Next Article in Journal
Isolation of Primary Human Saphenous Vein Endothelial Cells, Human Internal Thoracic Artery Endothelial Cells, and Human Adipose Tissue-Derived Microvascular Endothelial Cells from Patients Undergoing Coronary Artery Bypass Graft Surgery
Previous Article in Journal
Bioactive Compounds and Pharmacological Properties of the Polypore Fomes fomentarius, a Medicinal Wild Mushroom Collected from Morocco
Previous Article in Special Issue
Pharmacological Effect of Water-Extractable (Poly)Phenolic Polysaccharide–Protein Complexes from Prunus spinosa L. Wild Fruits
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Potential of Orally Administered Quercetin, Hesperidin, and p-Coumaric Acid in Suppressing Intra-/Extracellular Advanced Glycation End-Product-Induced Cytotoxicity in Proximal Tubular Epithelial Cells

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
Department of Pathology and Laboratory Medicine, Kanazawa Medical University, Uchinada 920-0293, Ishikawa, Japan
7
Department of Pathology, Kanazawa Medical University Hospital, Uchinada 920-0293, Ishikawa, Japan
8
Department of Spleen and Stomach Diseases, First Affiliated Hospital of Hebei University of Chinese Medicine, Shijiazhuang 050011, China
9
Hebei Key Laboratory of Turbidity Toxin Syndrome, Shijiazhuang 050011, China
10
Research Center, First Affiliated Hospital of Hebei University of Chinese Medicine, Shijiazhuang 050011, China
11
Inoue Iin Clinic, Kusatsu 525-0034, Shiga, Japan
12
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), 9216; https://doi.org/10.3390/ijms26189216
Submission received: 7 August 2025 / Revised: 14 September 2025 / Accepted: 19 September 2025 / Published: 21 September 2025

Abstract

Advanced glycation end-products (AGEs) are associated with the dysfunction of proximal tubular epithelial (PTE) cells in lifestyle diseases. Urinary stones induce cytotoxicity in PTE cells, and various medicines have been developed to mitigate or prevent their generation/accumulation. The leaves of Quercus salicina Blume/Q. stenophylla Makino—used in Japanese folk medicine—contain quercetin, hesperidin, and p-hydroxycinnamic (p-coumaric) acid, which can suppress the cytotoxicity of intra-/extracellular AGEs. This review investigated the effects of quercetin, hesperidin, and p-coumaric acid on PTE cells in terms of their metabolism following oral administration and the associated organs and bacteria. Current evidence indicates that, in PTE cells, non-metabolized quercetin and p-coumaric acid may suppress intra-/extracellular AGE-induced cytotoxicity, whereas the metabolites of quercetin and hesperidin may inhibit the generation of AGEs. However, little is known of the effects of p-coumaric acid metabolites. Quercetin, hesperidin, and p-coumaric acid may collectively suppress the cytotoxicity of intra-/extracellular AGEs in PTE cells. This review on the current paradigm of Q. salicina/Q. stenophylla extract provides a useful baseline for the design of further preclinical and clinical investigations.

1. Introduction

Proximal tubular epithelial (PTE) cells in the kidney regulate the reabsorption of minerals, glucose, amino acids, and raw urine, and regulate the volume of urine [1,2,3]. They are commonly damaged by various types of minerals contained in urinary stones, such as sodium and calcium, inflammation induced by various cytokines and intracellular proteins, and reactive oxygen species (ROS) [1,2,3,4,5]. PTE cell damage and dysfunction have been associated with lifestyle-related diseases (LSRDs) such as diabetes mellitus (DM), cardiovascular disease (CVD), hypertension, steatotic liver disease (SLD), and metabolic dysfunction–associated SLD (MASLD) [6]. Some researchers have revealed mechanisms by which DM induces and promotes the formation of urinary stones [3]. PTE cell death and dysfunction are likely associated with intra-/extracellular advanced glycation end-products (AGEs) [6,7,8,9]. Intracellular AGEs induce inflammation, cell death, and dysfunction in various organs such as the pancreas, liver, brain, and gut [8,9,10,11,12]. In HK-2 cells, a PTE cell line, the generation and accumulation of Nε-carboxymethyl-lysine (CML)-modified proteins were associated with impaired autophagy and the induction of cytotoxicity, such as inflammation and fibrosis [7]. We hypothesized that various AGEs may modify heat shock protein 90 (HSP90), which is associated with urinary stone adhesion [6]. Various types of AGE receptors are expressed on PTE cells, including the receptor for AGEs (RAGE) [13,14], Toll-like receptor 4 (TLR4) [15,16], and megalin [17,18,19,20]. Extracellular AGEs (e.g., in blood, saline, and urine [8,9,10,11,12]) interact with RAGE and TLR4 to induce inflammation and PTE cell death, whereas megalin stimulates the endocytosis system to promote AGE degradation [13,15,17,19]. Based on various studies of cell survival, PTE cell-related cytotoxicity and dysfunction may be mitigated by inhibiting the accumulation of intracellular AGEs [21,22,23,24,25,26,27,28] and blocking AGE-RAGE/TLR4 signaling [13,14,15,16,17].
Although various medicines have been developed for urinary stone adhesion and inflammation [6], this study focused on the effects of Quercus salicina Blume/Q. stenophylla Makino leaf extract [29,30,31,32], “Japanese folk medicine,” against modern European and Kampo medicines (traditional Japanese medicine influenced by traditional Chinese medicine) [29,30,31,32,33,34,35]. The aqueous extract of Q. salicina/Q. stenophylla leaves has been prescribed to remove urinary stones since the early modern period in Japan [29]. In the 20th century, Urocalun was developed based on this folk medicine for traditional and industrial applications [29]. The present review specifically investigates the roles of the components of Q. salicina/Q. stenophylla leaves—quercetin, hesperidin, and p-hydroxycinnamic (p-coumaric) acid [29,30,31,32]—in AGE-related cytotoxicity, which have been shown to inhibit (i) the generation of intracellular AGEs [36,37] and (ii) AGE-RAGE/TLR4 signaling [38,39,40,41]. Intracellular AGE generation is suppressed via the “carbonyl trap systems” of quercetin, hesperidin, and p-coumaric acid, which require a flavonoid skeleton and a phenolic acid containing a carboxyl group [12,21,22,23]. Accordingly, we hypothesized that quercetin, hesperidin, and p-coumaric acid would exert the same effects to protect PTE cells from intra-/extracellular AGE-related cytotoxicity. To test this hypothesis, we predicted and considered various conditions (see Section 5.6 and Section 5.7). We assessed the structural changes in compounds transported into the cells, considering an oral route of administration [29,30,31,32] and that (i) quercetin, hesperidin, and p-coumaric acid may not be metabolized or (ii) their metabolites are transported into PTE cells [42,43,44,45,46,47,48]. The metabolites of quercetin and hesperidin, which exhibit a flavonoid skeleton with a carbonyl group, can be transported into PTE cells to inhibit the generation of intracellular AGEs [12]. Similarly, high amounts of p-coumaric acid can avoid complete metabolism and be transported into the cells [40] to potentially suppress intracellular AGE generation and AGE-RAGE/TLR4 signaling [11,12]. This review identifies the key advances and gaps in this field, providing a useful reference for further investigation into the clinical applicability and translation of Q. salicina/Q. stenophylla leaf extract for mitigating PTE cell dysfunction/death and maintaining renal function.

