified in the crude drug extract. The data show that several natural products, such as glycyrrhizin, baicalin, 6-shogaol, quercetin, epigallocatechin-3-galate, and genistein, inhibit intracellular AGEs and suppress extracellular AGE inflammation. Furthermore, several natural products in the Hangeshashito water extract can suppress cytotoxicity in stomatitis.

1. Introduction

Stomatitis is a painful condition that affects the mouth lining, and there are numerous types and causes, such as traumatic stomatitis [1], aphthous stomatitis [2], and chemotherapy-/radiotherapy-induced stomatitis [3,4]. Aphthous stomatitis is diagnosed in the clinical stage when the cause of stomatitis has not been determined. The proliferation of viruses and bacteria, exposure to cytotoxic materials such as nicotine, and other lifestyle factors can cause stomatitis [5], indicating that it is associated with lifestyle-related diseases (LSRDs). Kampo medicines, which are based on traditional Chinese medicines, such as Hangeshashinto (Chinese name, Ban-Xia-Xie-Xin-Tang) [5,6,7,8], Orento [5,9], Orengedokuto [5,9], Inchinkoto [5,9], Byakkokaninjinto [5], Juzentaihoto [5], Hochuekkiito [5], and Shosaikoto, have been widely used to treat and prevent stomatitis [5]. In Japan, Hangeshashinto is widely used to treat stomatitis, gastrointestinal dysfunction, and nausea [5,6,7,8,9]. Data elucidating the effects and side effects of Kampo medicines have been accumulating for more than two thousand years. However, the mechanisms of Kampo medicines have not yet been fully elucidated due to the high number of natural products contained in each Kampo medicine and the digestion, absorption, and metabolization of natural products by oral administration [10]. While the oral administration of Hangeshashinto to patients with stomatitis, gut dysfunction, and nausea is described in traditional Chinese medicine [11,12,13,14,15], the treatment and prevention strategy for stomatitis described in Kampo is gargling and coating of the mouth with the Hangeshashinto water extract [16,17,18,19]. When gargled or coated on the mouth, natural products in the Hangeshinto water extract may directly protect the oral epithelial cells, thus avoiding digestion, absorption, and metabolism pathways [10]. Oh et al. and Endo et al. analyzed the components in the Hangeshashinto water extract using three-dimensional high-performance liquid chromatography (3D-HPLC) [6,20], and 19 natural products, such as liquiritigenin [21], glycyrrhizin [22], baicalin [23], and 6-shogaol [24,25], were identified. Because these natural products inhibit the production of prostaglandin E2 (PGE2), they can be used as a direct protection of oral epithelial cells [21,22,23,24,25]. In this review, we have focused on advanced glycation end-products (AGEs) associated with LSRDs, including diabetes mellitus (DM), cardiovascular disease (CVD), hypertension, fatty liver, and stomatitis [26,27,28,29]. AGEs originate from saccharides (e.g., glucose and fructose), their metabolites, and non-enzymatic production (e.g., methylglyoxal, glyoxal, and glyceraldehyde) [26,27,28,29]. These compounds react with proteins to produce AGEs; however, methylglyoxal, glyoxal, and glyceraldehyde can also be produced from lipids such as glycerol. Consequently, the definition of AGEs may be modified in the future [26,27,28,29]. Three major free types of AGE, Nε-carboxymethyl–lysine (CML), Nε-carboxyethyl–lysine (CEL), and Nδ-(5-hydro-5-methyl-4-imidazolone-2-yl)-ornithine (methylglyoxal-hydro-imidazolone, MG-H1), which are modified proteins, have been reported. These AGE-modified proteins are generated in the human body, and their dysfunction might affect various LSRDs [26,27,28,29]. Intracellular AGEs can be generated and accumulate in oral epithelial cells and esophageal and gastric epithelial cells [30,31,32]. Consequently, intracellular AGEs may induce diseases such as stomatitis. In contrast, extracellular AGEs, which are leaked into body fluids such as blood, urine, and saliva from various cells that produce intracellular and dietary AGEs, can induce cytotoxicity via receptors for AGEs (RAGE) and toll-like receptor 4 (TLR4) [26,27,28,29]. The major dietary AGEs are CML, CEL, and MG-H1 [10,27,28,29]. RAGE and TLR4 are expressed on oral epithelial cells and other cells in various organs [33,34,35]. Extracellular AGEs can be transported and expressed on oral epithelial cells, and the AGEs-RAGE/TLR4 axis induces cytotoxicity such as inflammation [33,34,35]. Extracellular AGEs can cause stomatitis; consequently, this review has focused on natural compounds that can suppress stomatitis. Specifically, the natural products in Hangeshashinto water extract are reviewed, and examples such as liquiritigenin [36], glycyrrhizin [37], baicalin [38,39], and 6-shogol [40,41,42], which inhibit the production of AGEs in vitro and suppress the AGEs-RAGE/TLR4 axis signaling, are discussed. The literature on natural products in seven crude drugs (Hange, Ogon, Oren, Ninjin, Kankyo, Taiso, and Kanzo), which are contained in Hangeshashinto, has also been reviewed. We discuss the natural products in each crude drug, such as quercetin [43], epigallocatechin-3-gallate [44], genistein [45], jasmonic acid [46], and gallic acid [47]. The relationship between each compound that occurs or promotes LSRDs and Kampo medicines remains unclear, although the effects of Kampo medicines have been revealed [10].

2. Various Types of Stomatitis

Stomatitis can be caused by various factors (Figure 1). Traumatic stomatitis can be caused by a traumatic physical stimulation, such as a burn [1,48], which causes oral epithelial cells to peel off. In contrast, aphthous stomatitis is characterized by ulcers, insomnia, psychological stress, and other factors, such as the excessive intake of saccharides and lipids, and we predict that AGEs specifically can induce stomatitis (See Section 6). Although the mechanisms of aphthous stomatitis remain unclear [2,49], diagnosis has been performed when an obvious factor in the patient’s mouth or other organs is not observed. Viruses and bacteria can cause herpetic and candidal stomatitis [50,51,52,53]; however, inflammation is usually induced to inhibit their proliferation. Protection against virus and bacteria proliferation is associated with lifestyle, and related stomatitis may be due to LSRDs. Nicotine is one of the main chemicals in all forms of tobacco [54,55]. Nicotine stomatitis, a LSRD, is characterized by an oral lesion, usually located on the hard palate, which develops due to the heat and chemical irritation caused by tobacco products. Vitamin B1 deficiency inhibits the recovery of oral epithelial cells against damage and induces stomatitis [56]. Furthermore, vitamins B2, B6, and B12 are required to produce proteins in oral epithelial cells and maintain metabolism, and thus, deficiencies in these vitamins may also induce stomatitis [56,57]. We believe that cases of stomatitis induced by B1, B2, B6, and B12 deficiencies should be classified as LSRDs. Chemotherapy and radiation treatments for cancer suppress the oral epithelial cell cycle, and consequently, damaged cells can induce inflammation leading to stomatitis [3,4]. Furthermore, suppression of the immune system in oral epithelial cells can also promote the proliferation of viruses and bacteria, which can also lead to stomatitis. Because chemotherapy and radiation do not occur in daily life, these cases are not classified as LSRDs [3,4,8,15].

3. Treatment of Stomatitis with Hangeshashinto and Predicted Mechanisms

3.1. Traditional Chinese and Kampo Medicines

To describe the Hangeshashinto treatment for stomatitis, we first introduce the relationship between traditional Chinese medical science and Kampo, which is traditional Japanese medicine [58,59,60,61,62]. Traditional Chinese medical science, which includes health preservation (healthy diet and exercise) [63], diagnosis [62,64,65], moxibustion [66], cupping [67,68], acupuncture [69,70,71], and crude drugs (natural medicines) [6,64,65,72,73,74], has been used for approximately 3000 years. The practices migrated to Japan during the ancient and middle periods, directly from the Chinese mainland via the Korean peninsula. They were adapted to the Japanese population, land, and medical plants, and renamed Kampo [59,60,61,62]. “Kam” or “Kan” in Japanese means “Han” in Chinese, referring to the ancient Chinese empire (from the third-century BC to the third-century AD), and “po” refers to the method of diagnosis and treatment (Figure 2a). Various traditional Chinese medicines involving plants, animal tissues, and minerals are used in Kampo [59,60,61,62,75,76]. Ban-Xia-Xie-Xin-Tang, a set of seven crude drugs, was one of the treatments adopted by Kampo, and referred to as Hangeshashinto (Table 1) [5,6,7,8,11]. In traditional Chinese medical science, Ban-Xia-Xie-Xin-Tang is orally administered, used as a gargle, and as a coating on the oral squamous [11]. The gargle and coating onto the oral squamous cells of Hangeshathinto are described in the Japanese Pharmacopoeia, which includes registered Kampo medicines (Figure 2b) [5,6,7,8]. This review has focused on the use of Hangeshashinto as a gargle and coating on oral squamous cells (see Section 3.2).

3.2. Hangeshashinto (Ban-Xia-Xie-Xin-Tang) Treatment Effects and Mechanisms

Many Kampo medicines are orally administered water extracts from crude drugs [73,74]. This includes the Hangeshashinto water extract, which is a dry powder (dry powder quality is regulated in the Japanese Pharmacopoeia) used to treat oral squamous cells, gut dysfunction, dyspepsia, vomiting, and nausea [5,6,7,8,11]. Hangeshashinto water extract has anti-oxidation, anti-inflammation, anti-bacteria, tissue recovery, pain relief, and intestinal bacteria induction effects [5,6,7,8,11,16,77]. However, the detailed mechanism has not yet been revealed because its various natural products are digested, absorbed, and metabolized in the body’s organs [10]. We predicted that the extract components would directly induce anti-oxidation, anti-inflammation, anti-bacterial, and pain relief when used as a gargle and coating for oral epithelial cells; however, this will require further investigation to elucidate fully [6,7,10,16,77]. Oh et al. reported that Hangeshashinto suppressed the expression of interleukin (IL)-6 and 8 in CAL27 (a human oral epithelial cell line) treated with Porphyromonas gingivalis pathogen-associated molecular pattern (PAMP) [6]. Hato et al. revealed that Hangeshashinto suppressed the expression of IL-1α and human β-defensin 1 in normal human oral keratinocytes, which were stimulated with lipopolysaccharide [7]. Hangeshasnito inhibited cyclooxygenase (COX)-2 and suppressed the production of prostaglandin E2 (PGE2) [16]. A scratch and cell migration test using normal human oral keratinocytes by Uezono et al. revealed that Hangeshashinto induced the secretion of the C-X-C motif chemokine ligand (CXCL) 12 from cells, and secreted CXCL 12 combined with C-X-C chemokine receptor 4 (CXCR4) to promote migration [77]. This effect was mediated by the phosphorylation of extracellular signal-regulated kinase (ERK), and the activity of ERK was associated with increased CXCL12 expression.

3.3. Natural Products in Hangeshashinto and Their Anti-Inflammation Effects

Oh et al. and Endo et al. analyzed the components in Hangeshashinto water extract using 3D-HPLC and identified approximately twenty natural products [6,20]. They classified flavonoid, chalcone, alkaloid, and gingerol groups. The flavonoids included liquiritin [6], liquiritin apioside [6], wogonin [6,20], wogonin-7-O-glucuronide [6], baicalin [20], orxylin A [6], and orxylin-7-O-glucuronide [6]. The chalcones included isoquritin [6], isoliquiritin apioside [6], and isoquitigenin [6]. The alkaloids included palmatine [6], berberine [6], epiberberine [6], jateprrhizine [6], coptisine [6], and magnoflorine [6]. The triterpene group included glycyrrhizin (glycyrric acid) [6,22]. The gingerol group (one of the monophenolic acid groups) included 6-gingerol [20] and 6-shougaol [6]. The structures of these natural products are presented in Figure 3, Figure 4, Figure 5 and Figure 6. Because the Hangeshashinto water extract inhibited COX-2 and suppressed the production of PGE2 [16], we surveyed reports on natural products that show inhibition of COX-2 and suppression of PGE2 production. Liquirtin [78], liquiritin apioside [79], liquiritigenin [21], worgonin [16,80], baicalin (baicalein-7-O-glucuronide) [16,23,80], orxylin A [81], isoquritin [82], isoquitigenin [82], palmatine [83], berberine [84], epiberberine [85], coptisine [86], glycyrrhizin [22], magnoflorine [87], 6-gingerol [16,24,88], and 6-shougaol [16,24,25] can inhibit COX-2 and/or suppress the production of PGE2. Because the Hangeshashinto water extract contains a high concentration of natural compounds, it cannot be suggested with certainty that these identified compounds are the sole cause of the inflammatory effects [6,20]. However, these results may help elucidate the mechanisms of Hangeshashinto.

4. AGEs

4.1. AGE Origins

Saccharides, saccharide metabolites, and their non-enzymatic reaction products react with proteins to generate AGEs via the Maillard reaction [26,27,28,29,89,90,91,92]. We introduce glucose [26,27,28,29,93,94], fructose [26,27,28,29,93,94], melibiose [95], and ribose [96,97] as major saccharides (Figure 7), and glyceraldehyde, glycolaldehyde, methylglyoxal, glyoxal, and 3-deoxyglucosone as metabolites and non-enzymatic reaction products of saccharides (Figure 8) [26,27,28,29]. Because fructose can be generated from glucose via the polyol pathway and methylglyoxal can be produced from glyoxal, the production routes of AGEs are intertwined [26,27,28,29]. However, we should consider the definition of AGEs because some AGEs may be advanced lipoxidation end-products (ALEs) [29]. AGEs can also be produced from lipids, such as methylglyoxal, which can be made from glycerin [98,99].

4.2. Free AGEs

While AGEs can react with proteins to generate AGEs via the Maillard reaction, they can also react with amino acids to produce free-type AGEs (Figure 9 and Figure 10) [26,27,28,29]. We introduced AGEs that were detected and identified in human and/or animal tissues/cultured cells, and body fluids, such as blood, saliva, and urine (Figure 9) [26,27,28,29,100,101,102,103], and AGEs synthesized in a tube [104], whose structure had been hypothesized [105] (Figure 10). Although the AGEs in Figure 9 are the major free types, CML, CEL, MG-H1, methylglyoxal-derived imidazolium cross-link (methylglyoxal–lysine dimer, MOLD), and pentosidine in the blood and urine have been selected as biomarkers for LSRDs or a dairy diet (see Section 4.5.5). Shigeta et al. reported that pyrrolopyridinium–lysine dimer-derived glyceraldehyde 1 and 2 (PPG1 and PPG2) were synthesized from glyceraldehyde and Nα-acetyl-lysine in the tube [104]. However, there are no reports that PPG1 and PPG2 were detected in vivo. Takeuchi et al. hypothesized the structure of two glyceraldehyde-derived AGEs, which they named “Toxic AGEs (TAGE)” in 2004; however, the structure has not been proved [105].

4.3. Previous AGE Categories and Potential Improvements

In the middle of the 20th century, a classical category for AGEs was established [26,27,28,29]. Glucose-, glyceraldehyde-, glycolaldehyde-, methylglyoxal-, glyoxal-, and 3-deoxyglusone-derived AGEs were named based on their original compounds as follows: AGE-1, -2, -3, -4, -5, and -6 [26,27,28,29]. However, Litwinowicz et al. detected and quantified melibiose-derived AGEs (MAGE) and determined that MAGE should be categorized as AGE-10 [95]. In contrast, one AGE can be generated from various origin compounds, such as CML, which is generated and produced from glucose, ribose, glycolaldehyde, and glyoxal (Figure 11) [28,96,97]. In the future, AGE categories based on the original compounds may be improved. Some researchers have suggested a unique category based on cytotoxicity and/or LSRDs [105,106,107,108]. Takeuchi et al. named a GA-AGE which was recognized by a polyclonal antibody they prepared, as “TAGE” in 2004; however, the structure emains unclear [105]. Shmkova et al. focused on the pridinium moiety of both glyceraldehyde- and glycolaldehyde-derived AGEs and named them as TAGE [106]. Shen et al. suggested that glyceraldehyde-, glycolaldehyde-, methylglyoxal-, and 3-deoxyglucosone-derived AGEs should be categorized as TAGE because they could directly induce cell damage [107]. Furthermore, Lee et al. used a cytotoxicity analysis to determine that MOLD is a typical TAGE [108].

4.4. Structure of AGE-Modified Proteins

AGEs can exist as AGE-modified proteins or animals in the organs [109,110,111,112] and body fluids of humans and animals [113,114,115]. Various methods have been used to identify the structures of AGE-modified proteins, including electrospray ionization–mass spectrometry (ESI-MS) and matrix-assisted laser desorption/ionization MS (MALDI-MS) analysis (see Section 4.6) [109,110,111,112,116,117]. Some AGEs, including free types, can be modified into one molecular protein, as shown in Figure 12a [27,28,29]. Because free-type AGEs generally have modified lysine or arginine residues in amino acid sequences, the location is limited compared with the number of whole amino acids. Although ESI-/MALDI-MS can be used to elucidate this structure, the operation is complex because the peptide that contains more than two free-type AGEs must be detected and identified [27,28,29]. In contrast, free-type AGEs, which include more than two lysine and/or arginine residues, show the potential of the free-type AGE structure in terms of intra-/intermolecular covalent bonding for proteins (Figure 12b) [27,28,29]. In this structure, we believe ESI-/MALDI-MS analysis should be used for identification; however, we understand the limitations of MS technology. The protein database of the ESI-/MALDI-MS equipment cannot be used to detect the structures of peptides that originate from complex proteins combined with free-type AGEs [27,28,29]. In contrast, researchers cannot prove this structure with only anti-AGE antibody analysis. If an anti-AGE antibody recognizes a protein epitope, it does not confirm the difficulty of the inter- or intramolecular covalent bonding capacity [27,28,29,118] (Figure 12b).

4.5. Intra-/Extracellular AGEs and LSRDs

4.5.1. Intracellular AGEs and LSRDs

AGEs are generated from saccharides, their metabolites, and non-enzymatic production in the cultured cells [116,119]. Sanavirathna et al. analyzed intracellular AGEs in a pancreatic ductal cell line (PANC-1) treated with glyceraldehyde, and identified argpirimidine-, MG-H1-, and GLAP-modified proteins [116]. The functions of these proteins may be affected by AGE modification. Takahashi et al. reported that intracellular CML-modified proteins increased in a human proximal tubular cell line (HK-2), which was incubated in a high glucose medium, had an Atg5 knockdown, and lysosome function was suppressed by intracellular glucose-derived AGEs [119]. Although their results were in vitro, they indicate a relationship between intracellular AGEs and LSRDs. In contrast, various AGEs (e.g., MG-H1, G-H1-modified proteins) were detected in cardiac tissues of patients (age >75 years) or patients with diabetes mellitus [109,110,111]. These AGEs may suppress cardiomyocyte function to induce CVD. The skeletal muscle in the obese mouse that ingests high-fat and high-saccharide foods accumulated CML- and CEL-modified proteins, and they were associated with adipose degeneration of skeletal muscle [120]. Furthermore, the accumulation of CML and CEL may induce sarcopenia.

4.5.2. AGEs in the Extracellular Matrix and LSRDs

Pentosidine-modified collagens are AGE-modified extracellular matrix proteins [29,121,122]. Intracellular collagens may be modified with free-type AGEs, such as pentosidine, and then secreted and released. Extracellular AGE modification may also occur. Pentosidine-modified collagen showed the dysfunction and may induce [29,121,122].