2. Urinary Stone/Inflammation in PTE Cells and Roles of Q. salicina/Q. stenophylla Leaf Extract

2.1. Cytotoxicity in PTE Cells

The kidney is the major organ for reabsorbing various essential materials such as minerals, glucose, amino acids, and peptides through the PTE cells [1,2,3,4,5]. PTE cell damage is closely associated with senescence, high urine pressure and volume, excessive minerals, inflammation, ROS, and urinary stone adhesion (Figure 1). Urinary stones can directly damage PTE cells, requiring treatment with approved medicines [3,4,5,6,49,50,51,52]. Urinary stones induce inflammation and the upregulation of proteins associated with apoptosis, such as HSP90 and annexin [4,5,6]. Although the mechanisms by which LSRDs such as DM, CVD, SLD, and MASLD occur and promote urinary stone formation remain unclear, some researchers have reported a relationship between DM and urinary stone production [3]. They noted that sodium-glucose cotransporter-2 (SGLT2) inhibition reduces urinary stone formation. SGLT2 inhibitors increase glycosuria and calcium excretion, which may help explain the mechanisms by which DM promotes urinary stone production. However, whether other LSRDs induce urinary stone formation remains unclear, although they are associated with their generation. In contrast, various molecules can interact with the RAGE and TLR4 expressed on PTE cells to induce inflammation and cell death [53,54,55]. PTE cell damage is thus associated with inflammation from LARDs. Moreover, high intra-/extracellular AGE expression in LSRDs induces PTE cell cytotoxicity and dysfunction (see Section 4) [6].

2.2. Medicines Against Urinary Stones

Among the modern European medicines developed from the 18th century, tamsulosin (Astellas Pharma Inc., Tokyo, Japan) [56], magnesium oxide (Maruishi Pharmaceutical Co., Ltd., Osaka, Japan) [57], potassium-/sodium citrate hydrate (Nippon Chemiphar Co., Ltd., Tokyo, Japan) [58], tiopronin (Travere Therapeutics Inc., San Diego, CA, USA) [59], thiazide and thiazide-like diuretics (Merck & Co., Inc., Rahway, NJ, USA) [60], and febuxostat (Teijin Pharma Ltd., Tokyo, Japan) [61,62] have been prescribed for urinary stone adhesion and PTE cell inflammation. These medicines generally comprise one or more low-molecular-weight compounds. Urocalun (Nippon Shinyaku Co., Ltd., Kyoto, Japan)—developed based on the leaf extract of Q. salicina/Q. stenophylla [29,30,31,63] and Choreito (Tsumura & Co., Tokyo, Japan) [a “Kampo” medicine (see Section 5.1), Chinese name: Zhu-Ling-Tang] [64,65]—has also been prescribed to remove urinary stones from the proximal tubule. Although Urocalun is currently prescribed in clinical, hospital, and pharmaceutical practice in Japan, Q. salicina/Q. stenophylla leaf extract is applied largely as a folk medicine.