4.5.3. AGEs in the Blood, Urine, Saliva, and LSRDs

AGEs, including free types, and AGE-modified proteins have been detected in blood [29,123,124,125], urine [29,126], and saliva [29,101]. Many researchers believe that AGEs in the body fluid leak into the blood and lymph vessels, and renal tubules from cells where intracellular AGEs were generated [29]. However, we suggest that AGEs may be generated in the blood/lymph vessel and renal tubule due to the detection of glyceraldehyde [127,128], glycolaldehyde [129], methylglyoxal [130], and glyoxal [130] in the blood. Because these compounds rapidly react with amino acids or proteins, some AGEs may be generated in the blood/lymph vessels and renal tubules. Furthermore, AGEs in foods and beverages have been reported as dietary AGEs (see Section 4.5.4). Various organs, such as the liver, heart, lung, gut, and oral squamous cells, express RAGE and TLR4; AGEs-RAGE/TLR4 signaling can induce dysfunction and cytotoxicity, such as excess inflammation [33,34,35,131,132,133,134]. In contrast, Wang et al. reported that glyceraldehyde-derived AGEs-modified bovine serum albumin, which was the model of AGEs and might contain various AGEs in the blood, induced dysfunction of cardiomyocytes via the suppression of ryanodine receptor 2 activity [135]. Lee et al. revealed that glyoxal-derived imidazolium cross-link (glyoxal–lysine dimer, GOLD) and MOLD induced oxidative damage and inflammation by interacting with RAGE [108,136].

4.5.4. Dietary AGEs and LSRDs

Various types of saccharides are contained in foods and beverages, and they are processed using heat treatments in factories and homes [28,89,133,137,138]. While multiple types of AGEs (free-type AGEs and AGE-modified proteins) can be produced, we acknowledge that CML, CEL, and MG-H1 are widely produced in many foods and beverages [89,133]. Many researchers believe that the AGEs detected in the body fluid will include dietary AGEs, and the AGEs-RAGE/TLR4 axis induces inflammation in various organs, promoting LSRDs such as cancer and gut ulcer [133,137].

4.5.5. AGEs in the Body Fluid as a Biomarker for LSRDs and Dietary Lifestyle

To diagnose LSRDS, such as non-alcoholic heptosteatosis (NASH), hyperuricemia, high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C), uric acid, creatinine, IL-1β, IL-6, IL-10, and urinary pH, have been measured in the blood and urine [139,140]. Because AGEs in the blood, urine, and saliva can be detected in humans and animals, various researchers have suggested their use as LSRD [29,95,114,115,140,141] and dietary lifestyle biomarkers [137,138]. Litwinowicz et al. suggested that MAGE in the blood could be a beneficial biomarker for alcoholic hepatitis [95] and insisted that it should be categorized as AGE-10 [95]. CML and MG-H1 are reduced in the blood after kidney transplantation and can thus be used as biomarkers for kidney function [114]. Kato et al. reported that MG-H1, but not CML, CEL, and Nε-carboxymethyl–lysine (CMA), was elevated in the blood of patients with nephropathy [141]. CML and CEL in the blood increased in obesity model mice fed high-fat and high-sugar diets [114], and various types of AGEs (each structure was not identified) in the blood of NASH model mice were also increased [142]. In contrast, CML, CEL, and MG-H1 in the blood reflect the intake of dietary AGEs and can be used as biomarkers for dietary lifestyle [137,138].

4.6. Identification and Quantification of AGEs

We introduce fluorescence, immunostaining, slot blotting, Western blotting, ELISA, GC-MS, ESI-/MALDI-MS, and nuclear magnetic resonance (NMR) as methods to identify and quantify AGEs [26,27,28,29]. AGEs are generally excited at a wavelength of approximately 370 nm, and fluorescence is emitted at approximately 440 nm [143,144]. Although fluorescence cannot be used to differentiate between different AGE structures, it can be used to quantify AGEs in the human skin and blood under non-invasive conditions, and is a beneficial analysis for clinical operations [144]. To analyze basic AGE research, HPLC and fluorescence can be combined, as well as hydrophilic interaction liquid chromatography (HILIC) [145]. Immunostaining, slot blotting, Western blotting, and ELISA analysis generally require anti-AGE antibodies [26,27,28,29]. Although immunostaining, slot blotting, and ELISA can be used to quantify AGEs, they cannot be used to analyze their molecular structures [26,27,28,29]. In the quantification with slot blot analysis, we believed that Takata’s method is beneficial because their lysis buffer (or modified Takata’s lysis buffer) promotes suitable probing of AGE-modified proteins onto polyvinylidene difluoride (PVDF) membranes [26,146,147,148,149]. This method was selected in thirteen studies from 2017 to 2025. Some researchers analyzed fluorescence to quantify AGEs using immunostaining and the ELISA method without using anti-AGE antibodies [117,150]. Western blotting analysis of AGEs can detect various AGE proteins in cell lysates, tissue lysates, and body fluids on the PVDF membrane [120,149]. To quantify AGEs with slot blotting and ELISA, AGE-modified proteins are required, not general recombinant proteins [26,146,147,148,151]. GC-MS [26,152,153,154,155], ESI-MS [26,89,111,123,124,141,156,157], and MALDI-MS [26,89,111,123,124,141,156,157] were used to analyze the mass of free-type AGEs and AGE-modified peptides (Figure 10 and Figure 12). Because free-type AGEs (e.g., CML and CEL) are non-volatile compounds, they should be esterified [26,152,153,154,155]. We believe that ESI- and MALDI-MS analyses are the most suitable for proving that free-type AGEs are modified on one molecular protein and that AGEs with two amino acid residue (e.g., pentosidine, MOLD, GOLD) link proteins with intra- and/or intermolecular covalent bonds, because methods that use fluorescence and anti-AGE antibodies are ineffective (Figure 10 and Figure 12) [26,109,110,111,118]. NMR is used for AGE identification, not quantification [26,28].

5. Mechanisms of Inhibition of Intra-/Extracellular AGE-Induced Cytotoxicity by Anti-AGE Compounds

Numerous strategies have been reported for the inhibition of intra-/extracellular AGE-induced cytotoxicity. The carbonyl trap system and activation of glyoxalase-1 can inhibit the generation of intracellular AGEs (Figure 13) [27,28,29]. The former can directly trap the origins of AGEs, which include ketone and aldehyde groups (e.g., glucose, fructose, glyceraldehyde, glycolaldehyde, methylglyoxal, and glyoxal), and reduce the effects of methylglyoxal and glyoxal because they are metabolized by glyoxalase-1 (Figure 7, Figure 8 and Figure 13). Researchers have reported that various compounds show a carbonyl trap system and activation of glyoxalase-1 [27,28,29]. Another method that has been investigated involves inhibiting glucose transport into cells. However, glucose and its metabolites/non-enzymatic products can be generated in cells via the glycolysis and peroxidation of lipids if glucose is not transported. Although AGEs can be degraded by autophagy and the ubiquitin–proteasome system [27,28,29], our understanding of the natural compounds that promote these mechanisms remains unclear. In contrast, anti-AGE compounds may block AGEs-RAGE/TLR4 because they combine with RAGE/TLR4 as antagonists and inhibit the cell signaling pathway (Figure 13) [27,28,29].

6. Potential of AGE-Induced Cytotoxicity and Dysfunction for Oral Squamous Cells

6.1. Potential of Intracellular AGE-Induced Cytotoxicity and Dysfunction for Oral Epithelial Cells

Oral epithelial cells are located on the oral squamous and typically undergo rapid turnover within 7–14 days [158]. Although intracellular AGEs have not been identified or quantified in oral epithelial cells, existing information about esophageal and gastric epithelial cells indicates that they can be generated and accumulate in oral epithelial cells [10,30,31,32,159]. Esophageal cells are remarkably similar to oral epithelial cells, and Yokoyama et al. reported that the condition of oral epithelial cells may reflect the risk of esophageal epithelial cells [159]. In contrast, intracellular AGEs such as CML and CEL in the esophageal epithelial cells increased in Goto–Kakizaki rats (DM model rats) [31]. The turnover period for gastric epithelial cells is similar to that of oral epithelial cells (7–14 days) [10]. Wang et al. revealed that intracellular AGEs were generated and accumulated to induce damage in gastric epithelial cancer cells and promote tumor formation [32]. Oya-Ito et al. reported that MGO-AGE-modified HSP27 was increased in RGK-1 cells (human gastric epithelial cell line) incubated with high glucose medium, and this MGO-AGE modification may be associated with the phosphorylation of HSP27 [30]. Because phosphorylation regulates the activity of HSP27, which suppresses apoptotic proteins and works as a chaperone system, MGO-AGE modifications may affect these functions [30]. Because glucose and fructose can be transported into oral epithelial cells as well as esophageal and gastric epithelial cells, we believe that intracellular AGEs can be generated and accumulate in oral epithelial cells. The phenomenon of intracellular AGEs in the oral epithelial cells should be further investigated to identify novel oral squamous cell types.

6.2. Potential of Extracellular AGE-Induced Cytotoxicity and Dysfunction for Oral Epithelial Cells

RAGE and TLR4 are expressed on oral epithelial cells as well as other organs [10,33,34,35,132]. Because AGEs in the body fluid (e.g., blood and saliva) can directly combine with RAGE and TLR4, AGEs-RAGE/TLR4 signaling can be induced. To date, there are no reports analyzing extracellular AGE-RAGE/TLR4 signaling in vitro or in vivo in oral epithelial cells. Many researchers have tried to elucidate how inflammatory proteins such as high-mobility group box 1 (HMGB-1) and lipopolysaccharides (LPS) induce inflammation and cell dysfunction via the RAGE/TLR4 to investigate inflammation in oral squamous [10,33,34,35,132]. However, inflammation stimulated by the AGEs-RAGE/TLL4 axis can be induced on oral epithelial cells as well as other cells in the liver, lung, gut, and small/large intestine. Oral epithelial cells are more directly exposed to AGEs in saliva and dietary AGEs than the cells in other organs. Oral squamous cells that induce extracellular AGEs-RAGE/TLR4 should be further researched, as well as the involved inflammatory proteins and lipopolysaccharides.

7. Natural Products in Hangeshashinto Water Extract Inhibit the Generation of Intracellular AGEs in Stomatitis

Some researchers have suggested inhibiting the generation of intracellular AGEs [28,121,149,160,161,162] and their degradation via the autophagy and ubiquitin–proteasome system [163,164]. To inhibit the generation of intracellular AGEs, the carbonyl trap system can be used against α-carbonyl compounds, the precursors of AGEs, such as glucose, fructose, glyceraldehyde, methylglyoxal, glyoxal, and 3-deoxyglucosone (Figure 7, Figure 8 and Figure 13). Furthermore, glyoxalase-1, which can metabolize methylglyoxal and glyoxal (Figure 8 and Figure 13), can suppress the transportation of glucose into cells [28,121,160,161,162,165,166]. Although the structures of the natural products that show carbonyl trap systems remain unclear, flavonoid skeletal compounds (Figure 3) [28,160,161], chalcone skeletal compounds (Figure 4) [28,160,161], alkaloid (Figure 5) [28,160], resveratrol compounds [28,167], and p-coumaric acid, which is similar to 6-gengerol and 6-shogaol in Figure 6, exhibit carbonyl trap effects [28,168]. Froldi et al. found that each glucose, glyoxal, and ribose was incubated with bovine serum albumin (BSA) in the tube. They also investigated the inhibition effects of baicalin for each glucose-, glyoxal-, and ribose-derived BSA [169]. Baicalin can inhibit the production of each AGE-modified BSA. Although Froldi et al. did not suggest whether these effects were induced by carbonyl trapping, we consider it because baicalin is a flavonoid skeletal compound. Furthermore, baicalin shows anti-α-glucosidase activity, which inhibits the production of glucose from polysaccharides and glycoproteins, and reduces the potential that glucose and its intermediates react with amino acid (e.g., lysine, arginine) residues in proteins [169]. Wang et al. incubated glucose and beef enzymatic treatment solution in a tube and quantified CML and CEL [36]. In this study, liquiritigenin, liquiritin, and glycyrrhizin inhibited the generation of both CML and CEL. We predicted that they would show the carbonyl trapping. In contrast, they were able to reduce the production of both methylglyoxal and glyoxal, and these results suggest that they activated glyoxalase-1 [36]. Carnovali et al. reported that the blood glucose level was reduced with the liquiritigenin treatment [170]. Liquiritigenin can inhibit the generation of intracellular AGEs in three ways [36,170]. Alvi et al. reported that glycyrrhizin blocked ribose-derived BSA generation in the tube where ribose and BSA were incubated [37]. Glycyrrhizin belongs to the triterpene structure compound, which contains one carboxylic acid and is glycosylated. The structure may directly inhibit the production of AGEs, though the mechanism remains unclear. Ahmad et al. reported that berberine inhibited the methylglyoxal reaction with human serum albumin (HAS) in a tube [171]. However, glycyrrhizin suppressed glyoxalase-1 in Plasmodium falciparum, but its activation up-regulated expression in rats [172,173,174]. Because glycyrrihizin was orally administered and may have been digested, absorbed, and metabolized in previous investigations [173,174], the effects of glycyrrihizin on the oral epithelial cells remain unclear. Oral epithelial cells treated with glycyrrihizin in vitro or in vivo must be investigated further to elucidate this issue. Both 6-gingerol and 6-shogaol were introduced as natural products as the methylglyoxal-trapping materials, and they inhibited the generation of methylglyoxal-modified proteins and free AGEs, which were derived from methylglyoxal [40,41,175]. To inhibit the generation of intracellular AGEs in the oral epithelial cells, natural products need to directly show their function because target cells are exposed [10]. If natural products inhibit or suppress the generation or accumulation of intracellular AGEs in vivo, then their metabolism in the small/large intestine may have the same effect. However, we believe that natural products which inhibit the function or generation of intracellular AGEs in vitro, such as baicalin [169], liquiritigenin [36,170], liquiritin [36], glycyrrhizin [36,37], berberine [171], 6-gengerol [40,41], and 6-shogol [40,41,175], should be investigated further as treatment and prevention strategies for stomatitis because they can directly affect for the oral epithelial cells.

8. Natural Products Obtained from Hangeshashinto Water Extract Suppress Extracellular AGEs-RAGE/TLR4 Signaling in Stomatitis

Baicalin may suppress the AGE-RAGE signaling pathway in vitro and in vivo (Figure 3) [38,39]. Qui et al. reported that baicalin suppressed AGE-RAGE signaling in a DM animal model treated with streptozotocin [38]. In contrast, Fu et al. isolated and cultured porcine aortic vascular endothelial (PAVE) cells from ten 30-day-old naturally farrowed, caryly weaned piglets [39]. Baicalin suppresses RAGE signaling to modulate apoptosis, and blocking RAGE signaling may be effective when extracellular AGEs are agonists. Liquiritin attenuates AGEs-RAGE/NF-κβ in human umbilical vein endothelial cells in vitro (Figure 3) [176]. In contrast, liquiritin apioside and wogonin regulate the AGE-RAGE signaling pathway in diabetic mice (Figure 3) [177,178]. They were metabolized in various organs and bacteria following oral administration. However, the results indicate that liquiritin, apioside, and worgonin obtained from Hangeshashinto water extract may suppress AGE-RAGE signaling in oral epithelial cells. Isoliquiritigenin suppresses the AGE-RAGE signaling pathway in vitro and in vivo (Figure 4) [179,180]. Isoliquiritigenin was previously shown to ameliorate the toxicity of extracellular AGEs on cultured human renal proximal tubular epithelial cells [179]. This function of isoliquiritigenin may translate to oral epithelial cells, providing a preventive strategy for stomatitis. Because isoliquiririgenin was administered orally for db/db mice in the latter investigation, the resulting metabolites may inhibit the AGE-RAGE axis [180]. Shi et al. did not indicate if isoliquiritigenin or its metabolites exhibit this function; however, their investigation introduced the potential that isoliquiritigenin may suppress the AGE-RAGE axis in stomatitis. Goto et al. reported that berberine improves the high-mobility group box-1 (HMGB1)-RAGE or -TLR4 axis in a rat cardiomyocyte cell line (H9C2) in vitro (Figure 5) [181]. HMGB1 and other proteins can activate RAGE and TLR4 to induce inflammation and cell dysfunction, and AGE-modified proteins are contained within their agonists. In contrast, Jiang et al. reported that berberine modulated AGE-induced ferroptosis in both human keratinocyte cell lines (HaCaT cells) and the skin of db/db mice [182]. We predict that berberine will modulate AGE-induced ferroptosis in oral epithelial cells and their metabolites, which may exhibit similar effects. Fan et al. reported that glycyrrhizin inhibits the HMGB1-RAGE axis in various tumor cells (Figure 6) [183]. While Fan et al. did not suggest that glycyrrihizin blocked the AGEs-RAGE axis, it may show this function because RAGE can combine various types of AGE-modified proteins. Furthermore, 6-shogaol inhibited AGE-induced IL-6 and intracellular adhesion molecule 1 (ICAM1) expression on human gingival fibroblasts in vitro. The 6-shogaol suppressed AGE-induced RAGE expression and activation of MAPKs/NF-κβ signaling (Figure 6) [42]. We believe that 6-shogaol inhibits AGE-RAGE signaling, but the effect may not be direct, because the ratio of the AGE-RAGE combination may be reduced.

9. Beneficial Natural Products in Seven Crude Drugs Obtained from Hangeshashinto

Plant leaves, blanch, roots, seeds, and seed vessels contain high levels of natural products [184,185]. The various solvents (e.g., water, water/methanol, ethanol, acetone) that can be extracted from plants also contain natural products; however, they have not yet been fully elucidated. Oh et al. and Endo et al. identified approximately 20 components in Hangeshashinto water extract using 3D-HPLC [6,20]. The identified compounds are not necessarily present in high concentrations in the extract. Because researchers generally divide individual natural products using the retention times and absorption/excitation wavelengths with 3D-HPLC, the compounds whose retention times and wavelengths are different from those of other compounds are more likely to be isolated. The natural products in each crude drug (tuber of Pinellia ternate Breitenbach, Araceae; root of Scutellaria baicalensis Georgi, Labiatae; rhizome of Coptis japonica Makino, Ranueculaceae; steamed rhizome of Zingiber officinale Roscoe, Zingiberaceae; root of Panax ginseng C.A. Meyer, Araliaceae; the root or stolon of Glycyrrhiza uralensis Fischer, Laguminosae; and fruit of Zizpbus jujura Miller var. inermis Pehder, Rhamnaceae) obtained from the Hangeshashinto water extract must be further investigated (Table 1) [186]. We introduce natural products obtained from each extract of crude drug in Hangeshashinto, which were extracted with various solvent such as water, ethanol, and methanol, and review their functions, including the inhibition of the generation of intracellular AGEs, and suppression of extracellular AGEs-RAGE/TLR4 signaling (see Section 10 and Section 11), using the “Standards of Reporting Kampo Products (STORK)” database which was established by Japanese researchers [186].

10. Natural Products in the Crude Drugs Obtained from Hangeshashinto That Inhibit the Generation of Intracellular AGEs

Natural products in plants can suppress the generation of intracellular AGEs. We introduced quercetin, chrysin, genistein, (+)-catechin, (−)-epicatechin, epigarocatechin-3-gallate, hesperidin, imperialine, curcumin, piperine, diphylorethohydroxyarmalol, and resveratrol in Figure 14 [28,121,160,161,187]. These natural products are involved in the carbonyl trap system, where they remove AGE-origin compounds [28,121,160,161,187]. In contrast, curcumin and resveratrol inhibit glyoxalase-1 to promote the generation of glyoxal- and methylglyoxal-derived AGEs. However, the carbonyl trap system can trap them to suppress the generation of AGEs [162]. Although the main effects of curcumin and resveratrol for oral epithelial cells remain unclear, we believe that they should be considered as candidates for natural products for the suppression of AGE-induced cytotoxicity. We reviewed whether these natural products were detected and isolated in each crude drug obtained from Hangeshathinto (Table 2) [188,189,190,191,192,193,194,195,196,197,198,199,200,201,202,203,204,205,206].

11. Natural Products in Crude Drugs from Hangeshashinto Suppress Extracellular AGEs-RAGE/TLR4 Signaling

We introduce ferulic acid [214], caffeic acid [215], gallica acid [216,217], luteolin [218], apigenin [219,220], fisetin [221], naringenin [222], and naringin (naringenin-7-O-rhamunoglucoside) [223], which are natural products that have been obtained from various plants and can suppress/attenuate extracellular AGEs-RAGE/TLR4 signaling and cytotoxicity (Figure 15). Although quercetin and p-coumaric acid were introduced as natural products that inhibited the generation of intracellular AGEs in Section 10 (Figure 14a,g), both quercetin [224,225,226,227,228,229,230] and p-coumaric acid [231,232,233,234] were able to suppress extracellular AGE-induced cytotoxicity via RAGE/TLR4. We investigated the natural products in each crude drug obtained from Hangeshashinto and introduced this information in Table 3 [235,236,237,238,239,240,241,242,243,244,245,246]. Naringenin is an aglycone type, and naringin is a glycosylation type of naringenin [223]. Although naringenin and naringin have been isolated from various citruses [247,248], we cannot observe a reference that naringin was isolated in seven crude drugs in Hangeshashinto. However, naringin-4‘-O-glucoside was isolated from Glycyriza glabra, and it is expected that naringin also will be isolated due to their structural similarities [249].

12. Limitations

While it is likely that AGE-induced oral squamous cell syndromes occur as a result of modern lifestyles, we targeted the intracellular AGEs in the oral epithelial cells and extracellular AGEs that directly combine with the surface. AGEs in other organs can promote cytotoxicity for oral epithelial cells via inflammation and saliva dysfunction; however, we were unable to review this phenomenon. Various natural products are generally contained in the roots, leaves, radix, and seeds of the plants [10,184,185]. Although the analysis of the Hangeshashinto water extract shows that it contains high amounts of natural products, we have only discussed nineteen compounds, and we did not present the ratio of these compounds in the water extract weight (e.g., μg/g dry weight). Therefore, we cannot accurately assess their bioactivity for anti-intra-/extracellular cytotoxicity in oral epithelial cells. Moreover, we did not review whether whole natural products show anti-intracellular and extracellular AGE-induced cytotoxicity in oral epithelial cells because these issues will be addressed as detection and isolation technologies develop for natural products in the Hangeshashinto extract. In contrast, we reviewed some natural products that show anti-AGE-induced cytotoxicity in seven crude drugs in Hangeshashinto. These natural products are typical compounds for anti-AGE materials in various plants. If we introduce the major compounds in each crude drug, then Section 9 and Section 10 would contain details for over 200 compounds [186], which is beyond the scope of this review. Therefore, we introduced some natural products that show anti-AGE-induced cytotoxicity and are contained in various plants, and reviewed whether they are included in each crude drug in Section 9 and Section 10. This information should suggest the possibility that more natural products, which suppress the AGE-induced cytotoxicity in oral epithelial cells, will be isolated in the Hangeshashinto water extract.

13. Conclusions

While intra-/extracellular AGEs may induce oral squamous cell syndromes, these syndromes have not been recognized in clinical or basic medical science. Hangeshashinto treatments are generally administered orally, and the mechanisms by which they attenuate the mouth, esophagus, and gut have been widely investigated. This review focused on treatments of oral epithelial cells that involve gargling and coating the mouth with Hangeshashinto water extract. Nineteen natural products that can suppress the generation of intracellular AGEs and extracellular AGEs-RAGE/TLR4 signaling have been isolated from the Hangeshashinto water extract. In addition, sixteen natural compounds in seven crude drugs obtained from Hangeshashinto may show anti-intra-/extracellular AGE effects. To inhibit the generation of various types of AGE-modified proteins, the carbonyl trap system and activation of glyoxalase-1, which are natural products in the water extract of Hangeshashinto or seven crude drugs, are beneficial because they can react with AGE-origin compounds. Although they can suppress the signaling of AGEs in the body fluid or the beverages/foods and RAGE/TLR4 in oral epithelial cells, the possibility that they can suppress various or whole types of AGEs-RAGE/TLR4 signaling remains unclear. Each natural compound may suppress an individual structure in the AGE-RAGE/TLR4 axis; this will require further investigation. Natural products in Hangeshashinto water extract may directly prevent and modulate stomatitis induced by intra-/extracellular AGEs. This review will be beneficial for future investigations that aim to reveal the mechanisms by which the Hangeshashinto water extract attenuates AGE-induced stomatitis.

Author Contributions

Conceptualization, T.T., J.M., K.M., S.Y. and Y.M.; Methodology, T.T., J.M., K.M., S.Y., Q.Y., N.O., H.N. and Y.M.; Writing—Original Draft Preparation, T.T., J.M., K.M., S.I., S.Y., J.H., Q.Y., X.G., S.M., T.N., N.O., H.N., T.M. and Y.M.; Writing—Review and Editing, T.T., J.M., K.M., S.I., S.Y., J.H., Q.Y., X.G., S.M., T.N., N.O., H.N., T.M. and Y.M.; Visualization for Figure 2, T.T., J.H., Q.Y., X.G., and Y.M.; Visualization for Figures without Figure 2, T.T., J.M., K.M., S.I., S.Y., S.M., T.N., N.O., H.N., T.M. and Y.M.; Visualization for Tables, T.T., J.M., K.M., S.Y. and Y.M. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by JSPS KAKENHI [grant number JP24K14802 (T.T.)].

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available in the article.

Acknowledgments

The references were reviewed and formatted by Sumie Saito from the Department of Urology at Kanazawa Medical University (Uchinada, Japan).

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
AGEAdvanced glycation end-product
ALEAdvanced lipoxidation end-product
BSABovine serum albumin
CELNε-carboxyethyl–lysine
CMLNε-carboxymethyl–lysine
COXCyclooxygenase
CXCLC-X-C motif chemokine ligand
CXCRC-X-C chemokine receptor 4
CVDCardiovascular disease
DODIC3-Deoxyglucosone-derived imidazolium cross-link
3D-HPLCThree-dimensional high-performance liquid chromatography
DMDiabetes mellitus
ELISAEnzyme-linked immunosorbent assay
ERKExtracellular signal-regulated kinase
ESIElectrospray ionization
ESI-MSElectrospray ionization–mass spectrometry
GAGlyceraldehyde
GCGas chromatography
GC–MSGas chromatography–mass spectrometry
G-H1Glyoxal-hydro-imidazolone
GLAPGlyceraldehyde-derived pyridinium
GODICGlyoxal-derived imidazolium cross-link
HASHuman serum albumin
HDL-CHigh-density lipoprotein cholesterol
HILICHydrophilic interaction liquid chromatography
HMGB1High-mobility group box-1
HPLCHigh-performance liquid chromatography
HSP90Heat shock protein 90
ILInterleukin
LDL-CLow-density lipoprotein cholesterol
LSRDLifestyle-related disease
MAGEMelibiose-derived advanced glycation end-product
MALDIMatrix-assisted laser desorption/ionization
MG-H1Methylglyoxal-hydro-imidazolone
MODICMethylglyoxal-derived imidazolium cross-link
MOLDMethylglyoxal–lysine dimer
MSMass spectrometry
MyD88Myeloid differentiation factor
NASHNon-alcoholic heptosteatosis
NF-κβNuclear factor-κβ
NMRNuclear magnetic resonance
PGE2Prostaglandin E2
PPGPyrrolopyridinium–lysine dimer-derived glyceraldehyde
PVDFPolyvinylidene fluoride
RAGEReceptor for advanced glycation end-product
ROSReactive oxygen species
sRAGESoluble receptor for advanced glycation end-product
SGLT1Sodium-glucose cotransporter 1
TAGEToxic advanced glycation end-product
TCMTraditional Chinese medicine
TLR4Toll-like receptor 4
TCMSTraditional Chinese medical science
TNF-αTumor necrosis factor alpha