3. Intra-/Extracellular AGEs

3.1. Origins of AGEs

Protein glycation occurs commonly in vivo and in vitro [66,67,68]. Although the proteins in, for example, glycated hemoglobin (HbA1c) can be recovered, the saccharides in AGEs cannot be recovered along with their metabolites/non-enzymatic products and protein molecules [6,7,8,9,66,67,68]. We investigated the origin of AGE-related glucose, fructose, glyceraldehyde, glycolaldehyde, glyoxal, methylglyoxal, and 3-deoxyglucosone (Figure 2) [6,9,11,12]. Glyceraldehyde, glycolaldehyde, glyoxal, and methylglyoxal are produced from saccharides (e.g., glucose, fructose) and lipids [6,69,70,71,72,73]. Because AGEs and advanced lipoxidation end-products (ALEs) are generated from the same molecules, some AGEs and ALEs have the same characteristics [6,12]. Therefore, the excess intake of saccharides and lipids can induce LSRDs via the generation of AGEs and ALEs [6,12]. This suggests that AGEs and ALEs require more in-depth classification.

3.2. Structure and Category of AGEs

3.2.1. Structure of Free AGEs

AGEs are generated from saccharides and their metabolites/non-enzymatic products and proteins, and they react and combine with proteins through non-enzymatic reactions [6,9,11,12]. Accordingly, AGEs are considered modified proteins. A “free type of AGE” can be generated through amino acid reactions, including CML, Nε-carboxyethyl-lysine (CEL), methylglyoxal-hydro-imidazolone (MG-H1), and methylglyoxal-lysine dimer (MOLD) [6,9,10,11,12], several of which have been detected in humans/animals (e.g., organs, cells, blood, and urine), as well as in foods and beverages (Figure 3) [6,9,10,11,12]. AGEs have also been artificially synthesized (e.g., trihydroxy-triosidine, lysine-hydroxy-triosidine, arginine-hydroxy-triosidine, and a pyrrolopyridinium lysine dimer derived from glyceraldehyde) [74,75]. In 2023, Takeuchi et al. [76] reported two hypothetical structures for glyceraldehyde-derived AGEs (GA-AGEs), which they named “Toxic AGEs (TAGE)” in 2004; however, the structures have not been proved (see Section 3.2.2).

3.2.2. Classic and Novel AGEs

Studies on AGE catalyzation since the mid-20th century have identified glucose-derived (Glc-AGEs, AGE-1), GA-derived (AGE-2), glycolaldehyde-derived (Glycol-AGEs, AGE-3), methylglyoxal-derived (MGO-AGEs, AGE-4), glyoxal-derived (GO-AGEs, AGE-5), and 3-deoxyglucosone-derived AGEs (3DG-AGEs, AGE-6) [9,11,12]. However, novel types of AGEs with different compositions may require novel categories. Lactaldehyde-derived and melibiose-derived AGEs (MAGE) were recently synthesized [77,78]. Litwinowicz et al. [78] identified MAGEs in the blood of patients with alcoholic liver disease, which was categorized as AGE-10. Because the MGO-AGEs MG-H1 and argpyrimidine can be generated from glyceraldehyde and methylglyoxal, they should be categorized as GA- and MGO-AGEs [11,12]. Sugawa et al. [79] reported that CML can be generated from ribose via the production of glyoxal. Moreover, although glucoselysine is produced from glucose, it can also be produced from fructose [80,81]. Various researchers have proposed other categorizations based on cytotoxicity. For example, Takeuchi et al. [76] defined some GA-AGEs as “TAGE” in 2004 based on their recognition by in-house-prepared polyclonal anti-AGE antibodies, though the structures of the TAGEs remain unclear. Simkova et al. [82] classified GA- and Glycol-AGEs containing pyridinium moieties as TAGEs. Shen et al. [83] categorized GA-, MGO-, GO-, and 3DG-AGEs as TAGEs. In contrast, Lee et al. [6,84] categorized MOLD (one of the MGO-AGEs), which contains two amino acids and has been confirmed in humans, as a TAGE that may be associated with LSRDs (Figure 3b). Various researchers have defined TAGEs based on their cytotoxicity and associations with LSRDs [6,76,82,83,84].

3.2.3. Crude AGE Pattern

Various AGE structures may be generated from one source compound [85]. Although this phenomenon has been demonstrated using electrospray ionization (ESI)-mass spectrometry (MS) (ESI-MS) and matrix-assisted laser desorption/ionization-MS (MALDI-MS) in vitro (in cultured cells) and in tube experiments, no report has named or categorized this phenomenon. However, understanding this phenomenon may be important for future animal models and clinical investigations. Therefore, we recently termed it a crude AGE pattern [6,11,12]. In previous reports, Senaviralthna et al. [85] showed that MG-H1-, argpyrimidine-, and glyceraldehyde-derived pyridinium (GLAP)-modified proteins were generated in human pancreatic ductal epithelial PANC-1 cells treated with glyceraldehyde using ESI-MS (Figure 4). MG-H1- and argpyrimidine-modified HSP90 have been artificially generated through incubation of methylglyoxal and HSP90 at 37 °C [6].