References

  1. Horvat, A.L.; Prpić, J.; Muhvić, U.M.; Pezelj-Ribarić, S.; Ivančić-Jokić, N.; Peršić, B.R.; Aleksijević, M.; Glažar, I. Oral Mucosal Lesions in Childhood. Dent. J. 2022, 10, 214. [Google Scholar] [CrossRef]
  2. Ashari, K.A.; Parvaneh, N.; Mirnia, K.; Ayati, M.; Saeedi, M.; Salehzaden, F.; Shahrooei, M.; Sangsari, R.; Rohani, P.; Ziaee, V. Three cases of autoinflammatory disease with novel NLRC4 mutations, and the first mutation reported in the CARD domain of NLRC4 associated with autoinflammatory infantile enterocolitis (AIFEC). Pediatr. Rheumatol. 2024, 22, 90. [Google Scholar] [CrossRef] [PubMed]
  3. Zhu, Z.; Pan, W.; Ming, X.; Wu, J.; Zhang, X.; Miao, J.; Cui, W. The effect of probiotics on severe oral mucositis in cancer patients undergoing chemotherapy and/or radiotherapy: A meta-analysis. J. Stomatol. Oral Maxillofac. Surg. 2024, 125, 101983. [Google Scholar] [CrossRef] [PubMed]
  4. Mahakunakorn, P.; Sangchart, P.; Panyatip, P.; Ratha, J.; Damrongrungruang, T.; Priprem, A.; Puthongking, P. In vitro cytoprotective and in vivo anti-oral mucositis effects of melatonin and its derivatives. PeerJ. 2024, 12, e17608. [Google Scholar] [CrossRef] [PubMed]
  5. Sunagawa, M.; Yamaguchi, K.; Tsukada, M.; Ebihara, N.; Ikemoto, H.; Hisamitsu, T. Kampo (Traditional Japanese Herbal) Formulae for Treatment of Stomatitis and Oral Mucositis. Medicines 2018, 10, 130. [Google Scholar] [CrossRef]
  6. Oh, H.; Makita, Y.; Masuno, K.; Imamura, Y. Hangeshashinto Inhibits Porphyromonas gingivalis Pathogen-Associated Molecular Patterns-Mediated IL-6 and IL-8 Production through Toll-Like Receptors in CAL27 Cells. Evid.-Based Complement. Altern. Med. 2024, 2024, 9866670. [Google Scholar] [CrossRef]
  7. Hato, H.; Kaneko, A.; Maeda, C.; Sakata, K.; Ono, Y.; Mizukami, Y.; Kono, T.; Kitagawa, Y. Comparison between hangeshashinto and dexamethasone for IL-1α and β-defensin 1 production by human oral keratinocytes. J. Oral Biosci. 2024, 66, 188–195. [Google Scholar] [CrossRef]
  8. Chan, W.J.; Adiwidjaja, J.; Mclachlen, A.; Boddy, A.V.; Haenett, J.E. Interactions between natural products and cancer treatments: Underlying mechanisms and clinical importance. Cancer Chemother. Pharmacol. 2023, 91, 103–119. [Google Scholar] [CrossRef]
  9. Nishimura, M.; Taniguchi, S.; Tamaoki, S.; Fujita, T. Inhibition of compound action potentials in the frog sciatic nerve by inchinkoto, a traditional Japanese medicine used for oral mucositis. J. Oral. Biosci. 2024, 66, 420–429. [Google Scholar] [CrossRef]
  10. Takata, T.; Motoo, Y. Novel In Vitro Assay of the Effects of Kampo Medicines against Intra/Extracellular Advanced Glycation End-Products in Oral, Esophageal, and Gastric Epithelial Cells. Metabolites 2023, 13, 878. [Google Scholar] [CrossRef]
  11. Kim, Y.H.; Kim, J.Y.; Kwon, O.; Jung, S.Y.; Joung, J.; Yang, C.S.; Lee, J.; Cho, J.; Son, C. Efficacy of a Traditional Herbal Formula, Banha-Sasim-Tang in Functional Dyspepsia Classified as Excess Pattern. Front. Pharmacol. 2021, 12, 698887. [Google Scholar] [CrossRef]
  12. Nishiwaki, R.; Inoue, Y.; Sugao, M.; Sugimasa, N.; Hamaguchi, T.; Noji, M.; Takeuchi, K.; Ito, Y.; Kato, T.; Yasuma, T.; et al. Hangeshashinto-Associated Mesenteric Phlebosclerosis and Highly Atypical Adenoma Requiring Laparoscopic Right Hemicolectomy. Diagnostics 2024, 14, 565. [Google Scholar] [CrossRef]
  13. Murai, T.; Matsuo, M.; Tanaka, H.; Manabe, Y.; Takaoka, T.; Hachiya, K.; Yamaguchi, T.; Otsuka, S.; Shibamoto, Y. Efficacy of herbal medicine TJ-14 for acute radiation-induced enteritis: A multi-institutional prospective Phase II trial. J. Radiat. Res. 2020, 61, 140–145. [Google Scholar] [CrossRef]
  14. Wang, Y.; Ren, Y.; Xiao, C.; Liu, H.; Fu, X.; You, F. Hangeshashinto for preventing oral mucositis in patients receiving cancer treatment: Protocol for a systematic review and meta-analysis. BMJ Open 2021, 11, e047627. [Google Scholar] [CrossRef]
  15. Kato, T.; Sakaguchi, H. Efficacy of Cryotherapy and Hangeshashinto for Radiation-induced Oral Stomatitis: Preliminary Study. In Vivo 2023, 37, 830–835. [Google Scholar] [CrossRef] [PubMed]
  16. Kono, T.; Kaneko, A.; Matsumoto, C.; Miyagi, C.; Ohbuchi, K.; Mozuhara, Y.; Miyano, K.; Uezono, Y. Multitargeted effects of hangeshashinto for treatment of chemotherapy-induced oral mucositis on inducible prostaglandin E2 production in human oral keratinocytes. Integra. Cancer Ther. 2014, 13, 435–445. [Google Scholar] [CrossRef] [PubMed]
  17. Fukamichi, H.; Matsumoto, C.; Omiya, Y.; Arimoto, T.; Morisaki, H.; Kataoka, H.; Kadena, M.; Funatsu, T.; Fukutake, M.; Kase, Y.; et al. Effects of Hangeshashinto on Growth of Oral Microorganisms. Evid.-Based Complement. Alternat. Med. 2015, 2015, 512947. [Google Scholar] [CrossRef] [PubMed]
  18. Ozawa, N.; Onda, T.; Hayashi, K.; Honda, G.; Shibahara, T. Effects of Topical Hangeshashinto (TJ-14) on Chemotherapy-Induced Oral Mucositis. Cancer Manag. Res. 2020, 12, 1069–1078. [Google Scholar] [CrossRef]
  19. Ogihara, T.; Kagawa, M.; Yamanaka, R.; Imai, S.; Itohara, K.; Hira, D.; Nakagawa, S.; Yonezawa, A.; Ito, M.; Nakagawa, T.; et al. Preparation and pharmaceutical properties of Hangeshashinto oral ointment and its safety and efficacy in Syrian hamsters with 5-fluorouracil-induced oral mucositis. J. Nat. Med. 2023, 77, 53–63, Erratum in J. Nat. Med. 2024, 78, 803. [Google Scholar] [CrossRef]
  20. Endo, M.; Oikawa, T.; Tonooka, M.; Hanawa, T.; Odaguchi, H.; Hori, M. Hangekobokuto, a traditional Japanese herbal medicine, ameliorates postoperative ileus through its anti-inflammatory action. J. Smooth Muscle Res. 2022, 58, 78–88. [Google Scholar] [CrossRef]
  21. Tu, C.; Ma, Y.; Song, M.; Yan, J.; Xiao, Y.; Wu, H. Liquiritigenin inhibits IL-1β-induced inflammation and cartilage matrix degradation in rat chondrocytes. Eur. J. Pharmacol. 2019, 858, 172445. [Google Scholar] [CrossRef]
  22. Zhou, S.; Liu, G.; Si, Z.; Yu, L.; Hou, L. Glycyrrhizin, an HMGB1 inhibitor, Suppresses Interleukin-1β-Induced Inflammatory Responses in Chondrocytes from Patients with Osteoarthritis. Cartilage 2020, 13, 947S–955S. [Google Scholar] [CrossRef]
  23. Li, B.; Wang, M.; Chen, S.; Li, M.; Zeng, J.; Wu, S.; Tu, Y.; Li, Y.; Zhang, R.; Huang, F.; et al. Baicalin Mitigates the Neuroinflammation through the TLR4/MyD88/NF-κB and MAPK Pathways in LPS-Stimulated BV-2 Microglia. Biomed. Res. Int. 2022, 2022, 3263446. [Google Scholar] [CrossRef]
  24. Eren, D.; Betul, Y.M. Revealing the effect of 6-gingerol, 6-shogaol and curcumin on mPGES-1, GSK-3β and β-catenin pathway in A549 cell line. Chem. Biol. Interact. 2016, 258, 257–265. [Google Scholar] [CrossRef]
  25. Wang, D.; Jiang, Y.; Yang, X.; Wei, Q.; Wang, H. 6-Shogaol reduces progression of experimental endometriosis in vivo and in vitro via regulation of VGEF and inhibition of COX-2 and PGE2-mediated inflammatory responses. Korean J. Physiol. Pharmacol. 2018, 22, 627–636. [Google Scholar] [PubMed]
  26. Takata, T. Is the Novel Slot Blot a Useful Method for Quantification of Intracellular Advanced Glycation End-Products? Metabolites 2023, 13, 564. [Google Scholar] [CrossRef] [PubMed]
  27. Takata, T.; Msauji, T.; Motoo, Y. Analysis of Crude, Diverse, and Multiple Advanced Glycation End-Product Patterns May Be Important and Beneficial. Metabolites 2024, 14, 3. [Google Scholar]
  28. Takata, T.; Inoue, S.; Masauji, T.; Miyazawa, T.; Motoo, Y. Generation and Accumulation of Various Advanced Glycation End-Products in Cardiomyocytes May Induce Cardiovascular Disease. Int. J. Mol. Sci. 2024, 25, 7319. [Google Scholar] [CrossRef]
  29. Takata, T.; Inoue, S.; Kunii, K.; Masauji, T.; Miyazawa, K. Slot Blot- and Electrospray Ionization-Mass Spectrometry/Matrix-Assisted Laser Desorption/Ionization-Mass Spectrometry-Based Novel Analysis Methods for the Identification and Quantification of Advanced Glycation End-Products in the Urine. Int. J. Mol. Sci. 2024, 25, 9632. [Google Scholar] [CrossRef]
  30. Oya-Ito, T.; Naito, Y.; Takagi, T.; Handa, O.; Matsui, H.; Yamada, M.; Shima, K.; Yoshikawa, T. Heat-shock protein 27 (Hsp27) as a target of methylglyoxal in gastrointestinal cancer. Biochem. Biophys. Acta. 2011, 1812, 769–781. [Google Scholar]
  31. Chen, P.; Gregersen, H.; Zhao, J. Advanced glycation end-product expression is upregulated in the gastrointestinal tract of type 2 diabetic rats. World J. Diabetes 2015, 6, 662–672. [Google Scholar] [CrossRef] [PubMed][Green Version]
  32. Wang, M.; Fang, H.; Xie, C. Advanced glycation end products in gastric cancer: A promising future. World J. Clin. Oncol. 2024, 15, 1117–1121. [Google Scholar] [CrossRef] [PubMed]
  33. Mark, R.; Bermejo, J.L.; Bierhaus, A.; Plinkert, P.K.; Angel, P.; Hess, J. The receptor for advanced glycation end products is dispensable in a mouse model of oral and esophageal carcinogenesis. Histol. Histopathol. 2013, 28, 1585–1594. [Google Scholar] [PubMed]
  34. Tancharoen, S.; Gando, S.; Binita, S.; Nagasato, T.; Kikuchi, K.; Nawa, Y.; Dararat, P.; Yamamoto, M.; Narkpinit, S.; Maruyama, I. HMGB1 Promotes Intraoral Palatal Wound Healing through RAGE-Dependent Mechanisms. Int. J. Mol. Sci. 2016, 17, 1961. [Google Scholar] [CrossRef]
  35. Sinduja, P.; Ramani, P.; Sekaran, S. Quantitative Assessment of Receptors of Advanced Glycation End Products Expression in Tissue Samples from Patients with oral Submucous Fibrosis, Leukoplakia, and Oral Squamous Cell Carcinoma. Contemp. Clin. Dent. 2024, 15, 71–76. [Google Scholar] [CrossRef]
  36. Wang, Q.; Chang, H.; Deng, P.; He, P.; Chen, Q.; Wang, Z.; Qui, F.; Oz, F.; Chen, E.; Zeng, M. Investigation on the simultaneous inhibition of advanced glycation end products, 4-methylimidazole and hydroxymethylfurfural in thermal reaction meat flavorings by liquiritigenin, liquiritin and glycyrrhizic acid and possible pathways. Food Res. Int. 2023, 173, 113414. [Google Scholar] [CrossRef]
  37. Alvi, S.S.; Nabi, R.; Khan, M.S.; Akhter, F.; Ahmad, S.; Khan, M.S. Glycyrrhizic Acid Scavenges Reactive Carbonyl Species and Attenuates Glycation-Induced Multiple Protein Modification: An In Vitro and In Silico Study. Oxid. Med. Cell. Longev. 2021, 2021, 7086951. [Google Scholar] [CrossRef]
  38. Qiu, S.; Wu, X.; Wu, Q.; Jin, X.; Li, H.; Roy, R. Pharmacological Action of Baicalin on Gestational Diabetes Mellitus in Pregnant Animals Induced by Streptozotocin via AGE-RAGE Signaling Pathway. Appl. Biochem. Biotechnol. 2024, 196, 1636–1651. [Google Scholar] [CrossRef]
  39. Fu, S.; Zhao, W.; Xiog, C.; Guo, L.; Guo, J.; Qiu, Y.; Hu, C.A.; Ye, C.; Liu, Y.; Wu, Z.; et al. Baicalin modulates apoptosis via RAGE, MAPK, and AP-1 in vascular endothelial cells during Haemophilus parasuis invasion. Innate Immun. 2019, 25, 420–432. [Google Scholar] [CrossRef]
  40. Zhu, Y.; Zhao, Y.; Wang, P.; Ahmedna, M.; Sang, S. Bioactive ginger constituents alleviate protein glycation by trapping methylglyoxal. Chem. Res. Toxicol. 2015, 28, 1842–1849. [Google Scholar] [CrossRef]
  41. Sampath, C.; Zhu, Y.; Sang, S.; Ahmedna, M. Bioactive compounds isolated from apple, tea, and ginger protect against dicarbonyl induced stress in cultured human retinal epithelial cells. Phytomedicine 2016, 23, 200–213. [Google Scholar] [CrossRef] [PubMed]
  42. Nonaka, K.; Bando, M.; Sakamoto, E.; Inagaki, Y.; Naruishi, K.; Yumoto, H.; Kido, J. 6-Shogaol Inhibits Advanced Glycation End-Products-Induced IL-6 and ICAM-1 Expression by Regulating Oxidative Responses in Human Gingival Fibroblasts. Molecules 2019, 24, 3705. [Google Scholar] [CrossRef] [PubMed]
  43. Li, L.; Xu, C.; Guo, Y.; Wang, H. Screening potential treatments for mpox from Traditional Chinese Medicine by using a data-driven approach. Medicine 2023, 102, e35116. [Google Scholar] [CrossRef] [PubMed]
  44. Yen, C.; Zhao, F.; Yu, Z.; Zhu, X.; Li, C.G. Interactions Between Natural Products and Tamoxifen in Breast Cancer: A Comprehensive Literature Review. Front. Pharmacol. 2022, 13, 847113. [Google Scholar] [CrossRef]
  45. Akash, S.; Bibi, S.; Biswas, P.; Mukerjee, N.; Khan, D.A.; Hasan, M.N.; Sultana, N.A.; Hosen, M.E.; Jardan, Y.A.B.; Nafidi, H.; et al. Revolutionizing anti-cancer drug discovery against breast cancer and lung cancer by modification of natural genistein: An advanced computational and drug design approach. Front. Oncol. 2023, 13, 1228865. [Google Scholar] [CrossRef]
  46. Duan, X.; Chen, L.; Liu, Y.; Chen, H.; Wang, F.; Hu, Y. Integrated physicochemical, hormonal, and transcriptomic analysis reveals the underlying mechanism of callus formation in Pinellia ternata hydroponic cuttings. Front. Plant Sci. 2023, 14, 1189499. [Google Scholar] [CrossRef]
  47. Song, Y.; Hing, H.; Son, J.S.; Kwon, Y.O.; Lee, H.H.; Kim, H.J.; Park, J.H.; Son, M.J.; Oh, J.; Yoon, M. Investigation of Ginsenosides and Antioxidant Activities in the Roots, Leaves, and Stems of Hydroponic-Cultured Ginseng (Panax ginseng Meyer). Prev. Nutr. Food Sci. 2019, 24, 283–292. [Google Scholar] [CrossRef]
  48. Xu, Y.; Woo, S.B.; Treister, N.S. Thalidomide for management of refractory oral mucosal diseases. Oral Surg. Oral Med. Pathol. Oral Radiol. 2024, 137, 372–378. [Google Scholar] [CrossRef]
  49. Yang, Y.; Zhang, J.; Yuan, C.; Cheng, Z. Recurrent aphthous stomatitis and neoplasms of the mouth and pharynx: A two-sample Mendelian randomization study. BMC Cancer 2024, 24, 1372. [Google Scholar] [CrossRef]
  50. Coppola, N.; Cantile, T.; Adamo, D.; Canfora, F.; Baidares, S.; Riccitiello, F.; Musella, G.; Mignogna, M.D.D.; Leuci, S. Supportive care and antiviral treatments in primary herpetic gingivostomatitis: A systematic review. Clin. Oral. Investig. 2023, 27, 6333–6344. [Google Scholar] [CrossRef]
  51. Uesugi, A.; Tsushima, F.; Miyamoto, Y.; Harada, H. Pollen Food Allergy Syndrome Caused by Japanese Radish: A Case Report. Indian J. Dermatol. 2023, 68, 123. [Google Scholar] [CrossRef]
  52. Ye, X.; Liu, Y.; Chen, D.; Liao, B.; Wang, J.; Shen, J.; Gou, L.; Zhou, Y.; Zhou, X.; Liao, G.; et al. Moxidectin elevates Candida albicans ergosterol levels to synergize with polyenes against oral candidiasis. Appl. Microbiol. Biotechnol. 2024, 108, 509. [Google Scholar] [CrossRef]
  53. Pezzotti, G.; Adachi, T.; Imamura, H.; Ikegami, S.; Kitahara, R.; Yamamoto, T.; Kanakura, N.; Zhu, W.; Ishibashi, K.; Okumura, K.; et al. Raman Spectroscopic Algorithms for Assessing Virulence in Oral Candidiasis: The Fight-or-Flight Response. Int. J. Mol. Sci. 2024, 25, 11410. [Google Scholar] [CrossRef]
  54. Manpreet, K.; Ajmal, M.B.; Raheel, S.A.; Saleem, M.C.; Mubeen, K.; Gaballah, K.; Faden, A.; Kujan, O. Oral health status among transgender young adults: A cross-sectional study. BMC Oral Health 2021, 21, 575. [Google Scholar] [CrossRef]
  55. Dashti, H.; Sundaram, D. The association between nicotine stomatitis and waterpipe smoking. Tob. Induc. Dis. 2024, 22, 118. [Google Scholar] [CrossRef]
  56. Nolan, A.; Mclntosh, W.B.; Allam, B.F.; Lamey, P.J. Recurrent aphthous ulceration: Vitamin B1, B2 and B6 status and response to replacement therapy. J. Oeal. Pathol. Med. 1991, 20, 389–391. [Google Scholar] [CrossRef]
  57. Lakshmi, A.V.; Ramalakshmi, B.A. Effect of pyridoxine or riboflavin supplementation on plasma homocysteine levels in women with oral lesions. Natl. Med. J. India 1998, 11, 171–172. [Google Scholar] [PubMed]
  58. Hus, P.; Chen, J.; Kuo, S.; Wang, W.; Jan, F.; Yang, S.; Yang, C. San-Zhong-Kui-Jian-Tang Exerts Antitumor Effects Associated With Decreased Cell Proliferation and Metastasis by Targeting ERK and the Epithelial-Mesenchymal Transition Pathway in Oral Cavity Squamous Cell Carcinoma. Intrgr. Cancer Ther. 2022, 21, 15347354221134921. [Google Scholar]
  59. Shirahata, T.; Kanzawa, A.; Uematsu, M.; Fuchino, H.; Kawano, N.; Kawahara, N.; Yoshimatsu, K.; Hanawa, T.; Odaguchi, H.; Kobayashi, Y. Near-Infrared Metabolic Profiling for Discrimination of Apricot and Peach Kernels. Chem. Pharm. Bull. 2022, 70, 863–867. [Google Scholar] [CrossRef] [PubMed]
  60. Motoo, Y.; Cameron, S. Kampo medicines for supportive care of patients with cancer: A brief review. Integr. Med. Res. 2022, 11, 100839. [Google Scholar] [CrossRef]
  61. Motoo, Y.; Yasui, H. Analysis on Kampo case reports from the viewpoint of “Yasui Classification”. Tradit. Kampo Med. 2024, 11, 60–64. [Google Scholar] [CrossRef]
  62. Chung, H.; Moroi, M.; Hojo, Y.; Chen, F.; Yukawa, K.; Motoo, Y.; Arai, I. The Status of Nationwide Implementation of Integrative Medicine Programs by Japanese Local Government from a “Social Model” Viewpoint. Prespect. Integr. Med. 2024, 3, 98–105. [Google Scholar] [CrossRef]
  63. Chen, H.; Chen, D.; Zhou, S.; Chi, K.; Wu, J.; Hung, F. Multiple tophi deposits in the spine: A case report. World J. Clin. Case 2022, 10, 10647–10654. [Google Scholar] [CrossRef]
  64. Guo, Y.; Jia, X.; Du, P.; Wang, J.; Du, Y.; Li, B.; Xue, Y.; Jiang, J.; Cai, Y.; Yang, Q. Mechanistic insights into the ameliorative effects of Xianglianhuazhuo formula on chronic atrophic gastritis through ferroptosis mediated by YY1/miR-320a/TFRC signal pathway. J. Ethnopharmacol. 2024, 323, 117608. [Google Scholar] [CrossRef]
  65. Guo, Y.; Li, Z.; Cheng, N.; Jia, X.; Wang, J.; Ma, H.; Zhao, R.; Li, B.; Xue, Y.; Cai, Y.; et al. High-throughput sequencing analysis of differential microRNA expression in the process of blocking the progression of chronic atrophic gastritis to gastric cancer by Xianglian Huazhuo formula. J. Tradit. Chin. Med. 2024, 44, 703–712. [Google Scholar]
  66. Gong, C.; Li, H.; Li, H.; Li, Q.; Gu, P.; Xiao, Q.; Jia, Y.; Xiao, Q.; Mi, Y.; Wei, S.; et al. Efficacy and mechanism of long-snake moxibustion for treating insomnia in breast cancer survivors: Study protocol for a randomized controlled trial. Front. Neurol. 2025, 16, 1524412. [Google Scholar] [CrossRef]
  67. Samadi, M.; Huang, L.; Mo, P.; Hernandez, M.; Hunh, I.Y.; Jan, Y. Effects of negative pressure of four-cup cupping therapy on hemodynamic responses of the gastrocnemius. J. Bodyw. Mov. Ther. 2025, 42, 446–451. [Google Scholar] [CrossRef]
  68. Kashkash, F.; Swed, S.; Mustafa, A.; Azizi, S.; Bakkour, A.; Abdalla, A.; Alkammar, M.; Hafez, W. Acinetobacter-Induced Endocarditis Post Cupping Therapy: Case Report. Clin. Case Rep. 2025, 13, e70490. [Google Scholar] [CrossRef]
  69. Meng, X.; Guo, X.; Zhang, J.; Moriya, J.; Kobayashi, J.; Yamaguchi, R.; Yamada, S. Acupuncture on ST36, CV4 and KI1 Suppresses the Progression of Methionine- and Choline-Deficient Diet-Induced Nonalcoholic Fatty Liver Disease in Mice. Metabolites 2019, 9, 299. [Google Scholar] [CrossRef]
  70. Han, J.; Guo, X.; Meng, X.; Zhang, J.; Yamaguchi, R.; Motoo, Y.; Yamada, S. Acupuncture improved lipid metabolism by regulating intestinal absorption in mice. World J. Gatsroenterol. 2020, 26, 5118–5129. [Google Scholar] [CrossRef]
  71. Li, H.; Wang, D. Acupuncture, a Promising Therapy for Insulin Resistance and Non-Alcoholic Fatty Liver Disease. Int. J. Gen. Med. 2024, 17, 4917–4928. [Google Scholar] [CrossRef] [PubMed]
  72. Motoo, Y.; Arai, I.; Tsutani, K. Use of Kampo Diagnosis in Randomized Controlled Trials of Kampo Products in Japan: A Systematic Review. PLoS ONE 2014, 9, e104422. [Google Scholar] [CrossRef] [PubMed]
  73. Uema, M.; Hyuga, M.; Yonemitsu, K.; Hyuga, S.; Amakura, Y.; Uchiyama, N.; Mizoguchi, K.; Odaguchi, H.; Goda, Y. Antiviral Effect of Ephedrine Alkaloids-Free Ephedra Herb Extract against SARS-CoV-2 In Vitro. Microorganisms 2023, 11, 534. [Google Scholar] [PubMed]
  74. Huang, X.; Hyuga, S.; Amakura, Y.; Hyuga, M.; Uchiyama, N.; Hakamatsuka, T.; Goda, Y.; Odaguchi, H.; Hanawa, T.; Kobayashi, Y. Overlooked switch from transient sedation to sustained excitement in the Biphasic effects of Ephedra Herb extract administered orally to mice. J. Ethnopharmacol. 2023, 301, 115827. [Google Scholar]
  75. Arai, I. Clinical studies of traditional Japanese herbal medicines (Kampo): Need for evidence by the modern scientific methodology. Integr. Med. Res. 2021, 10, 100722. [Google Scholar] [CrossRef]
  76. Arai, I.; Kawahara, N. Kampo pharmaceutical products in the Japanese health-care system: Legal status and quality assurance. Tradit. Kampo Med. 2019, 6, 3–11. [Google Scholar]
  77. Miyano, K.; Hasegawa, S.; Asai, N.; Uzu, M.; Yatsuoka, W.; Ueno, T.; Nonaka, M.; Fujii, H.; Uezono, Y. The Japanese Herbal Medicine Hangeshashinto Induces Oral Keratinocyte Migration by Mediating the Expression of CXCL12 Through the Activation of Extracellular Signal-Regulated Kinase. Front. Pharmacol. 2022, 12, 695039. [Google Scholar] [CrossRef]
  78. Zhu, Y.; Duan, A.; Yu, Q.; Tian, S.; Zhou, Z.; Li, P.; Pan, D.; Tao, H.; Zhu, Q. Screening bioactive compounds from Fangji Huangqi decoction for treating rheumatoid arthritis via COX-2 magnetic ligand fishing combined with in vivo validation. J. Ethnopharmacol. 2025, 337, 11875. [Google Scholar] [CrossRef]
  79. Park, E.; Lee, M.; Jeon, W.; Seo, C.; You, S.; Shin, H. Paljung-San, a traditional herbal medicine, attenuates benign prostatic hyperplasia in vitro and in vivo. J. Ethnopharmacol. 2018, 218, 109–115. [Google Scholar] [CrossRef]
  80. Ji, Y.; Han, J.; Lee, N.; Yoon, J.; Youn, K.; Ha, H.J.; Yoon, E.; Kim, D.H.; Jun, M. Neuroprotective Effects of Baicalein, Wogonin, and Oroxylin A on Amyloid Beta-Induced Toxicity via NF-κB/MAPK Pathway Modulation. Molecules 2020, 25, 5087. [Google Scholar] [CrossRef]