3.2.4. Diverse AGE Patterns

Amino acid residues such as lysine and arginine are targets of non-enzymatic glycation, and proteins can be modified by various free types of AGEs in vitro and in vivo [86,87]. This has been demonstrated by researchers who analyzed AGEs using ESI-MS and MALDI-MS. These findings suggest that researchers should focus on the diverse types of AGEs that modify proteins, rather than on a single type of AGE modification, when analyzing AGE-induced cytotoxicity such as protein dysfunction. In contrast, a single type of AGE can modify various proteins, and these phenomena have been generally recognized [11,12]. However, these phenomena have not been named or categorized. Therefore, we proposed type 1 and 2 diverse AGE patterns (Figure 5) [11,12,88]. In the type 1 diverse AGE pattern, a certain AGE structure can modify a certain protein (1A: different types of AGE structures modified by the same amino acid residue; 1B: same type of AGE structures modified by different amino acid residues; 1C: different types of AGE structures modified by different amino acid residues; Figure 5a). These patterns can be validated using MS such as ESI-MS and MALDI-MS [11,12,87,88]. In the type 2 diverse AGE pattern, one type of AGE structure can modify various proteins (Figure 5b), which can be validated through Western blot analysis using anti-AGE antibodies.

3.2.5. Multiple AGE Patterns

Based on the type 1 and 2 diverse AGE patterns, we proposed type 1 and 2 multiple AGE patterns, as these phenomena have not yet been named or categorized in the field of AGE research (Figure 6) [11,12,87,88]. These patterns can be validated using MS such as ESI-MS and MALDI-MS [6,11,12,87,88]. In the type 1 multiple AGE pattern, certain AGE structures can modify one protein molecule but not a specific type of protein (Figure 6a). In the type 2 multiple AGE pattern, the AGE structure is modified by more than two proteins through intermolecular covalent bonds (Figure 6b).

3.3. Intracellular AGEs

Various organs such as the heart, liver, pancreas, and proximal tubule express intracellular AGEs, with crude, diverse, and multiple AGE patterns [11,12]. Intracellular AGEs can be generated and accumulated in normal human tissue, especially in patients with LSRDs. 2-Ammnonio-6-[4-(hydroxymethyl)-3-oxidopyridinium-1-yl]-hexanoate-lysine (4-hydroxymethyl-OP-lysine) modified with ryanodine receptor 2 (RyR2) and MG-H1 and G-H1 modified with the F-actin tropomyosin filament can induce beating dysfunction in cardiomyocytes [12]. The accumulation of CEL in skeletal muscles can promote the formation of adipose tissue [9,12]. Glucoselysine in the lens may promote cataract formation in model rats [80,81].

3.4. Extracellular AGEs

3.4.1. AGEs in the Blood, Saliva, and Urine from Various Organs

Once intracellular AGEs are generated in various organs, they can disperse and be secreted into the blood, saliva, and urine [9,11,12]. Some cytotoxic AGEs may leak into the body fluid, where they induce cytotoxicity and cell dysfunction. These AGEs interact with RAGE and TLR4 expressed on the cell membranes of various organs to induce cytotoxicity and inflammation. AGE-RAGE/TLR4 signaling is closely related to LSRD pathogenesis. Megalin expressed in the proximal tubule activates the endocytosis system to degrade AGEs. Since extracellular AGEs reflect the dynamics of intracellular AGEs, they are useful biomarkers of organ dysfunction and disease development [9,11,12]. Litwinowicz et al. [78] reported a novel MAGE biomarker for alcoholic liver disease. Katsuta et al. [89] found that blood CML and MG-H1 levels may be associated with kidney function after transplantation. The AGE structures can be determined through enzyme-linked immuno-sorbent assay (ELISA) and MS, such as ESI-/MALDI-MS.

3.4.2. AGEs in Extracellular Matrix

AGEs such as pentosidine can exist in the extracellular matrix [11,12,88]. Pentosidine-modified collagen tissue exhibits various dysfunctions compared with normal collagen tissue. We believe that intracellular collagens may be modified by AGEs and subsequently released.

3.4.3. Dietary AGEs

AGEs such as CML, CEL, and MG-H1 are produced in various foods and beverages, and can be consumed by humans [9,11,12,88]. These AGEs interact with RAGE and TLR4 in various organs as well as AGEs in the body fluid. The AGEs in the blood and urine may closely reflect diet and lifestyle.

3.5. Identification and Quantification of AGEs

The identification and quantification of AGEs are generally performed through fluorescence microscopy [10,11,12], Western blot [10,11,12,25], slot blotting [10,11,12,25,88,90], ELISA [10,11,12,91], MS such as gas chromatography-MS (GC-MS), ESI-MS, MALDI-MS [10,11,12,90], and nuclear magnetic resonance (NMR) [11,12,77]. Fluorescence microscopy and high-performance liquid chromatography (HPLC) are suitable since AGEs emit fluorescence [10,11,12]. Western blotting, slot blotting, and ELISA use anti-AGE antibodies to detect AGEs, though they do not provide structural information [10,11,12,25,90]. Standard AGE-modified proteins are prepared in test tubes, whereas normal recombinant proteins can be prepared in cells [10,88,92]. In the quantification, we recommend that slot blotting be performed based on Takata’s method, using Takata’s lysis buffer (modified or not) to stably probe AGE-modified proteins onto polyvinylidene fluoride (PVDF) membranes [10,25,88,90]. Therefore, this method was selected in thirteen studies from 2017 to 2025 [10,25,90]. For MS, quantification must be performed for AGEs with defined structures and with appropriate standards [10,11,12,89]. For quantification, GC-, ESI-, and MALDI-MS systems can be connected with HPLC platforms. NMR can identify AGEs but not quantify them [10,12,77,78]. Western blotting using only anti-AGE antibodies is inappropriate for detecting type 1 diverse and type 1 and 2 multiple AGE patterns; ESI-/MALDI-MS must instead be performed (Figure 5a and Figure 6) [6,11,12,88].

4. Cytotoxicity of Intra-/Extracellular AGEs in PTE Cells

4.1. Cytotoxicity of Intracellular AGEs in PTE Cells

Takahashi et al. [7] suggested that the generation/accumulation of intracellular AGEs (likely various Glc-AGEs) in a human PTE cell line (HK-2 cells) promoted the production of lysosomes to degrade AGE-modified proteins. They revealed that CML-modified proteins increased in HK-2 cells in which Atg5 was knocked down and incubated in high-glucose medium [7]. They insisted that intracellular AGE-modified proteins increase in the proximal tubular cells, where the dysfunction of autophagy is induced, and promote inflammation and fibrosis of cells. Although they analyzed only CML-modified proteins in HK-2 cells with Western blot, other types of AGE-modified proteins might be generated in these cells in their study because glucose is the origin for various types of AGEs (e.g., CEL) [9,11,12]. In contrast, the role of HSP90 as a receptor for urinary stones in HK-2 cells may be modified by various types of AGEs based on diverse and multiple AGE patterns [6]. Since MG-H1 and argpyrimidine-modification may cause dysfunction in human recombinant HSP90, we hypothesized that HSP90 expressed in tubular epithelial cells is modified by these AGEs under high-glucose and/or fructose conditions [6]. Current evidence suggests that various types of AGEs may be generated in PTE cells depending on the crude, diverse, and multiple AGE patterns (Figure 4, Figure 5 and Figure 6).

4.2. Cytotoxicity of Extracellular AGEs in PTE Cells

RAGE [13,14], TLR4 [15,16], and megalin [17,18,93] are expressed on the surface of PTE cells. Although megalin participates in the endocytosis of AGEs from the blood and urine [17,18], the AGE-RAGE/TLR4 axis induces cytotoxicity and inflammation in various organs such as the liver, heart, lung, and gut [9,13,14,15,16]. Chen et al. [14] reported that activation of AGE-RAGE signaling in the kidney upregulated the expression of tumor necrosis factor alpha (TNF-α), interleukin (IL)-1 beta (IL-1β), IL-6, and IL-18 in renal tissues, and increased the blood levels of these cytokines. AGE-RAGE signaling strongly activates the TLR4/myeloid differentiation factor 88 (MyD88)/nuclear factor-κβ (NF-κβ) pathway to exacerbate inflammation [14]. AGE-TLR4 interactions also induce inflammation [9]. Although urinary stones adhere to receptor proteins such as HSP90, annexins [4,5], and CD44 in PTE cells to induce inflammation, the roles of the AGE-RAGE/TLR4 axis in inflammation require further investigation. Finally, the soluble receptor for AGEs (sRAGE)—a cleaved-type RAGE released/secreted from cell membranes in the blood and urine—can also react with AGEs and potentially inhibit the cytotoxic effect associated with AGE-RAGE interactions [94].

5. Potential of Quercetin, Hesperidin, and p-Coumaric Acid for Suppressing Intra-/Extracellular AGE-Induced Cytotoxicity in PTE Cells

5.1. Q. salicina/Q. stenophylla Leaf Extract

Modern European and traditional medicines are commonly used in Japan. Traditional Japanese medical science often adopts Kampo [32,33,34,95,96] and folk medicines [29,30,31]. Traditional Chinese medical science involves a variety of therapeutic approaches, such as health management (diet and exercise) [97], crude drugs (natural medicines) [33,34,95,96,98,99], moxibustion/cupping treatment [32,33,34,95,96,99], and acupuncture [100,101,102], and was introduced in ancient Japan from the Chinese mainland and Korean peninsula and modified to establish Kampo (“Kam” or “Kan” in Japanese means “Han” in Chinese, referring to the ancient Chinese empire; “po” refers to the method of diagnosis or treatment; Figure 7). Kampo medicines contain multiple crude drugs, whereas original Japanese medicine is prepared using one crude drug (e.g., specific plant extract). Although much of traditional Japanese medical science is considered Kampo, Japanese folk medical science traditionally incorporates crude plant extracts [29,30,31]. Q. salicina/Q. stenophylla leaf extract has been used in folk medicine to remove urinary stones from the proximal tubules since the early modern period in Japan. It is unclear whether this extract was used as a regular authentic medicine in other areas such as China, Korea, and Vietnam, as this information did not reach Japan during the ancient to early modern periods. Therefore, we believe that Q. salicina/Q. stenophylla leaf extract is one of the Japanese folk medicines. The most important contemporary derivative, Urocalun, was only developed from the aqueous extract of Q. salicina/Q. stenophylla in the 20th century [29,30,31].