  81. Li, C.; Fong, S.Y.K.; Mei, Q.; Lin, G.; Zuo, Z. Influence of mefenamic acid on the intestinal absorption and metabolism of three bioactive flavones in Radix Scutellariae and potential pharmacological impact. Pharm. Biol. 2014, 52, 291–297. [Google Scholar] [CrossRef] [PubMed]
  82. Kim, J.; Park, S.J.; Yun, K.; Cho, Y.; Park, H.; Lee, K. Isoliquiritigenin isolated from the roots of Glycyrrhiza uralensis inhibits LPS-induced iNOS and COX-2 expression via the attenuation of NF-kappaB in RAW 264.7 macrophages. Eur. J. Pharmacol. 2008, 584, 175–184. [Google Scholar] [CrossRef] [PubMed]
  83. Yan, B.; Wang, D.; Dong, S.; Cheng, Z.; Na, L.; Sang, M.; Yang, H.; Yang, Z.; Zhang, S.; Yan, Z. Palmatine inhibits TRIF-dependent NF-κB pathway against inflammation induced by LPS in goat endometrial epithelial cells. Int. Immunopharmacol. 2017, 45, 194–200. [Google Scholar] [CrossRef] [PubMed]
  84. Del Gaudio, M.P.; Kraus, S.I.; Melzer, T.M.; Bustos, P.S.; Ortega, M.G. Oral treatment with Berberine reduces peripheral nociception: Possible interaction with different nociceptive pathways activated by different allogeneic substances. J. Ethnopharmacol. 2024, 321, 117564. [Google Scholar] [CrossRef]
  85. Ma, S.; Feng, C.; Dai, G.; Song, Y.; Zhou, G.; Zhang, X.; Miao, C.; Yu, H.; Ju, W. In silico target fishing for the potential bioactive components contained in Huanglian Jiedu Tang (HLJDD) and elucidating molecular mechanisms for the treatment of sepsis. Chin. J. Nat. Med. 2015, 13, 30–40. [Google Scholar] [CrossRef]
  86. Luo, C.; Chen, H.; Wang, Y.; Lin, G.; Li, C.; Tan, L.; Su, Z.; Lai, X.; Xie, J.; Zeng, H. Protective effect of coptisine free base on indomethacin-induced gastric ulcers in rats: Characterization of potential molecular mechanisms. Life Sci. 2018, 193, 47–56. [Google Scholar] [CrossRef]
  87. Haque, M.A.; Jantan, I.; Harikrishnan, H.; Wahab, S.M.A. Magnoflorine Enhances LPS-Activated Pro-Inflammatory Responses via MyD88-Dependent Pathways in U937 Macrophages. Planta Med. 2018, 84, 1255–1264. [Google Scholar] [CrossRef]
  88. Hwang, Y.; Kim, T.; Kim, R.; Ha, H. The Natural Product 6-Gingerol Inhibits Inflammation-Associated Osteoclast Differentiation via Reduction of Prostaglandin E2 Levels. Int. J. Mol. Sci. 2018, 19, 2068. [Google Scholar] [CrossRef]
  89. Sharma, C.; Kaur, A.; Thind, S.S.; Singh, B.; Raina, S. Advanced glycation End-products (AGEs): An emerging concern for processed food industries. J. Food Sci. Technol. 2015, 52, 7561–7676. [Google Scholar] [CrossRef]
  90. Plemmenos, G.; Piperi, C. Pathogenic Molecular Mechanisms in Periodontitis and Peri-Implantitis: Role of Advanced Glycation End Products. Life 2022, 12, 218. [Google Scholar] [CrossRef]
  91. Pan, Y.; Huang, Z.; Cai, H.; Li, Z.; Zhu, J.; Wu, D.; Xu, W.; Qiu, H.; Zhang, N.; Li, G.; et al. WormCNN-Assisted Establishment and Analysis of Glycation Stress Models in C. elegans: Insights into Disease and Healthy Aging. Int. J. Mol. Sci. 2024, 25, 9675. [Google Scholar] [CrossRef] [PubMed]
  92. Silva, R.M.G.D.; Barbosa, F.C.; Santos, H.H.; Granero, F.O.; Figueiredo, C.C.M.; Granero, F.O.; Figueiredo, C.C.M.; Nicolau-Junior, N.; Hamaguchi, A.; Silva, L.P. Antioxidant and anti-glycation activities of Mandevilla velutina extract and effect on parasitemia levels in Trypanosoma cruzi experimental infection: In vivo, in vitro and in silico approaches. J. Ethnopharmacol. 2025, 337, 118994. [Google Scholar] [CrossRef] [PubMed]
  93. Ohno, R.; Ichimaru, K.; Tanaka, S.; Sugawa, H.; Katsuta, N.; Sakae, S.; Tominaga, Y.; Ban, I.; Shirakawa, J.; Yamaguchi, Y.; et al. Glucoselysine is derived from fructose and accumulates in the eye lens of diabetic rats. J. Biol. Chem. 2019, 294, 17326–17338. [Google Scholar] [CrossRef] [PubMed]
  94. Yamaguchi, H.; Matsumura, T.; Sugawa, H.; Niimi, N.; Sango, K.; Nagai, R. Glucoselysine, a unique advanced glycation end-product of the polyol pathway and its association with vascular complications in type 2 diabetes. J. Biol. Chem. 2024, 300, 107479. [Google Scholar] [CrossRef]
  95. Litwinowicz, K.; Waszczuk, E.; Kuzan, A.; Bronowicka-Szydełko, A.; Gostomska-Pampuch, K.; Naporowski, P.; Gamian, A. Alcoholic Liver Disease Is Associated with Elevated Plasma Levels of Novel Advanced Glycation End-Products: A Preliminary Study. Nutrients 2022, 14, 5266. [Google Scholar] [CrossRef]
  96. Sugawa, H.; Ikeda, T.; Tominaga, Y.; Katsuta, N.; Nagai, R. Rapid formation of N ε-(carboxymethyl)lysine (CML) from ribose depends on glyoxal production by oxidation. RSC Chem. Biol. 2024, 5, 1140–1146. [Google Scholar] [CrossRef]
  97. Ban, I.; Sugawa, H.; Nagai, R. Protein Modification with Ribose Generates Nδ-(5-hydro-5-methyl-4-imidazolone-2-yl)-ornithine. Int. J. Mol. Sci. 2022, 23, 1224. [Google Scholar] [CrossRef]
  98. Melvin, M.; Avery, K.C.; Ballentine, R.M.; Flora, J.W.; Gardner, W.; Karles, G.D.; Pithawalla, Y.B.; Smith, D.C.; Ehman, K.D.; Wagner, K.A. Formation of Diacetyl and Other α-Dicarbonyl Compounds during the Generation of E-Vapor Product Aerosols. ACS Omega 2020, 5, 17565–17575. [Google Scholar] [CrossRef]
  99. Sugiura, K.; Koike, S.; Suzuki, T.; Ogasawara, Y. Oxidative Formation of Methylglyoxal in Glycerol Preparations during Storage. Biol. Pharm. Bull. 2020, 43, 879–883. [Google Scholar] [CrossRef]
  100. LeWinter, M.M.; Taatjes, D.; Ashikaga, T.; Palmer, B.; Bishop, N.; VanBuren, P.; Bell, S.; Donaldson, C.; Meyer, M.; Margulies, K.B.; et al. Abundance, localization, and functional correlates of the advanced glycation end-product carboxymethyl lysine in human myocardium. Physol. Rep. 2017, 5, e13462. [Google Scholar]
  101. Jung, W.K.; Park, S.; Kim, H.R.; Ryu, H.Y.; Kim, Y.H.; Kim, J. Advanced Glycation End Products Increase Salivary Gland Hypofunction in d-Galactose-Induced Aging Rats and Its Prevention by Physical Exercise. Curr. Issues Mol. Biol. 2021, 43, 2059–2067. [Google Scholar] [CrossRef]
  102. Matsui, T.; Joo, H.D.; Lee, J.M.; Ju, S.M.; Tao, W.H.; Higashimoto, Y.; Fukami, K.; Yamagishi, S. Development of a monoclonal antibody-based ELISA system for glyceraldehyde-derived advanced glycation end products. Immunol. Lett. 2015, 167, 141–146. [Google Scholar] [CrossRef]
  103. Kehm, R.; Rückriemen, J.; Weber, D.; Deubel, S.; Grune, T.; Höhn, A. Endogenous advanced glycation end products in pancreatic islets after short-term carbohydrate intervention in obese, diabetes-prone mice. Nutr. Diabetes 2019, 9, 9. [Google Scholar] [CrossRef] [PubMed]
  104. Shigeta, T.; Sasamoto, K.; Yamamoto, T. Glyceraldehyde-derived advanced glycation end-products having pyrrolopyridinium-based crosslinks. Biochem. Biophys. Rep. 2021, 26, 100963. [Google Scholar] [CrossRef] [PubMed]
  105. Takeuchi, M.; Suzuki, H.; Takeda, K.; Sakai-Sakasai, A. Toxic advanced glycation end-products (TAGE) are major structures of cytotoxic AGEs derived from glyceraldehyde. Med. Hypotheses 2024, 183, 111248. [Google Scholar] [CrossRef]
  106. Simkova, P.; Capcarova, M. The influence of toxic advanced glycation end-products (TAGEs) on the development of diabetic nephropathy. Arch. Ecotoxicol. 2024, 6, 13–16. [Google Scholar] [CrossRef]
  107. Shen, C.; Lu, C.; Cheng, C.; Li, K.; Kuo, Y.; Wu, C.; Liu, C.; Hsien, S.; Tsai, C.; Yu, C. Advanced Glycation End-Products Acting as Immunomodulators for Chronic Inflammation, Inflammaging and Carcinogenesis in Patients with Diabetes and Immune-Related Diseases. Biomedicines 2024, 12, 1699. [Google Scholar] [CrossRef]
  108. Lee, H.; Gu, M.; Lee, J.; Kim, Y. Methylglyoxal-Lysine Dimer, an Advanced Glycation End Product, Induces Inflammation via Interaction with RAGE in Mesangial Cells. Mol. Nutr. Food Res. 2021, 65, e200799. [Google Scholar]
  109. Papadaki, M.; Holewinski, R.J.; Prevus, S.B.; Martin, T.G.; Stachowski, M.J.; Li, A.; Blair, C.A.; Moravec, C.S.; Van Eyk, J.E.; Campbell, K.S.; et al. Diabetes with heart failure increases methylglyoxal modifications in the sarcomere, which inhibit function. JCI. Insight 2018, 3, e121264. [Google Scholar] [CrossRef]
  110. Ruiz-Meana, M.; Minguet, M.; Bou-Teen, D.; Miro-Casas, E.; Castans, C.; Castellano, J.; Bonzon-Kulichenko, E.; Igual, A.; Rodriguez-Lecoq, R.; Vázquez, J.; et al. Ryanodine Receptor Glycation Favors Mitochondrial Damage in the Senescent Heart. Circulation 2019, 139, 949–964. [Google Scholar] [CrossRef]
  111. Papadaki, M.; Kampaengsri, T.; Barrick, S.K.; Campbell, S.G.; von Lewiski, D.; Rainer, P.P.; Harris, S.P.; Greenberg, M.J.; Kirk, J.A. Myofilament glycation in diabetes reduces contractility by inhibiting tropomyosin movement, is rescued by cMyBPC domains. J. Mol. Cell. Cardiol. 2022, 162, 1–9. [Google Scholar] [CrossRef] [PubMed]
  112. Morioka, Y.; Teshigawara, K.; Tomono, Y.; Wang, D.; Izushi, Y.; Wake, H.; Liu, K.; Takahashi, H.K.; Mori, S.; Nishibori, M. The specific localization of advanced glycation end-products (AGEs) in rat pancreatic islets. J. Pharmacol. Sci. 2017, 134, 218–224. [Google Scholar] [CrossRef] [PubMed]
  113. Indyk, D.; Bronowicka-Szydełko, A.; Gamian, A.; Kuzan, A. Advanced glycation end products and their receptors in serum of patients with type 2 diabetes. Sci. Rep. 2021, 11, 13264. [Google Scholar] [CrossRef] [PubMed]
  114. Katsuta, N.; Nagai, M.; Saruwatari, K.; Nakamura, M.; Nagai, R. Mitochondrial stress and glycoxidation increase with decreased kidney function. J. Clin. Biochem. Nutr. 2023, 72, 147–156. [Google Scholar] [CrossRef]
  115. Kato, S.; Matsumura, T.; Sugawa, H.; Nagai, R. Correlation between serum advanced glycation end-products and vascular complications in patient with type 2 diabetes. Sci. Rep. 2024, 14, 18722. [Google Scholar] [CrossRef]
  116. Senavirathna, L.; Ma, C.; Chen, R.; Pan, S. Proteomic Investigation of Glyceraldehyde-Derived Intracellular AGEs and Their Potential Influence on Pancreatic Ductal Cells. Cells 2021, 10, 1005. [Google Scholar] [CrossRef]
  117. Kinoshita, S.; Mera, K.; Ichikawa, H.; Shimasaki, S.; Nagai, M.; Taga, Y.; Iijima, K.; Hattori, S.; Fujiwara, Y.; Shirakawa, J.; et al. Nω-(Carboxymethyl)arginine Is One of the Dominant Advanced Glycation End Products in Glycated Collagens and Mouse Tissues. Oxidative Med. Cell. Longev. 2019, 2019, 9073451. [Google Scholar] [CrossRef]
  118. Takata, T.; Inoue, S.; Kunii, K.; Masauji, T.; Moriya, J.; Motoo, Y.; Miyazawa, K. Advanced Glycation End-Product-Modified Heat Shock Protein 90 May Be Associated with Urinary Stones. Diseases 2025, 13, 7. [Google Scholar] [CrossRef]
  119. Takahashi, A.; Takabatake, Y.; Kimura, T.; Maejima, I.; Namba, T.; Yamamoro, T.; Matsuda, J.; Minami, S.; Kaimori, J.; Matsui, I.; et al. Autophagy Inhibits the Accumulation of Advanced Glycation End Products by Promoting Lysosomal Biogenesis and Function in the Kidney Proximal Tubules. Diabetes 2017, 66, 1359–1372. [Google Scholar] [CrossRef]
  120. Mastrocola, R.; Collino, M.; Nigro, D.; Chiazza, F.; D’Antona, G.; Aragno, M.; Minetto, M.A. Accumulation of advanced glycation end-products and activation of the SCAP/SREBP Lipogenetic pathway occur in diet-induced obese mouse skeletal muscle. PLoS ONE 2015, 10, e0119587. [Google Scholar] [CrossRef]
  121. Añazco, C.; Riedelsberger, J.; Vega-Montoto, L.; Rojas, A. Exploring the Interplay between Polyphenols and Lysyl Oxidase Enzymes for Maintaining Extracellular Matrix Homeostasis. Int. J. Mol. Sci. 2023, 24, 10985. [Google Scholar] [CrossRef]
  122. Quansah, E.; Shaik, T.A.; Çevik, E.; Wang, X.; Höppener, C.; Meyer-Zedler, T.; Deckert, V.; Schmitt, M.; Popp, J.; Krafft, C. Investigating biochemical and structural changes of glycated collagen using multimodal multiphoton imaging, Raman spectroscopy, and atomic force microscopy. Anal. Bioanal. Chem. 2023, 415, 6257–6267. [Google Scholar] [CrossRef]
  123. Nomi, Y.; Kudo, H.; Miyamoto, K.; Okura, T.; Yamamoto, K.; Shimohiro, H.; Kitao, S.; Ito, Y.; Egawa, S.; Kawahara, K.; et al. Free advanced glycation end product distribution in blood components and the effect of genetic polymorphisms. Biochemie 2020, 179, 69–76. [Google Scholar] [CrossRef]
  124. Okura, T.; Ueta, E.; Nakmura, R.; Fujioka, Y.; Sumi, K.; Matsumoto, K.; Shoji, K.; Matsuzawa, K.; Izawa, S.; Nomi, Y.; et al. High Serum Advanced Glycation End Products Are Associated with Decreased Insulin Secretion in Patients with Type 2 Diabetes: A Brief Report. J. Diabetes Res. 2017, 2017, 5139750. [Google Scholar] [CrossRef]
  125. Damasiewicz-Bodzek, A.; Łabuz-Roszak, B.; Kumaszka, B.; Tadeusiak, B.; Tyrpień-Golder, K. The Assessment of Serum Concentrations of AGEs and Their Soluble Receptor (sRAGE) in Multiple Sclerosis Patients. Brain Sci. 2021, 11, 1021. [Google Scholar] [CrossRef]
  126. Kashiwabara, S.; Hosoe, H.; Ohno, R.; Nagai, R.; Shiraki, M. Development and Evaluation of Novel ELISA for Determination of Urinary Pentosidine. J. Nutr. Sci. Vitaminol. 2019, 65, 526–533. [Google Scholar] [CrossRef]
  127. Martin-Morales, A.; Arakawa, T.; Sato, M.; Matsumura, Y.; Mano-Usui, F.; Ikeda, K.; Inagaki, N.; Sato, K. Development of a Method for Quantitation of Glyceraldehyde in Various Body Compartments of Rodents and Humans. J. Agric. Food Chem. 2021, 69, 13246–13254. [Google Scholar] [CrossRef] [PubMed]
  128. Hassel, B.; Sørnes, K.; Elsais, A.; Cordero, P.R.; Frøland, A.S.; Rise, F. Glyceraldehyde metabolism in mouse brain and the entry of blood-borne glyceraldehyde into the brain. J. Neurochem. 2024, 168, 2868–2879. [Google Scholar] [CrossRef] [PubMed]
  129. Gustafsson, B.; Hedin, U.; Caidahl, K. Glycolaldehyde and maleyl conjugated human serum albumin as potential macrophage-targeting carriers for molecular imaging purposes. Contrast Media Mol. Imaging 2015, 10, 37–42. [Google Scholar] [CrossRef] [PubMed]
  130. Mirza, M.A.; Kandhro, A.J.; Memon, S.Q.; Khuhawar, M.Y.; Arain, R. Determination of glyoxal and methylglyoxal in the serum of diabetic patients by MEKC using stilbenediamine as derivatizing reagent. Electrophoresis 2007, 28, 3940–3947. [Google Scholar] [CrossRef]
  131. Kido, R.; Hiroshima, Y.; Kido, J.; Ikuta, T.; Sakamoto, E.; Inagaki, Y.; Naruishi, K.; Yumoto, H. Advanced glycation end-products increase lipocalin 2 expression in human oral epithelial cells. J. Periodontal Res. 2020, 55, 539–550. [Google Scholar] [CrossRef]
  132. Lin, Y.; Zhang, W.; Liu, L.; Li, W.; Li, Y.; Li, B. ENO1 Promotes OSCC Migration and Invasion by Orchestrating IL-6 Secretion from Macrophages via a Positive Feedback Loop. Int. J. Mol. Sci. 2023, 24, 737. [Google Scholar] [CrossRef]
  133. Phuong-Nguyen, K.; Mcneill, B.A.; Aston-Mourney, K.; Rivera, L.R. Advanced Glycation End-Products and Their Effects on Gut Health. Nutrients 2023, 15, 405. [Google Scholar] [CrossRef] [PubMed]
  134. Hayashi, K.; Sato, K.; Ochi, S.; Kawano, S.; Munesue, S.; Harashima, A.; Oshima, Y.; Kyoi, T.; Yamamoto, Y. Inhibitory Effects of Saururus chinensis Extract on Receptor for Advanced Glycation End-Products-Dependent Inflammation and Diabetes-Induced Dysregulation of Vasodilation. Int. J. Mol. Sci. 2022, 23, 5757. [Google Scholar] [CrossRef] [PubMed]
  135. Wang, J.; Tang, Z.; Zhang, Y.; Qiu, C.; Zhu, L.; Zhao, N.; Liu, Z. Matrine alleviates AGEs- induced cardiac dysfunctions by attenuating calcium overload via reducing ryanodine receptor 2 activity. Eur. J. Pharmacol. 2019, 842, 118–124. [Google Scholar] [CrossRef] [PubMed]
  136. Lee, H.; Gu, M.J.; Kim, Y.; Lee, J.; Lee, S.; Choi, I.; Ha, S.K. Glyoxal-Lysine Dimer, an Advanced Glycation End Product, Induces Oxidative Damage and Inflammatory Response by Interacting with RAGE. Antioxidants 2021, 10, 1486. [Google Scholar] [CrossRef]
  137. Wada, K.; Nakashima, Y.; Yamakawa, M.; Hori, A.; Seishima, M.; Tanabashi, S.; Matsushita, S.; Tokimitsu, N.; Nagata, C. Dietary advanced glycation end products and cancer risk in Japan: From the Takayama study. Cancer Sci. 2022, 113, 2839–2848. [Google Scholar] [CrossRef]
  138. Chen, J.; Radjabzadeh, D.; Medina-Gomez, C.; Voortman, T.; van Meurs, J.B.J.; Ikram, M.A.; Uitterlinden, A.G.; Kraaij, R.; Zillikens, M.C. Advanced Glycation End Products (AGEs) in Diet and Skin in Relation to Stool Microbiota: The Rotterdam Study. Nutrients 2023, 15, 2567. [Google Scholar] [CrossRef]
  139. Takata, T.; Taniguchi, S.; Mae, Y.; Kageyama, K.; Fujino, Y.; Iyama, T.; Hikita, K.; Sugihara, T.; Isomoto, H. Comparative assessment of the effects of dotinurad and febuxostat on the renal function in chronic kidney disease patients with hyperuricemia. Sci. Rep. 2025, 15, 8990. [Google Scholar] [CrossRef]
  140. Shakthiya, T.; Chand, L.; Annamalai, R. A Recent Update on Candidate Biomarkers in the Pathogenesis of Diabetic Retinopathy. Open Biomark. J. 2025, 15, e18753183372247. [Google Scholar] [CrossRef]
  141. Kato, A.; Sugawa, H.; Tabe, K.; Ito, K.; Nakashima, H.; Nagai, R. Rapid pretreatment for multi-sample analysis of advanced glycation end products and their role in nephropathy. J. Clin. Biochem. Nutr. 2022, 70, 256–261. [Google Scholar] [CrossRef] [PubMed]
  142. Zhang, M.; Zhao, W.; Zhang, Z.; He, M.; Zhang, Y.; Song, B.; Liu, J.; Zhang, H. FPS-ZM1 attenuates the deposition of lipid in the liver of diabetic mice by sterol regulatory element binding protein-1c. BMC Endocr. Disord. 2024, 24, 164. [Google Scholar] [CrossRef] [PubMed]
  143. Ameera, K.; Mukunda, D.C.; Basha, S.; Rodrigues, J.; Biswas, S.; Mazumder, N.; Prabhu, V.; Prabhu, M.M.; Mahato, K.K. Fluorescence in probing the biochemical and conformational changes in non-enzymatically glycated proteins. Appl. Spectrosc. Rev. 2025, 60, 638–670. [Google Scholar] [CrossRef]
  144. Pinto, R.S.; Ferreira, G.S.; Silvestre, G.C.R.; Santana, M.F.M.; Nunes, V.S.; Ledesma, L.; Pinto, P.R.; de Assis, S.I.S.; Machado, U.F.; de Silva, E.S.; et al. Plasma advanced glycation end products and soluble receptor for advanced glycation end products as indicators of sterol content in human carotid atherosclerotic plaques. Diabetes Vasc. Dis. Res. 2022, 19, 14791641221085269. [Google Scholar] [CrossRef]
  145. Yan, Y.; Hemmler, D.; Schmitt-Kopplin, P. HILIC-MS for Untargeted Profiling of the Free Glycation Product Diversity. Metabolites 2022, 12, 1179. [Google Scholar] [CrossRef]
  146. Takata, T.; Murayama, H.; Masauji, T. Slot Blot Analysis of Intracellular Glyceraldehyde-Derived Advanced Glycation End Products Using a Novel Lysis Buffer and Polyvinylidene Difluoride Membrane. Bio-Protoc. 2024, 14, e5038. [Google Scholar] [CrossRef]
  147. Takata, T.; Sakasai-Sakai, A.; Takeuchi, M. Intracellular Toxic Advanced Glycation End-Products May Induce Cell Death and Suppress Cardiac Fibroblasts. Metabolites 2022, 12, 615. [Google Scholar] [CrossRef]
  148. Takata, T.; Sakasai-Sakai, A.; Takeuchi, M. Intracellular Toxic Advanced Glycation End-Products in 1.4E7 Cell Line Induce Death with Reduction of Microtubule-Associated Protein 1 Light Chain 3 and p62. Nutrients 2022, 14, 332. [Google Scholar] [CrossRef]
  149. Miyazaki, S.; Takino, J.; Nagamine, K.; Takeuchi, M.; Hori, T. RasGRP2 Attenuates TAGE Modification of eNOS in Vascular Endothelial Cells. Biol. Pharm. Bull. 2025, 48, 262–266. [Google Scholar] [CrossRef]
  150. Tominaga, Y.; Sugawa, H.; Hirabayashi, K.; Ikeda, T.; Hoshi, Y.; Nagai, R. Drosera tokaiensis extract containing multiple phenolic compounds inhibits the formation of advanced glycation end-products. Arch. Biochem. Biophys. 2020, 693, 108586. [Google Scholar] [CrossRef]
  151. Sembiring, E.R.; Fuad, A.M.; Suryadi, H. Expression and purification of recombinant human granulocyte colony-stimulating factor (rG-CSF) from Pichia pastoris. Indones. J. Biotechnol. 2024, 29, 205–212. [Google Scholar] [CrossRef]
  152. Baskal, S.; Tsikas, D. Free L-Lysine and Its Methyl Ester React with Glyoxal and Methylglyoxal in Phosphate Buffer (100 mM, pH 7.4) to Form Nε-Carboxymethyl-Lysine, Nε-Carboxyethyl-Lysine and Nε-Hydroxymethyl-Lysine. Int. J. Mol. Sci. 2022, 23, 3446. [Google Scholar] [CrossRef] [PubMed]
  153. Baskal, S.; Kaiser, A.; Meis, C.; Kruger, R.; Tsikas, D. Specific and sensitive GC-MS analysis of hypusine, Nε-(4-amino-2-hydroxybutyl)lysine, a biomarker of hypusinated eukaryotic initiation factor eIF5A, and its application to the bi-ethnic ASOS study. Amino Acids 2022, 54, 1083–1099. [Google Scholar] [CrossRef] [PubMed]
  154. Baskal, S.; Bollenbach, A.; Mels, C.; Kruger, R.; Tsikas, D. Development, validation of a GC-MS method for the simultaneous measurement of amino acids, their PTM metabolites and AGEs in human urine, and application to the bi-ethnic ASOS study with special emphasis to lysine. Amino Acids 2022, 54, 615–641. [Google Scholar] [CrossRef] [PubMed]
  155. Baskal, S.; Büttner, P.; Werner, S.; Besler, C.; Lurz, P.; Thiele, H.; Tsikas, D. Profile of urinary amino acids and their post-translational modifications (PTM) including advanced glycation end-products (AGEs) of lysine, arginine and cysteine in lean and obese ZSF1 rats. Amino Acids 2022, 54, 643–652. [Google Scholar] [CrossRef]
  156. Katsuta, N.; Takahashi, H.; Nagai, M.; Sugawa, H.; Nagai, R. Changes in S-(2-succinyl)cysteine and advanced glycation end-products levels in mouse tissues associated with aging. Amino Acids 2022, 54, 653–661. [Google Scholar] [CrossRef]