5.2. Limited Data on the Effects of Q. salicina/Q. stenophylla Leaf Extract Components

We focused on the possibility that medicines selected to prevent and treat urinary stones may also suppress intra-/extracellular AGE-induced cytotoxicity in PTE cells. However, no study has assessed the inhibitory effects of modern European medicines such as tamsulosin [56], magnesium oxide [57], potassium-/sodium citrate hydrate [58], tiopronin [59], thiazide and thiazide-like diuretics [60], and febuxostat [61,62] and traditional medicines such as Urocalun [29] and Choreito [64,65] on the generation of intracellular AGEs or AGE-RAGE/TLR4 signaling. Demonstrating that these urinary stone medicines exert anti-AGE effects in PTE cells would provide important information for the treatment and prevention of PTE cytotoxicity. Although the chemical structures of modern European medicines have been identified, their potential anti-AGE effects remain unclear [56,57,58,59,60,61,62]. Urocalun and Choreito contain multiple components, some of which have been identified, and they are known to include anti-AGE compounds. At this stage, we focused on the components of Q. salicina/Q. stenophylla leaf extract, which are major constituents of Urocalun. They are categorized as Japanese folk medicines rather than Kampo medicines, as research on folk medicines has not been as actively promoted compared with Kampo medicines. Accordingly, little is known of the effects of Q. salicina/Q. stenophylla leaf extract on AGE expression and signaling, despite its widespread adoption along with Urocalun. Because of the lack of studies, we characterized the effects of each compound based on the available mechanistic research. Therefore, direct treatments using the extract are unsuitable for describing the in vitro effects of the various components, whereas treatments with the respective components or their metabolites may effectively elucidate their effects. This approach is especially important given the metabolic processes that natural compounds and their metabolites are exposed to during transport to PTE cells following oral administration.

5.3. Quercetin, Hesperidin, and p-Coumaric Acid

Quercetin, hesperidin, and p-coumaric acid have been detected in various extracts of Q. salicina/Q. stenophylla (Figure 8) [6,29,30,31,32]. Quercetin [103,104], hesperidin [104,105,106,107], and p-coumaric acid [108,109] are produced by and accumulate in various plants. Although quercetin and hesperidin are flavonoids, the former has an aglycon structure and the latter has a glycoside structure [12,110,111]. Hesperetin-7-O-rutinoside, because hesperetin (aglycon-structured hesperidin) is modified with two hexoses. Although various flavonoids (e.g., rutin, naringin, quercetin, hesperidin, and myricetin) and phenolic acids (e.g., gallic acid, pyrogallol, caffeic acid, p-coumaric acid, and m-coumaric acid) have been isolated [32], we selected quercetin and hesperidin as typical examples of aglycon- and glycoside-type flavonoids, and p-coumaric acid as an example of phenolic acid. Although Aung et al. [32] characterized the leaf contents (μg/g) of various natural compounds such as flavonoids and phenolic acids, this study focused only on structure.

5.4. Suppression of Intracellular AGE Generation by Quercetin, Hesperidin, and p-Coumaric Acid in Cells Other than PTE Cells

Various studies have investigated the effects of low-molecular-weight plant compounds in suppressing intracellular AGE accumulation [112]. Quercetin, hesperidin, and p-coumaric acid have each been reported to inhibit the generation of intracellular AGEs through the molecular suppression of AGE generation [6,12,21,22,23,24,25,26]. The process is primarily facilitated through the “carbonyl trap system” (not the activation of glyoxalase [6,12,36,37]), which targets the source compounds such as glyceraldehyde, glycolaldehyde, methylglyoxal, glyoxal, and 3-deoxyglucosone [6,12]. The carbonyl trap system targets the ketone and aldehyde groups of these source compounds and functions as a non-enzymatic reaction (a simple organic chemical reaction that does not require enzymes). Many experiments have demonstrated that the carbonyl trap system can inhibit the generation of AGEs in vitro and in tube [6,12]. Quercetin, hesperidin, and p-coumaric acid may suppress the generation of various free AGEs, as well as diverse and multiple AGE patterns. The effects of quercetin and hesperidin are specifically determined by the flavonoid skeleton [110,111,113,114].

5.5. Inhibition of Extracellular AGE-RAGE/TLR4 Cell Signaling by Quercetin, Hesperidin, and p-Coumaric Acid in Cells Other than PTE Cells

To suppress cytotoxicity-related inflammation, various studies have investigated the effects of low-molecular-weight plant compounds on AGE-RAGE/TLR4 cell signaling [9,12]. Quercetin has been found to suppress the AGE-RAGE axis [38,115]. Quercetin-induced suppression of TLR4/NF-κβ signaling has been shown to inhibit inflammation and cell death in vitro and in vivo [116,117,118,119,120]. Therefore, we believe quercetin can suppress RAGE/TLR4-related cytotoxicity. However, because quercetin can exert cytotoxicity through RAGE, the inhibitor for the AGE-RAGE/TLR4 axis should be considered carefully [121,122]. Hesperidin can suppress RAGE/NF-κβ and TLR4/NF-κβ signaling in vitro and in vivo [39,123,124,125]. Various studies have indicated that p-coumaric acid can suppress the AGE-RAGE axis in vitro and in vivo [40,126]. Because p-coumaric acid can also suppress inflammation via TLR4/NF-κβ signaling in vivo [41,127], it may inhibit AGE-RAGE/TLR4-induced inflammation. The suppressive effects of quercetin, hesperidin, and p-coumaric acid on cytotoxicity via RAGE/TLR4 signaling have been demonstrated in non-clinical investigations.