  157. Suzuki, R.; Fijiwara, Y.; Saito, M.; Arakawa, S.; Shirakawa, J.; Yamanaka, M.; Komohara, Y.; Marumo, K.; Nagai, R. Intracellular Accumulation of Advanced Glycation End Products Induces Osteoblast Apoptosis Via Endoplasmic Reticulum Stress. J. Bone Miner. Res. 2020, 35, 1992–2003. [Google Scholar] [CrossRef]
  158. Singh, V.; Singh, A.K. Oral mucositis. Natl. J. Maxillofac. Surg. 2020, 11, 159–168. [Google Scholar] [CrossRef]
  159. Yokoyama, A.; Watanabe, K.; Inoue, Y.; Hirano, T.; Tamaoki, M.; Hirohashi, K.; Kawaguchi, S.; Ishida, Y.; Takeuchi, Y.; Kishimoto, Y.; et al. Somatic mosaicism in the buccal mucosa reflects lifestyle and germline risk factors for esophageal squamous cell carcinoma. Sci. Transl. Med. 2025, 17, eadq6740. [Google Scholar] [CrossRef]
  160. Yadav, N.; Palkhede, J.D.; Kim, S. Anti-Glucotoxicity Effect of Phytoconstituents via Inhibiting MGO-AGEs Formation and Breaking MGO-AGEs. Int. J. Mol. Sci. 2023, 24, 7672. [Google Scholar] [CrossRef]
  161. Bednarska, K.; Fecka, I.; Scheijen, J.L.J.M.; Ahles, S.; Vangrieken, P.; Schalkwijk, C.G. A Citrus and Pomegranate Complex Reduces Methylglyoxal in Healthy Elderly Subjects: Secondary Analysis of a Double-Blind Randomized Cross-Over Clinical Trial. Int. J. Mol. Sci. 2023, 24, 13168. [Google Scholar] [CrossRef]
  162. Schmidt, B.; Ferreira, C.; Alves Passos, C.L.; Silva, J.L.; Fialho, E. Resveratrol, Curcumin and Piperine Alter Human Glyoxalase 1 in MCF-7 Breast Cancer Cells. Int. J. Mol. Sci. 2020, 21, 5244. [Google Scholar] [CrossRef] [PubMed]
  163. Takeda, K.; Sakai-Sakasai, A.; Kajinami, K.; Takeuchi, M. A Novel Approach: Investigating the Intracellular Clearance Mechanism of Glyceraldehyde-Derived Advanced Glycation End-Products Using the Artificial Checkpoint Kinase 1 d270KD Mutant as a Substrate Model. Cells 2023, 12, 2838. [Google Scholar] [CrossRef] [PubMed]
  164. Senavirathna, L.; Hasapes, C.; Chen, R.; Pan, S. Accumulation of Intracellular Protein Advanced Glycation End Products Alters Cellular Homeostasis for Protein Clearance in Pancreatic Ductal Epithelial Cells. Biochemistry 2024, 63, 1723–1729. [Google Scholar] [CrossRef] [PubMed]
  165. Sekar, P.; Hsiao, G.; Hsu, S.; Huang, D.; Lin, W.; Chan, C. Metformin inhibits methylglyoxal-induced retinal pigment epithelial cell death and retinopathy via AMPK-dependent mechanisms: Reversing mitochondrial dysfunction and upregulating glyoxalase 1. Redox Biol. 2023, 64, 102786. [Google Scholar] [CrossRef]
  166. Prisco, S.Z.; Hartweck, L.; Keen, J.L.; Vogel, N.; Kazmirczak, F.; Eklund, M.; Hemnes, A.R.; Brittain, E.L.; Prins, K.W. Glyoxylase-1 combats dicarbonyl stress and right ventricular dysfunction in rodent pulmonary arterial hypertension. Front. Cardiovasc. Med. 2022, 9, 940932. [Google Scholar] [CrossRef]
  167. Shen, Y.; Xu, Z.; Sheng, Z. Ability of resveratrol to inhibit advanced glycation end product formation and carbohydrate-hydrolyzing enzyme activity, and to conjugate methylglyoxal. Food Chem. 2017, 216, 153–160. [Google Scholar] [CrossRef]
  168. Lee, S.M.; Zheng, L.W.; Jung, Y.; Hwang, G.; Kim, Y. Effects of hydroxycinnamic acids on the reduction of furan and α-dicarbonyl compounds. Food Chem. 2019, 312, 126085. [Google Scholar] [CrossRef]
  169. Froldi, G.; Djeujo, F.M.; Bulf, N.; Caparelli, E.; Ragazzi, E. Comparative Evaluation of the Antiglycation and Anti-α-Glucosidase Activities of Baicalein, Baicalin (Baicalein 7- O-Glucuronide) and the Antidiabetic Drug Metformin. Pharmaceutics 2022, 14, 2141. [Google Scholar] [CrossRef]
  170. Carnovali, M.; Luzi, L.; Terruzzi, L.; Banfi, G.; Mariotti, M. Liquiritigenin Reduces Blood Glucose Level and Bone Adverse Effects in Hyperglycemic Adult Zebrafish. Nutrients 2019, 11, 1042. [Google Scholar] [CrossRef]
  171. Ahmad, Y.; Nabi, F.; Siddiqui, S.; Khan, R.H.; Habib, S.; Moin, S. Assessing the role of Berberine as an inhibitor of advanced glycation end products (AGEs) formation using in vitro and molecular interaction studies. Arch. Biochem. Biophys. 2025, 766, 110292. [Google Scholar] [CrossRef] [PubMed]
  172. Soeiro, M.N.C.; Vergoten, G.; Bailly, C. Mechanism of action of glycyrrhizin against Plasmodium falciparum. Mem. Inst. Oswaldo Cruz 2021, 116, e210084. [Google Scholar] [CrossRef] [PubMed]
  173. Abu El-Asrar, A.M.; Siddiquei, M.M.; Nawaz, M.I.; Geboes, K.; Mohammad, G. The proinflammatory cytokine high-mobility group box-1 mediates retinal neuropathy induced by diabetes. Mediat. Inflamm. 2014, 2014, 746415. [Google Scholar] [CrossRef] [PubMed]
  174. Wang, Z.; Hsieh, C.; Liu, W.; Yin, M. Glycyrrhizic acid attenuated glycative stress in kidney of diabetic mice through enhancing glyoxalase pathway. Mol. Nutr. Food. Res. 2014, 58, 1426–1435. [Google Scholar] [CrossRef]
  175. Huang, Q.; Wang, P.; Zhu, Y.; Lv, L.; Sang, S. Additive Capacity of [6]-Shogaol and Epicatechin To Trap Methylglyoxal. J. Agric. Food. Chem. 2017, 65, 8356–8362. [Google Scholar] [CrossRef]
  176. Zhang, X.; Song, Y.; Han, X.; Feng, L.; Wang, R.; Zhang, M.; Zhu, M.; Jia, X.; Hu, S. Liquiritin attenuates advanced glycation end products-induced endothelial dysfunction via RAGE/NF-κB pathway in human umbilical vein endothelial cells. Mol. Cell Biochem. 2013, 374, 191–201. [Google Scholar] [CrossRef]
  177. Dai, X.; Liu, Y.; Liu, T.; Zhang, Y.; Wang, S.; Xu, T.; Yin, J.; Shi, H.; Ye, Z.; Zhu, R.; et al. SiJunZi decoction ameliorates bone quality and redox homeostasis and regulates advanced glycation end products/receptor for advanced glycation end products and WNT/β-catenin signaling pathways in diabetic mice. J. Ethnopharmacol. 2024, 219, 117167. [Google Scholar] [CrossRef]
  178. Li, Z.; Zhao, Z.; Chen, S.; Wang, X.; Wang, D.; Nie, X.; Yao, Y. Ge-Gen-Qin-Lian decoction alleviates the symptoms of type 2 diabetes mellitus with inflammatory bowel disease via regulating the AGE-RAGE pathway. BMC Complement. Med. Ther. 2024, 24, 225. [Google Scholar] [CrossRef]
  179. Lin, C.; Lin, Y.; Paul, C.R.; Hsieh, D.J.; Day, C.H.; Chen, R.; Kuo, C.; Ho, T.; Shibu, M.A.; Kai, C.; et al. Isoliquiritigenin ameliorates advanced glycation end-products toxicity on renal proximal tubular epithelial cells. Environ. Toxicol. 2022, 37, 2096–2102. [Google Scholar] [CrossRef]
  180. Shi, Q.; Zhou, T.; Hou, W.; Zhou, Y.; Deng, S.; Song, Y. Isoliquiritigenin Protects Against Diabetic Nephropathy in db/db Mice by Inhibiting Advanced Glycation End Product-Receptor for Advanced Glycation End Product Axis. Drug. Dev. Res. 2025, 86, e70051. [Google Scholar] [CrossRef]
  181. Goto, K.; Fujiwara- Tani, R.; Nukaga, S.; Miyagawa, Y.; Kawahara, I.; Nishida, R.; Ikemoto, A.; Sasaki, R.; Ogata, R.; Kishi, S.; et al. Berberine Improves Cancer-Derived Myocardial Impairment in Experimental Cachexia Models by Targeting High-Mobility Group Box-1. Int. J. Mol. Sci. 2024, 25, 4735. [Google Scholar] [CrossRef] [PubMed]
  182. Jiang, C.; Lao, G.; Ran, J.; Zhu, P. Berberine alleviates AGEs-induced ferroptosis by activating NRF2 in the skin of diabetic mice. Exp. Biol. Med. 2024, 249, 10280. [Google Scholar] [CrossRef] [PubMed]
  183. Fan, A.; Gao, M.; Tang, X.; Jiao, M.; Wang, C.; Wei, Y.; Gong, Q.; Zhong, J. HMGB1/RAGE axis in tumor development: Unraveling its significance. Front. Oncol. 2024, 14, 1336191. [Google Scholar] [CrossRef]
  184. Takata, T.; Masauji, T.; Motoo, Y. Potential of the Novel Slot Blot Method with a PVDF Membrane for Protein Identification and Quantification in Kampo Medicines. Membranes 2023, 13, 896. [Google Scholar] [CrossRef]
  185. Grzegorczyk-Karolak, I.; Gołąb, K.; Gburek, J.; Wysokińska, H.; Matkowski, A. Inhibition of Advanced Glycation End-Product Formation and Antioxidant Activity by Extracts and Polyphenols from Scutellaria alpina L. and S. altissima L. Molecules 2016, 21, 739. [Google Scholar] [CrossRef]
  186. Motoo, Y.; Hakamatsuka, T.; Kawahara, N.; Arai, I.; Tsutani, K. Standards of Reporting Kampo Products (STORK) in research articles. J. Integr. Med. 2017, 15, 182–185. [Google Scholar] [CrossRef]
  187. Bhuiyan, M.N.; Mitsuhashi, S.; Sugetomi, K.; Ubukata, M. Quercetin inhibits advanced glycation end product formation via chelating metal ions, trapping methylglyoxal, and trapping reactive oxygen species. Biosci. Biotechnol. Biochem. 2017, 81, 882–890. [Google Scholar] [CrossRef]
  188. Long, P.; Xia, Y.; Yang, Y.; Cao, J. Network-based pharmacology and molecular docking exploring the “Bupleuri Radix-Scutellariae Radix” mechanism of action in the viral hepatitis B treatment. Medicine 2022, 101, e31835. [Google Scholar] [CrossRef]
  189. Wei, M.; Li, H.; Li, Q.; Qiao, Y.; Ma, Q.; Xie, R.; Wang, R.; Liu, Y.; Wei, C.; Li, B.; et al. Based on Network Pharmacology to Explore the Molecular Targets and Mechanisms of Gegen Qinlian Decoction for the Treatment of Ulcerative Colitis. Biomed. Res. Int. 2020, 2020, 5217405. [Google Scholar] [CrossRef]
  190. Shi, X.; Guo, J.; Saravelos, S.; Hung, X.; Xia, E.; Feng, L.; Li, T. The use of intrauterine balloon therapy in reproductive medicine and surgery: A guidance for practice. Hum. Fertil. 2023, 26, 742–756. [Google Scholar] [CrossRef]
  191. Chawes, B.L.; Rechnitzer, C.; Schmiegelow, K.; Tvede, M. [Procalcitonin for early diagnosis of bacteraemia in children with cancer]. Ugeskr. Laeger 2007, 169, 138–142. [Google Scholar] [PubMed]
  192. Yimam, M.; Brownell, L.; Pantier, M.; Jia, Q. UP446, analgesic and anti-inflammatory botanical composition. Pharmacogn. Res. 2013, 5, 139–145. [Google Scholar] [CrossRef]
  193. Yimam, M.; Brownell, L.; Hodges, M.; Jia, Q. Analgesic effects of a standardized bioflavonoid composition from Scutellaria baicalensis and Acacia catechu. J. Diet. Suppl. 2012, 9, 155–165. [Google Scholar] [CrossRef] [PubMed]
  194. Jiang, J.; Eliaz, I.; Sliva, D. Suppression of growth and invasive behavior of human prostate cancer cells by ProstaCaid™: Mechanism of activity. Int. J. Oncol. 2011, 38, 1675–1682. [Google Scholar] [PubMed]
  195. Lee, C.J.; Lee, J.H.; Seok, J.H.; Hur, G.M.; Park, Y.C.; Seol, I.C.; Kim, Y.H. Effects of baicalein, berberine, curcumin and hesperidin on mucin release from airway goblet cells. Planta Med. 2003, 69, 523–526. [Google Scholar]
  196. Yao, H.; Li, S.; Hu, J.; Chen, Y.; Huang, L.; Lin, J.; Li, G.; Lin, X. Chromatographic fingerprint and quantitative analysis of seven bioactive compounds of Scutellaria barbata. Planta Med. 2011, 77, 388–393. [Google Scholar] [CrossRef]
  197. Singh, S.; Sharma, N.; Shukla, S.; Behl, T.; Gupta, S.; Anwer, M.K.; Vargas-De-La-Cruz, C.; Bungau, S.G. Understanding the Potential Role of Nanotechnology in Liver Fibrosis: A Paradigm in Therapeutics. Molecules 2023, 28, 2811. [Google Scholar] [CrossRef]
  198. Duan, L.; Jin, D.; An, X.; Zhang, Y.; Zhao, S.; Zhou, R.; Duan, Y.; Zhang, Y.; Liu, X.; Lian, F. The Potential Effect of Rhizoma coptidis on Polycystic Ovary Syndrome Based on Network Pharmacology and Molecular Docking. Evid.-Based Complement. Altern. Med. 2021, 2021, 5577610. [Google Scholar] [CrossRef]
  199. Wang, Y.; Chen, M.; Yu, H.; Yuan, G.; Luo, L.; Xu, X.; Xu, Y.; Sui, X.; Leung, E.L.; Wu, Q. The Role and Mechanisms of Action of Natural Compounds in the Prevention and Treatment of Cancer and Cancer Metastasis. Front. Biosci. 2022, 27, 192. [Google Scholar] [CrossRef]
  200. Li, Y.; Zhang, N.; Peng, X.; Ma, W.; Qin, Y.; Yao, Z.; Huang, C.; Zhang, X. Network pharmacology analysis and clinical verification of Jishe Qushi capsules in rheumatoid arthritis treatment. Medicine 2023, 102, e34883. [Google Scholar] [CrossRef]
  201. Kim, J. Investigation of Phenolic, Flavonoid, and Vitamin Contents in Different Parts of Korean Ginseng (Panax ginseng C.A. Meyer). Prev. Nutr. Food Sci. 2016, 21, 263–270. [Google Scholar] [CrossRef] [PubMed]
  202. Zhang, T.; Zhang, Z.; Dong, K.; Li, G.; Zhu, H. Yizhijiannao Granule and a combination of its effective monomers, icariin and Panax notoginseng saponins, inhibit early PC12 cell apoptosis induced by beta-amyloid (25-35). Neural Regen. Res. 2012, 7, 1845–1850. [Google Scholar] [PubMed]
  203. Du, G.; Wang, C.; Qi, L.; Zhang, Z.; Calway, T.; He, T.; Du, W.; Yuan, C. The synergistic apoptotic interaction of panaxadiol and epigallocatechin gallate in human colorectal cancer cells. Phytother. Res. 2013, 27, 272–277. [Google Scholar] [CrossRef] [PubMed]
  204. Zhang, Q.; Yang, H.; An, J.; Zhang, R.; Chen, B.; Hao, D. Therapeutic Effects of Traditional Chinese Medicine on Spinal Cord Injury: A Promising Supplementary Treatment in Future. Evid.-Based Complement. Altern. Med. 2016, 2016, 89587211. [Google Scholar] [CrossRef]
  205. Liu, J.; Xu, X.; Zhou, J.; Sun, G.; Li, Z.; Zhai, L.; Wang, J.; Ma, R.; Zhao, D.; Jiang, R.; et al. Phenolic acids in Panax ginseng inhibit melanin production through bidirectional regulation of melanin synthase transcription via different signaling pathways. J. Ginseng Res. 2023, 47, 714–725. [Google Scholar] [CrossRef]
  206. Moon, P.; Han, N.; Lee, J.S.; Kim, H.; Jeong, H. p-coumaric acid, an active ingredient of Panax ginseng, ameliolates atopic dermatitis-like skin lesions through inhibition of thymic stromal lymphopoietin in mice. J. Ginseng Res. 2021, 45, 176–182. [Google Scholar] [CrossRef]
  207. Aiexander, C.; Parsaee, A.; Vasefi, M. Polyherbal and Multimodal Treatments: Kaempferol- and Quercetin-Rich Herbs Alleviate Symptoms of Alzheimer’s Disease. Biology 2023, 12, 1453. [Google Scholar]
  208. Kim, J.; Shin, H.; Seo, C. Chemical interaction between Paeonia lactiflora and Glycyrrhiza uralensis, the components of Jakyakgamcho-tang, using a validated high-performance liquid chromatography method: Herbal combination and chemical interaction in a decoction. J. Sep. Sci. 2014, 37, 2704–2715. [Google Scholar] [CrossRef]
  209. Rapaport, J.; Sadgrove, N.J.; Arruda, S.; Swearingen, A.; Abidi, Z.; Sadick, N. Real-World, Open-Label Study of the Efficacy and Safety of a Novel Serum in Androgenetic Alopecia. J. Drugs Dermatol. 2023, 22, 559–564. [Google Scholar] [CrossRef]
  210. Chociej, P.; Foss, K.; Jabłońska, M.; Ustarbowska, M.; Sawicki, T. The Profile and Content of Polyphenolic Compounds and Antioxidant and Anti-Glycation Properties of Root Extracts of Selected Medicinal Herbs. Plant Food Hum. Nutr. 2024, 79, 468–473. [Google Scholar] [CrossRef]
  211. Kim, Y.; Oh, J.; Jang, C.H.; Lim, J.S.; Lee, J.S.; Kim, J. In Vivo Anti-inflammatory Potential of Viscozyme®-Treated Jujube Fruit. Foods 2020, 9, 1033. [Google Scholar] [CrossRef]
  212. Yan, M.; Wang, Y.; Watharkar, R.B.; Pu, Y.; Wu, C.; Lin, M.; Lu, D.; Liu, M.; Bao, J.; Xia, Y. Physicochemical and antioxidant activity of fruit harvested from eight jujube (Ziziphus jujuba Mill.) cultivars at different development stages. Sci. Rep. 2022, 12, 2272. [Google Scholar] [PubMed]
  213. Yuan, S.; Li, W.; Li, Q.; Wang, L.; Cao, J.; Jiang, W. Defense Responses, Induced by p-Coumaric Acid and Methyl p-Coumarate, of Jujube (Ziziphus jujuba Mill.) Fruit against Black Spot Rot Caused by Alternaria alternata. J. Agric. Food Chem. 2019, 67, 2801–2810. [Google Scholar] [CrossRef] [PubMed]
  214. Bekheit, S.O.; Kolieb, E.; El-Awady, E.E.; Alwaili, M.A.; Alharthi, A.; Khodeer, D.M. Cardioprotective Effects of Ferulic Acid Through Inhibition of Advanced Glycation End Products in Diabetic Rats with Isoproterenol-Induced Myocardial Infarction. Pharmaceuticals 2025, 18, 319. [Google Scholar] [CrossRef] [PubMed]
  215. Toma, L.; Sanda, G.M.; Niculescu, L.S.; Deleanu, M.; Stancu, C.S.; Sima, A.V. Caffeic acid attenuates the inflammatory stress induced by glycated LDL in human endothelial cells by mechanisms involving inhibition of AGE-receptor, oxidative, and endoplasmic reticulum stress. BioFactors 2017, 43, 685–697. [Google Scholar] [CrossRef]
  216. Umadevi, S.; Gopi, V.; Elangovan, V. Regulatory mechanism of gallic acid against advanced glycation end products induced cardiac remodeling in experimental rats. Chem.-Biol. Interact. 2014, 208, 28–36. [Google Scholar] [CrossRef]
  217. Umadevi, S.; Gopi, V.; Vellaichamy, E. Inhibitory effect of gallic acid on advanced glycation end products induced up-regulation of inflammatory cytokines and matrix proteins in H9C2 (2-1) cells. Cardiovasc. Toxicol. 2013, 13, 396–405. [Google Scholar] [CrossRef]
  218. Zhang, N.; Chen, P.; Liang, X.; Sun, J.; Liu, Q.; Guan, S.; Wang, Q. Luteolin targets the AGE-RAGE signaling to mitigate inflammation and ferroptosis in chronic atrophic gastritis. Aging 2024, 16, 10918–10930. [Google Scholar] [CrossRef]
  219. Zhou, Q.; Cheng, K.; Gong, J.; Li, E.T.S.; Wang, M. Apigenin and its methylglyoxal-adduct inhibit advanced glycation end products-induced oxidative stress and inflammation in endothelial cells. Biochem. Pharmacol. 2019, 166, 231–241. [Google Scholar] [CrossRef]
  220. Ma, Y.; Ma, Z.; Zhang, Y.; Luo, C.; Huang, P.; Tong, J.; Ding, H.; Liu, H. Apigenin and baicalein ameliorate thoracic aortic structural deterioration and cognitive deficit via inhibiting AGEs/RAGE/NF-κB pathway in D-galactose-induced aging rats. Eur. J. Pharmacol. 2024, 976, 176660. [Google Scholar] [CrossRef]
  221. Garg, S.; Malhotra, R.K.; Khan, S.I.; Sarkar, S.; Susrutha, P.N.; Singh, V.; Goyal, S.; Nag, T.C.; Ray, R.; Bhatia, J.; et al. Fisetin attenuates isoproterenol-induced cardiac ischemic injury in vivo by suppressing RAGE/NF-κB mediated oxidative stress, apoptosis and inflammation. Phytomedicine 2019, 56, 147–155. [Google Scholar] [CrossRef] [PubMed]
  222. Zhou, Q.; Xu, H.; Zhao, Y.; Liu, B.; Cheng, K.; Xhen, F.; Wang, M. 6-C-(E-Phenylethenyl)-naringenin, a Styryl Flavonoid, Inhibits Advanced Glycation End Product-Induced Inflammation by Upregulation of Nrf2. J. Agric. Food Chem. 2022, 70, 3842–3851. [Google Scholar] [CrossRef] [PubMed]
  223. Syed, A.A.; Reza, M.I.; Shafiq, M.; Kumariya, S.; Singh, P.; Husain, A.; Hanif, K.; Gayen, J.R. Naringin ameliorates type 2 diabetes mellitus-induced steatohepatitis by inhibiting RAGE/NF-κB mediated mitochondrial apoptosis. Life Sci. 2020, 257, 118118. [Google Scholar] [CrossRef]
  224. Zu, G.; Sun, K.; Li, L.; Zu, X.; Han, T.; Huang, H. Mechanism of quercetin therapeutic targets for Alzheimer disease and type 2 diabetes mellitus. Sci. Rep. 2021, 11, 22959. [Google Scholar] [CrossRef] [PubMed]
  225. Han, J.; Hou, J.; Liu, Y.; Liu, P.; Zhao, T.; Wang, X. Using Network Pharmacology to Explore the Mechanism of Panax notoginseng in the Treatment of Myocardial Fibrosis. J. Diabetes Res. 2022, 2022, 8895950. [Google Scholar] [CrossRef]
  226. Kang, C.; Li, X.; Liu, P.; Liu, Y.; Niu, Y.; Zeng, X.; Liu, J.; Zhao, H.; Qiu, S. Quercetin inhibits the activity and function of dendritic cells through the TLR4/IRAK4/NF-κB signalling pathway. Contemp. Oncol. 2023, 27, 182–189. [Google Scholar]
  227. Gong, X.; Huang, Y.; Ma, Q.; Jing, M.; Zhan, K.; Zhao, G. Quercetin Alleviates Lipopolysaccharide-Induced Cell Damage and Inflammation via Regulation of the TLR4/NF-κB Pathway in Bovine Intestinal Epithelial Cells. Curr. Issues Mol. Biol. 2022, 44, 5234–5246. [Google Scholar] [CrossRef]
  228. Zhang, H.; Li, Y.; Liu, Z.; Wang, J. Quercetin effectively improves LPS-induced intestinal inflammation, pyroptosis, and disruption of the barrier function through the TLR4/NF-κB/NLRP3 signaling pathway in vivo and in vitro. Food Nutr. Res. 2022, 66, 8948. [Google Scholar] [CrossRef]
  229. Jiang, M.; Wang, K.; Huang, Y.; Zhang, X.; Yang, T.; Zhan, K.; Zhao, G. Quercetin Alleviates Lipopolysaccharide-Induced Cell Oxidative Stress and Inflammatory Responses via Regulation of the TLR4-NF-κB Signaling Pathway in Bovine Rumen Epithelial Cells. Toxins 2023, 15, 512. [Google Scholar] [CrossRef]
  230. Shams, S.G.E.; Eissa, R.G. Amelioration of ethanol-induced gastric ulcer in rats by quercetin: Implication of Nrf2/HO1 and HMGB1/TLR4/NF-κB pathways. Heliyon 2022, 8, e11159. [Google Scholar] [CrossRef]
  231. Yu, X.; Zhang, D.; Xiao, C.; Zhou, Y.; Li, X.; Wang, L.; He, Z.; Reilly, J.; Xiao, Z.; Shu, X. P-Coumaric Acid Reverses Depression-Like Behavior and Memory Deficit Via Inhibiting AGE-RAGE-Mediated Neuroinflammation. Cells 2022, 11, 1594. [Google Scholar] [CrossRef]
  232. Wu, Q.; Kong, Y.; Liang, Y.; Niu, M.; Feng, N.; Zhang, C.; Qi, Y.; Guo, Z.; Xiao, J.; Zhou, M.; et al. Protective mechanism of fruit vinegar polyphenols against AGEs-induced Caco-2 cell damage. Food Chem. X 2023, 19, 100736. [Google Scholar] [CrossRef]
  233. Zabad, O.M.; Samra, Y.A.; Eissa, L.A. P-Coumaric acid alleviates experimental diabetic nephropathy through modulation of Toll like receptor-4 in rats. Life Sci. 2019, 238, 116965. [Google Scholar] [CrossRef] [PubMed]
  234. Truong, T.M.T.; Seo, S.H.; Chung, S.; Kang, I. Attenuation of hepatic fibrosis by p-Coumaric acid via modulation of NLRP3 inflammasome activation in C57BL/6 mice. J. Nutr. Biochem. 2023, 112, 109204. [Google Scholar] [CrossRef] [PubMed]
  235. Feng, T.; Zhang, M.; Xu, Q.; Song, F.; Wang, L.; Gai, S.; Tang, H.; Wang, S.; Zhou, L.; Li, H. Exploration of molecular targets and mechanisms of Chinese medicinal formula Acacia Catechu -Scutellariae Radix in the treatment of COVID-19 by a systems pharmacology strategy. Phytother. Res. 2022, 36, 4210–4229. [Google Scholar] [CrossRef] [PubMed]
  236. Zhang, X.; Wang, J.; Fu, J.; Hu, J.; Zhang, H.; Ye, M.; Yang, X.; Yu, H.; Xu, H.; Lu, J.; et al. Dissecting the antitumor effects of Scutellaria barbata: Initial insights into the metabolism of scutellarin and luteolin by gut microbiota. J. Pharm. Biomed. Anal. 2024, 248, 116325, Correction in J. Pharm. Biomed. Anal. 2024, 249, 116393. [Google Scholar] [CrossRef]
  237. Li, H.; Luo, D.; Wei, R.; Sun, M.; Zhang, X.; Deng, H.; Bian, W.; Wei, H.; Huang, Y. Investigating the Mechanism of Rhizoma Coptidis-Eupatorium fortunei Medicine in the Treatment of Type 2 Diabetes Based on Network Pharmacology and Molecular Docking. Biomed. Res. Int. 2022, 2022, 7978258. [Google Scholar] [CrossRef]
  238. Chen, J.; Liu, Z.; Liu, Y.; Ji, X.; Li, X.; Wei, Y.; Zi, F.; Tan, Y. Differences in autotoxic substances and microbial community in the root space of Panax notoginseng coinducing the occurrence of root rot. Appl. Environ. Microbiol. 2024, 90, e0228723. [Google Scholar] [CrossRef]
  239. Li, Z.; Liu, J.; Zhao, D.; Jiang, R.; Sun, L. Panax ginseng Meyer cv. Silvatica phenolic acids protect DNA from oxidative damage by activating Nrf2 to protect HFF-1 cells from UVA-induced photoaging. J. Ethnopharmacol. 2023, 302, 115883. [Google Scholar]
  240. Park, W.H.; Chang, M.S.; Yang, W.M.; Bae, H.; Kim, N.I.; Park, S.K. Cytoprotective effect of Panax ginseng on gallic acid-induced toxicity in TM3 mouse Leydig cells. Fitoterapia 2007, 78, 577–579. [Google Scholar] [CrossRef]
  241. Umukoro, S.; Kalejaye, H.A.; Ben-Azu, B.; Ajayi, A.M. Naringenin attenuates behavioral derangements induced by social defeat stress in mice via inhibition of acetylcholinesterase activity, oxidative stress and release of pro-inflammatory cytokines. Biomed. Pharmacother. 2018, 105, 714–723. [Google Scholar] [CrossRef] [PubMed]
  242. Cao, Y.; Xu, W.; Huang, Y.; Zeng, X. Licochalcone B, a chalcone derivative from Glycyrrhiza inflata, as a multifunctional agent for the treatment of Alzheimer’s disease. Nat. Prod. Res. 2020, 34, 736–739. [Google Scholar] [CrossRef]
  243. Dey, S.; Deepak, M.; Setty, M.; D’Souza, P.; Agarwl, A.; Sangli, G.K. Bioactive caffeic acid esters from Glycyrrhiza glabra. Nat. Prod. Res. 2009, 23, 1657–1663. [Google Scholar] [CrossRef]
  244. Niu, W.; Wu, F.; Cui, H.; Cao, W.; Xhao, Y.; Wu, Z.; Fan, M.; Liang, C. Network Pharmacology Analysis to Identify Phytochemicals in Traditional Chinese Medicines That May Regulate ACE2 for the Treatment of COVID-19. Evid.-Based Complement. Altern. Med. 2020, 2020, 7493281. [Google Scholar] [CrossRef]
  245. Li, S.; Li, W.; Wang, Y.; Asada, Y.; Koike, K. Prenylflavonoids from Glycyrrhiza uralensis and their protein tyrosine phosphatase-1B inhibitory activities. Bioorganic Med. Chem. Lett. 2010, 20, 5398–5401. [Google Scholar] [CrossRef]
  246. Yamagishi, N.; Yamamoto, Y.; Noda, C.; Hatayama, T. Naringenin inhibits the aggregation of expanded polyglutamine tract-containing protein through the induction of endoplasmic reticulum chaperone GRP78. Biol. Pharm. Bull. 2012, 35, 1836–1840. [Google Scholar] [CrossRef]
  247. Khamseekaew, J.; Iampanichakul, M.; Potue, P.; Maneesai, P.; Tangsucharit, P.; Rattanakanokchai, S.; Pakdeechote, P. The Alleviative Effect of Naringin Against Cardiovascular Dysfunction and Remodeling in Hypertensive Rats by Suppressing the Angiotensin II Pathway. Food Sci. Nutr. 2025, 13, e70484. [Google Scholar] [CrossRef] [PubMed]
  248. Alifarsangi, A.; Esmaeili-Mahani, S.; Salarkia, E.; Alizadeh, E.; Abbasnejad, M. Investigation of the Inhibitory Effect of Naringin on the Development of Morphine Physical Dependency in Male Rats. Addict. Health 2025, 17, 1543. [Google Scholar] [CrossRef] [PubMed]
  249. Eltahir, A.O.E.; Omoruyi, S.I.; Augustine, T.N.; Luckay, R.C.; Hussein, A.A. Neuroprotective Effects of Glycyrrhiza glabra Total Extract and Isolated Compounds. Pharmaceuticals 2024, 17, 852. [Google Scholar] [CrossRef]
Figure 1. Types of stomatitis. Traumatic stomatitis [1,48], aphthous stomatitis [2,49], herpetic stomatitis [50,51], candidal stomatitis [52,53], nicotine stomatitis [54,55], lack of vitamin B1, B2, B6, and B12 [56,57], and intra-/extracellular AGEs stomatitis are described in this review (see Section 6). The black line indicates that each case of stomatitis is confined to the clinical stage.
Figure 1. Types of stomatitis. Traumatic stomatitis [1,48], aphthous stomatitis [2,49], herpetic stomatitis [50,51], candidal stomatitis [52,53], nicotine stomatitis [54,55], lack of vitamin B1, B2, B6, and B12 [56,57], and intra-/extracellular AGEs stomatitis are described in this review (see Section 6). The black line indicates that each case of stomatitis is confined to the clinical stage.
Ijms 26 09118 g001
Figure 2. Relationship between traditional Chinese medical science (TCMS) and Kampo, and specifically, the Hangeshashinto (Ban-Xia-Xie-Xin-Tang) treatment [5,6,7,8,11,59,60,61,62]. (a) TCMS was exported and instructed into antient Japan from antient China, and was modified to “Kampo” in Japan. (b) Ban-Xia-Xie-Xin-Tang and the infrmomation of the method of treatment were exported and instructed into antient Japan from antient China. Ban-Xia-Xie-Xin-Tang (Chinese name) had been called Hangeshashinto in as Japanese name. Pink and green squares indicate TCMS and Kampo (traditional Japanese medicine), respectively. Ban-Xia-Xie-Xin-Tang and Hangeshashinto are composed of the same components. Hangeshashinto is generally administered as an oral treatment; however, gargling and coating of the oral squamous cells were used in ancient practice. Blue arrow indicate the medical science, medicine, and information of treatment of antient China were exported and instructed into antient Japan, and they were modified in Japan. Red term indicate the important and focued treatment compared with other treatment of the traditional Chinese and Japanese meidicines (See Section 3.2).
Figure 2. Relationship between traditional Chinese medical science (TCMS) and Kampo, and specifically, the Hangeshashinto (Ban-Xia-Xie-Xin-Tang) treatment [5,6,7,8,11,59,60,61,62]. (a) TCMS was exported and instructed into antient Japan from antient China, and was modified to “Kampo” in Japan. (b) Ban-Xia-Xie-Xin-Tang and the infrmomation of the method of treatment were exported and instructed into antient Japan from antient China. Ban-Xia-Xie-Xin-Tang (Chinese name) had been called Hangeshashinto in as Japanese name. Pink and green squares indicate TCMS and Kampo (traditional Japanese medicine), respectively. Ban-Xia-Xie-Xin-Tang and Hangeshashinto are composed of the same components. Hangeshashinto is generally administered as an oral treatment; however, gargling and coating of the oral squamous cells were used in ancient practice. Blue arrow indicate the medical science, medicine, and information of treatment of antient China were exported and instructed into antient Japan, and they were modified in Japan. Red term indicate the important and focued treatment compared with other treatment of the traditional Chinese and Japanese meidicines (See Section 3.2).
Ijms 26 09118 g002
Figure 3. Flavonoid compounds obtained from the Hangeshathito water extract [6,20]. Liquiritin [6], liquiritin apioside [6], wogonin [6,20], worgonin-7-O-glucuronide [6], baicalin (baicalein-7-O-glucuronide) [6], oroxylin [6], and oroxylin-7-O-glucoside [6].
Figure 3. Flavonoid compounds obtained from the Hangeshathito water extract [6,20]. Liquiritin [6], liquiritin apioside [6], wogonin [6,20], worgonin-7-O-glucuronide [6], baicalin (baicalein-7-O-glucuronide) [6], oroxylin [6], and oroxylin-7-O-glucoside [6].
Ijms 26 09118 g003
Figure 4. Chalcon compounds identified in the Hangeshashinto water extract [6]. Isoquritin [6], isoliquiritin apioside [6], and isoquitigenin [6].
Figure 4. Chalcon compounds identified in the Hangeshashinto water extract [6]. Isoquritin [6], isoliquiritin apioside [6], and isoquitigenin [6].
Ijms 26 09118 g004
Figure 5. Alkaloid compounds obtained from the Hangeshathito water extract [6]. Berberine [6], epiberberine [6], jateprrhizine [6], coptisine [6], and magnoflorine [6].
Figure 5. Alkaloid compounds obtained from the Hangeshathito water extract [6]. Berberine [6], epiberberine [6], jateprrhizine [6], coptisine [6], and magnoflorine [6].
Ijms 26 09118 g005
Figure 6. Triterpene and gingerol groups (one of the monophenolic acid groups) obtained from the Hangeshashinto water extract. [6,20,22]. (a) Glycyrrhizin [6,22], (b) 6-gingerol [20], and 6-shogaol [6].
Figure 6. Triterpene and gingerol groups (one of the monophenolic acid groups) obtained from the Hangeshashinto water extract. [6,20,22]. (a) Glycyrrhizin [6,22], (b) 6-gingerol [20], and 6-shogaol [6].
Ijms 26 09118 g006
Figure 7. Major saccharides involved in the generation of AGEs. Glucose [26,27,28,29,93,94], fructose [26,27,28,29,93,94], melibiose [95], and ribose [96,97].
Figure 7. Major saccharides involved in the generation of AGEs. Glucose [26,27,28,29,93,94], fructose [26,27,28,29,93,94], melibiose [95], and ribose [96,97].
Ijms 26 09118 g007
Figure 8. The metabolites and non-enzymatic reaction products of glucose, fructose, and ribose. AGEs are generated from glyceraldehyde, glycolaldehyde, methylglyoxal, glyoxal, and 3-deoxyglucosone [26,27,28,29]. The open red square identifies a triose.
Figure 8. The metabolites and non-enzymatic reaction products of glucose, fructose, and ribose. AGEs are generated from glyceraldehyde, glycolaldehyde, methylglyoxal, glyoxal, and 3-deoxyglucosone [26,27,28,29]. The open red square identifies a triose.
Ijms 26 09118 g008
Figure 9. Free-type AGEs [26,27,28,29]. (a) Free-type AGEs with one amino acid residue. CML, Nε-carboxymethyl–lysine; CEL, Nε-carboxyethyl–lysine; GLAP, glyceraldehyde-related pyridinium; GA-pyridine; argpyrimidine; GH-1, Nδ-(5-hydro-4-imidazolone-2-yl)-ornithine (glyoxal-hydro-imidazolone); MG-H1, Nδ-(5-hydro-5-methyl-4-imidazolone-2-yl)-ornithine (methylglyoxal-hydro-imidazolone). (b) Free-type AGEs with two amino acid residues. MOLD, methylglyoxal-derived imidazolium cross-link (methylglyoxal–lysine dimer); pentosidine; glucospane; GODIC, glyoxal-derived imidazolium cross-link; MODIC, methylglyoxal-derived imidazolium cross-link; and DODIC, 3-doxyglucosone-derived imidazolium cross-link.
Figure 9. Free-type AGEs [26,27,28,29]. (a) Free-type AGEs with one amino acid residue. CML, Nε-carboxymethyl–lysine; CEL, Nε-carboxyethyl–lysine; GLAP, glyceraldehyde-related pyridinium; GA-pyridine; argpyrimidine; GH-1, Nδ-(5-hydro-4-imidazolone-2-yl)-ornithine (glyoxal-hydro-imidazolone); MG-H1, Nδ-(5-hydro-5-methyl-4-imidazolone-2-yl)-ornithine (methylglyoxal-hydro-imidazolone). (b) Free-type AGEs with two amino acid residues. MOLD, methylglyoxal-derived imidazolium cross-link (methylglyoxal–lysine dimer); pentosidine; glucospane; GODIC, glyoxal-derived imidazolium cross-link; MODIC, methylglyoxal-derived imidazolium cross-link; and DODIC, 3-doxyglucosone-derived imidazolium cross-link.
Ijms 26 09118 g009
Figure 10. Free-type AGEs were synthesized in a tube, and their structure was predicted [104,105]. (a) PPG1, pyrrolopyridinium–lysine dimer-derived glyceraldehyde 1; PPG2, pyrrolopyridinium–lysine dimer-derived glyceraldehyde 2. They were synthesized from glyceraldehyde and Nα-acetyl-lysine in the tube in 2020, but were not detected in vivo in 2025 [104]. (b) 1,4-dihydropyrazine compounds 1 and 2 were named as “Toxic AGEs (TAGE)” by Takeuchi et al. They hypothesized the structure of TAGE; however, it has not been proved [105].
Figure 10. Free-type AGEs were synthesized in a tube, and their structure was predicted [104,105]. (a) PPG1, pyrrolopyridinium–lysine dimer-derived glyceraldehyde 1; PPG2, pyrrolopyridinium–lysine dimer-derived glyceraldehyde 2. They were synthesized from glyceraldehyde and Nα-acetyl-lysine in the tube in 2020, but were not detected in vivo in 2025 [104]. (b) 1,4-dihydropyrazine compounds 1 and 2 were named as “Toxic AGEs (TAGE)” by Takeuchi et al. They hypothesized the structure of TAGE; however, it has not been proved [105].
Ijms 26 09118 g010
Figure 11. Various routes in the production of CML from glucose and ribose [28,96,97]. Glyoxal and glycolaldehyde are produced through enzymatic reaction and autooxidation. CML, Nε-carboxymethyl–lysine.
Figure 11. Various routes in the production of CML from glucose and ribose [28,96,97]. Glyoxal and glycolaldehyde are produced through enzymatic reaction and autooxidation. CML, Nε-carboxymethyl–lysine.
Ijms 26 09118 g011
Figure 12. AGE-modified proteins [27,28,29]. Blue circles indicate the amino acids in proteins X, Y, and Z. Black and red indicate the number of amino acids. (a) AGEs, including free types, can be modified into one molecular protein, X. CEL, Nε-cassrboxyethyl-lysine; Arg-P, argpyrimidine; CML, Nε-carboxymethyl–lysine; MG-H1, Nδ-(5-hydro-5-methyl-4-imidazolone-2-yl)-ornithine (methylglyoxal-hydro-imidazolone). (b) D1 and 2; model of free-type AGEs that contains two amino acid residues. Proteins Y and Z are linked with D1 via an intermolecular covalent bond. D1 connected both 5th in protein Y and 6th in protein Z. In contrast, the 7th and 15th amino acids in protein Y are associated with D2 via an intramolecular covalent bond.
Figure 12. AGE-modified proteins [27,28,29]. Blue circles indicate the amino acids in proteins X, Y, and Z. Black and red indicate the number of amino acids. (a) AGEs, including free types, can be modified into one molecular protein, X. CEL, Nε-cassrboxyethyl-lysine; Arg-P, argpyrimidine; CML, Nε-carboxymethyl–lysine; MG-H1, Nδ-(5-hydro-5-methyl-4-imidazolone-2-yl)-ornithine (methylglyoxal-hydro-imidazolone). (b) D1 and 2; model of free-type AGEs that contains two amino acid residues. Proteins Y and Z are linked with D1 via an intermolecular covalent bond. D1 connected both 5th in protein Y and 6th in protein Z. In contrast, the 7th and 15th amino acids in protein Y are associated with D2 via an intramolecular covalent bond.
Ijms 26 09118 g012
Figure 13. Mechanisms of inhibition of intra-/extracellular AGE-induced cytotoxicity [27,28,29]. The pink square represents a cell. Blue circles indicate anti-AGE compounds. Brown circles and squares indicate full-length RAGE or TLR4. Orange squares indicate protein. The yellow hexagrams indicate AGEs. The yellow circles indicate degraded AGEs. Black arrows indicate the Maillard reaction and other non-enzymatic reactions involving proteins and AGE precursors (e.g., methylglyoxal, glyoxal, glyceraldehyde, glycolaldehyde). The red arrow indicates activation of GLO-1. The green arrow indicates the carbonyl trap system. The gray arrow indicates AGE degradation via autophagy or the ubiquitin–proteasome system. The blue arrow indicates a cell signaling pathway to induce inflammation. The black line indicates inhibition effects. AGE, advanced glycation end-products; RAGE, receptor for advanced glycation end-products; TLR4, toll-like receptor 4. GLO-1, glyoxalase-1.
Figure 13. Mechanisms of inhibition of intra-/extracellular AGE-induced cytotoxicity [27,28,29]. The pink square represents a cell. Blue circles indicate anti-AGE compounds. Brown circles and squares indicate full-length RAGE or TLR4. Orange squares indicate protein. The yellow hexagrams indicate AGEs. The yellow circles indicate degraded AGEs. Black arrows indicate the Maillard reaction and other non-enzymatic reactions involving proteins and AGE precursors (e.g., methylglyoxal, glyoxal, glyceraldehyde, glycolaldehyde). The red arrow indicates activation of GLO-1. The green arrow indicates the carbonyl trap system. The gray arrow indicates AGE degradation via autophagy or the ubiquitin–proteasome system. The blue arrow indicates a cell signaling pathway to induce inflammation. The black line indicates inhibition effects. AGE, advanced glycation end-products; RAGE, receptor for advanced glycation end-products; TLR4, toll-like receptor 4. GLO-1, glyoxalase-1.
Ijms 26 09118 g013
Figure 14. Natural products obtained from various plants that inhibit the generation of intracellular AGEs [28,121,160,161,187]: (a) quercetin, (b) chrisin, (c) genistein, (d) (+)-catechin, (e) (−)-epicatechin, (f) epigallocatechin-3-gallate, (g) p-hydroxy cinnamic acid (p-coumaric acid), (h) aspalatin, (i) resveratrol, (j) hesperidin, (k) imperialine, (l) curcumin, (m) piperine, and (n) diphlorethohydroxycarnalol.
Figure 14. Natural products obtained from various plants that inhibit the generation of intracellular AGEs [28,121,160,161,187]: (a) quercetin, (b) chrisin, (c) genistein, (d) (+)-catechin, (e) (−)-epicatechin, (f) epigallocatechin-3-gallate, (g) p-hydroxy cinnamic acid (p-coumaric acid), (h) aspalatin, (i) resveratrol, (j) hesperidin, (k) imperialine, (l) curcumin, (m) piperine, and (n) diphlorethohydroxycarnalol.
Ijms 26 09118 g014
Figure 15. Natural products in various plants that inhibit extracellular AGEs-RAGE/TLR4 signaling to attenuate cytotoxicity [28,121,160,161,187]; (a) ferulic acid, (b) caffeic acid, (c) gallic acid, (d) luteolin, (e) apigenin, (f) fisetin, (g) naringenin, and (h) naringin.
Figure 15. Natural products in various plants that inhibit extracellular AGEs-RAGE/TLR4 signaling to attenuate cytotoxicity [28,121,160,161,187]; (a) ferulic acid, (b) caffeic acid, (c) gallic acid, (d) luteolin, (e) apigenin, (f) fisetin, (g) naringenin, and (h) naringin.
Ijms 26 09118 g015
Table 1. Crude drugs obtained from Hangeshashinto, including Hange, Ogon, Oren, Kankyo, Ninjin, Kanzo, and Taiso. The names of crude drugs are described using Japanese, English, and Latin names.
Table 1. Crude drugs obtained from Hangeshashinto, including Hange, Ogon, Oren, Kankyo, Ninjin, Kanzo, and Taiso. The names of crude drugs are described using Japanese, English, and Latin names.
No.Japanese NameEnglish DescriptionDescription of Latin Name
1HangePinellia tuberTuber of Pinellia ternate Breitenbach, Araceae
2OgonScutellaria rootRoot of Scutellaria baicalensis Georgi, Labiatae
3OrenCoptidis rhizomeRhizome of Coptis japonica Makino, Ranueculaceae
4KankyoGinger rhizomeSteamed rhizome of Zingiber officinale Roscoe, Zingiberaceae
5NinjinGinseng rootRoot of Panax ginseng C.A. Meyer, Araliaceae
6KanzoGlycyrrhiza rootRoot or stolon of Glycyrrhiza uralensis Fischer, Laguminosae
7TaisoJujube fruitFruit of Zizpbus jujura Miller var. inermis Pehder, Rhamnaceae
Table 2. Crude drugs obtained from Hangeshashinto and their natural products can inhibit the generation of the intracellular AGEs to attenuate cytotoxicity. The crude drug names are described using Japanese and Latin names.
Table 2. Crude drugs obtained from Hangeshashinto and their natural products can inhibit the generation of the intracellular AGEs to attenuate cytotoxicity. The crude drug names are described using Japanese and Latin names.
No.Japanese NameDescription of Latin NameNatural ProductReference
1HangeTuber of Pinellia ternate Breitenbach, AraceaeNo informationNo information
2OgonRoot of Scutellaria baicalensis Georgi, Labiataequercetin[188,189]
genistein[190,191]
(+)-catechin[192,193]
epigallocatechin-3-gallate[194]
hesperidin[195]
p-coumaric acid[196]
curcumin[195,197]
3OrenRhizome of Coptis japonica Makino, Ranueculaceaequercetin[198]
4KankyoSteamed rhizome of Zingiber officinale Roscoe, ZingiberaceaeNo informationNo information
5NinjinRoot of Panax ginseng C.A. Meyer, Araliaceaequercetin[199,200]
(+)-catechin[201,202]
epigallocatechin-3-gallate[203,204]
p-coumaric acid[205,206]
6KanzoRoot or stolon of Glycyrrhiza uralensis Fischer, Laguminosaequercetin[43,207]
genistein[45]
(+)-catechin[208]
epigallocatechin-3-gallate[44,209]
p-coumaric acid[210]
7TaisoFruit of Zizpbus jujura Miller var. inermis Pehder, Rhamnaceaequercetin[211]
(+)-catechin[212]
(−)-epicatechin[212]
p-coumaric acid[213]
Table 3. Crude drugs in Hangeshashinto and their natural products that can inhibit extracellular AGEs-RAGE/TLR4 signaling to attenuate cytotoxicity. The names of crude drugs are described using Japanese and Latin names.
Table 3. Crude drugs in Hangeshashinto and their natural products that can inhibit extracellular AGEs-RAGE/TLR4 signaling to attenuate cytotoxicity. The names of crude drugs are described using Japanese and Latin names.
No.Japanese NameDescription of Latin NameNatural ProductReference
1HangeTuber of Pinellia ternate Breitenbach, AraceaeNo informationNo information
2OgonRoot of Scutellaria baicalensis Georgi, Labiataefisetin[235]
luteolin[236]
quercetin[188,189]
p-coumaric acid[196]
3OrenRhizome of Coptis japonica Makino, Ranueculaceaeluteolin[237]
quercetin[198]
4KankyoSteamed rhizome of Zingiber officinale Roscoe, ZingiberaceaeNo informationNo information
5NinjinRoot of Panax ginseng C.A. Meyer, Araliaceaeferulic acid[238]
caffeic acid[205,239]
gallic acid[47,240]
naringenin[241]
quercetin[199,200]
p-coumaric acid[205,208]
6KanzoRoot or stolon of Glycyrrhiza uralensis Fischer, Laguminosaecaffeic acid[242,243]
gallic acid[244]
apigenin[245]
naringenin[246]
quercetin[43,207]
p-coumaric acid[210]
7TaisoFruit of Zizpbus jujura Miller var. inermis Pehder, Rhamnaceaequercetin[211]
p-coumaric acid[212]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Takata, T.; Moriya, J.; Miyazawa, K.; Inoue, S.; Yamada, S.; Han, J.; Yang, Q.; Guo, X.; Mizuta, S.; Nakahashi, T.; et al. Potential of Natural Products in Hangeshashinto Water Extract on the Direct Suppression of Stomatitis Induced by Intra-/Extracellular Advanced Glycation End-Products. Int. J. Mol. Sci. 2025, 26, 9118. https://doi.org/10.3390/ijms26189118