5.6. The Issue of Whether Quercetin, Hesperidin, and p-Coumaric Acid Can Affect PTE Cells

There are no reports that orally administered quercetin, hesperidin, or p-coumaric acid suppress cytotoxicity of PTE cells induced by intra-/extracellular AGEs in vitro, in vivo (animal models), or in patients. Although these three compounds may inhibit the generation of intracellular AGEs and suppress extracellular AGE-RAGE/TLR4 signaling in PTE cells in vitro, whether these functions are exhibited in vivo or in clinical investigations remains unclear. However, (i) the carbonyl trap system can act regardless of cell type because it is a simple organic chemical reaction [6,12], and (ii) RAGE and TLR4 are expressed on PTE cells, with downstream NF-κβ signaling similar to other cell types [13,14,15,16]. Therefore, these compounds may protect PTE cells against intra-/extracellular AGE-induced cytotoxicity in vitro and in vivo. To hypothesize that orally administered quercetin, hesperidin, and p-coumaric acid protect PTE cells from AGE-induced cytotoxicity, we predicted and considered various conditions, as shown below. If quercetin, hesperidin, and p-coumaric acid cannot be transported into PTE cells, they will be unable to exert their anti-cytotoxic effects. This is an important issue that must be clarified to determine whether orally administered quercetin, hesperidin, and p-coumaric acid can exert anti-AGE effects in PTE cells in vivo and in patients. Based on this theme, we also need to consider the possibility that these compounds, or their metabolites, are transported into PTE cells and exert anti-AGE effects, since orally administered compounds are absorbed and metabolized [9].

5.7. Transportation Potential of Non-Metabolites and/or Metabolites of Quercetin, Hesperidin, and p-Coumaric Acid into PTE Cells

5.7.1. Absorption and Metabolization of Quercetin

A microbalance of quercetin has been detected in human blood [128]. This suggests that quercetin (i) can resist various metabolic systems and (ii) maintain the aglycon structure following metabolism by various enzymes and bacteria in different organs. Various quercetin metabolites have been recorded in human blood [129,130,131]. Although whole-metabolite profiles remain elusive, quercetin-3-O-glucoside (Q3G), quercetin 3-O-glucuronide (Q3GA), 3′-O-methylquercetin-O-glucuronide (3′-O-methyl-Q3GA), quercetin-4′-O-glucoside (Q4′G), quercetin-4′-O-glucuronide (Q4′GA), 3′-O-methylquercetin-4-O-glucuronide (3′-O-methyl-Q4′GA), quercetin-3′-glucuronide (Q3′GA), and quercetin-3′-O-sulfate (Q3S) have been reported in human blood (Figure 9) [129,130,131]. Quercetin can be metabolized into structures such as glycosides, glucuronides, and sulfates, though mainly non-metabolized quercetin is transported into PTE cells. Q3G and Q4′G can be transported to the small intestine via SGLT1 (Figure 10) [132,133,134]. Q3G and Q4′G are hydrolyzed via lactase phlorizin hydrolase (LPH) expressed on the membrane of small intestinal epithelial cells to produce quercetin, which can then be absorbed into PTE cells (Figure 10) [134,135,136]. Intracellular Q3G and Q4′G are hydrolyzed by β-glucosidase to produce quercetin, which is metabolized to glucuronide via phase II enzyme systems (Figure 10) [136]. Although little is known of quercetin-glucuronides, Q3GA, 3′-O-methyl-Q3GA, Q4′GA, 3′-O-methyl-Q4′GA, and Q3′GA may be produced in this step. Quercetin-glucuronides are dispersed into the lymph fluid and blood from the large intestine epithelial and hepatic parenchymal cells [136]. Quercetin-glucosides are transported from the small intestine into the large intestine, where bacteria hydrolyze them to produce quercetin [137,138]. The transport potential of quercetin has been confirmed in human colon Caco-2 cells (Figure 11) [139,140,141]. Murota et al. [142] reported that Q3G and Q4′G were likely absorbed into Caco-2 cells via SGLT1 as the transporter (Figure 11). In the large intestine, glucuronide-quercetins may be produced via β-galactosidase and phase II enzymes, and subsequently leak into lymph fluid and blood. We assessed the hepatic metabolism of quercetin [143,144,145,146,147]. Quercetin has been shown to protect against fibrosis and lipogenesis in hepatic tissue [143,144]. Kaci et al. [145] predicted that G3S may be produced as a metabolite. Hou et al. [146] suggested that quercetin metabolites may protect hepatic cells. Michala et al. [147] detected 3′-O-methylquercetin-7-O-glucuronide (3′-O-methyl-Q7GA) and 4′-O-methylquercetin-7-O-glucuronide in human hepatic parenchymal HepG2 cells treated with quercetin (4′-O-methyl-Q7GA) (Figure 12).