AMA Style

Takata T, Moriya J, Miyazawa K, Inoue S, Yamada S, Han J, Yang Q, Guo X, Mizuta S, Nakahashi T, et al. Potential of Natural Products in Hangeshashinto Water Extract on the Direct Suppression of Stomatitis Induced by Intra-/Extracellular Advanced Glycation End-Products. International Journal of Molecular Sciences. 2025; 26(18):9118. https://doi.org/10.3390/ijms26189118

Chicago/Turabian Style

Takata, Takanobu, Junji Moriya, Katsuhito Miyazawa, Shinya Inoue, Sohsuke Yamada, Jia Han, Qian Yang, Xin Guo, Shuichi Mizuta, Takeshi Nakahashi, and et al. 2025. "Potential of Natural Products in Hangeshashinto Water Extract on the Direct Suppression of Stomatitis Induced by Intra-/Extracellular Advanced Glycation End-Products" International Journal of Molecular Sciences 26, no. 18: 9118. https://doi.org/10.3390/ijms26189118

APA Style

Takata, T., Moriya, J., Miyazawa, K., Inoue, S., Yamada, S., Han, J., Yang, Q., Guo, X., Mizuta, S., Nakahashi, T., Onai, N., Nakano, H., Masauji, T., & Motoo, Y. (2025). Potential of Natural Products in Hangeshashinto Water Extract on the Direct Suppression of Stomatitis Induced by Intra-/Extracellular Advanced Glycation End-Products. International Journal of Molecular Sciences, 26(18), 9118. https://doi.org/10.3390/ijms26189118

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

Article Metrics

Back to TopTop