5.7.2. Absorption and Metabolization of Hesperidin

Hesperidin has never been recorded in the blood of humans, rats, or mice, suggesting that it cannot escape metabolism. Although hesperidin cannot be hydrolyzed by α-glucosidase in the small/large intestine, it can be hydrolyzed to hesperetin by bacterial β-glucosidase and absorbed into the small/large intestine (Figure 13) [148]. Hesperetin in the small/large intestine can be metabolized via the phase II enzyme system to hesperetin-glucuronide. Hesperidin can be transported into the small [149,150] and large intestine (Figure 13) [151,152]. Brand et al. [152] reported that UDP-glucuronosyltransferases (UGTs) and sulfotransferases (SULTs) in the human small/large intestines and liver can metabolize hesperidin. In the cytosolic fractions of these tissues, hesperetin-3′-O-glucuronide, hesperetin-7′-O-glucuronide, hesperetin-3′-O-sulfate, and hesperetin-7′-O-sulfate were identified following incubation with hesperidin (Figure 14) [151,152]. Hesperidin has been recorded in the liver of animal models, and its metabolites in a liver with fatty-liver characteristics suppressed inflammation and fibrosis [153,154]. In human hepatic parenchymal Hepa-RG and stellate LX-2 cells, hesperidin exerted antioxidant effects by increasing the protein levels of copper/zinc superoxide dismutase (SOD1), manganese superoxide dismutase (SOD2), and catalase, and by suppressing lipid oxidation [155]. These effects may have been associated with the anti-lipidation activities of its metabolites.

5.7.3. Absorption and Metabolization of p-Coumaric Acid

P-coumaric acid has been detected in human blood and can be absorbed into the small/large intestine because of its resistance to enzymatic and bacterial metabolism [156,157]. In plants, p-coumaric acid can be metabolized by some enzymes such as 4-coumaric CoA-ligase (4CL), hydroxycinnamoyl transferase (HCT), p-coumaroyl-shikimic acid-3-hydroxytransferase (C3′H), and caffeoyl shikimic esterase (CSE) to produce caffeic acid (in this pathway, p-coumaroyl-CoA, p-coumaroyl-shikimic acid, and caffeoyl-shikimic acid are produced as internal metabolites) (Figure 15) [158,159]. However, the metabolic dynamics of p-coumaric acid in the human body remain unclear. Because high amounts of p-coumaric acid are received with oral administration and detected in the blood, it is likely that small amounts may be metabolized owing to slight structural variation.

5.7.4. Transportation of Non-Metabolites or Metabolites of Quercetin, Hesperidin, and p-Coumaric Acid into PTE Cells

This study focused on the capacity of quercetin, hesperidin, p-coumaric acid, and their metabolites for transport into PTE cells. Based on their predicted structures, current evidence indicates that the metabolites of quercetin and hesperidin can be transported into PTE cells. Because quercetin and p-coumaric acid have been detected in human blood [129,156,157], the non-metabolized forms may be transported into PTE cells [although some molecules may be metabolized in the small or large intestine, the metabolites may eventually revert to their original structure (Figure 10 and Figure 11)]. In contrast, the evidence suggests that hesperidin cannot be transported into PTE cells because it has never been detected in human blood. Quercetin, quercetin-glucoside, and hesperidin are metabolized in the small/large intestine and liver, and their metabolites can also be transported into PTE cells (Figure 10, Figure 11, Figure 12, Figure 13 and Figure 14). Because p-coumaric acid shows resistance to enzymatic and bacterial metabolism [156,157], we predict that, along with its metabolites, some p-coumaric acid may be transported into PTE cells. In contrast, we cannot introduce the metabolites of p-coumaric acid.

5.7.5. Potential for Suppressing Intra-/Extracellular AGE-Induced Cytotoxicity by Non-Metabolites/Metabolites of Quercetin, Hesperidin, p-Coumaric Acid in PTE Cells

Based on Section 5.4 and Section 5.5, we hypothesize that orally administered quercetin and p-coumaric acid can suppress intra-/extracellular AGE-induced cytotoxicity in PTE cells, provided they are effectively transported into the target cells while maintaining their non-metabolic structures, although the metabolic processing of quercetin and p-coumaric acid requires further characterization [21,22,23,24,25,26,27,28,156,157]. Because the inhibition of intracellular AGEs by the carbonyl trap system of quercetin and p-coumaric acid can occur regardless of cell type, we believe they may inhibit intracellular AGE formation in PTE cells. Most hesperidin molecules may be metabolized before being transported; thus, it is unclear whether they participate in the suppression of intra-/extracellular AGE cytotoxicity in PTE cells. In contrast, the metabolites of quercetin and hesperidin may exert inhibitory effects owing to their robust flavonoid skeleton. Indeed, many flavonoid-based natural products have been shown to suppress intracellular AGE generation (Figure 8, Figure 9, Figure 10, Figure 11, Figure 12, Figure 13 and Figure 14) [12]. However, whether the metabolites of quercetin and hesperidin suppress cytotoxicity via the AGE-RAGE/TLR4 axis is unclear. In particular, further characterization of the metabolites of p-coumaric acid is needed to determine their anti-AGE effects in humans. If p-coumaric acid is able to be metabolized to caffeic acid in the human body, and it can be transported into PTE cells, it may inhibit the generation/accumulation of intracellular AGEs (Figure 15) [160,161].

6. Limitations

The complete composition of Q. salicina/Q. stenophylla leaf extract remains unclear, and the contents of quercetin, hesperidin, and p-coumaric acid may fluctuate depending on the collection site and year. This is a limitation compared with modern European medicines. Although we considered the metabolism of quercetin, hesperidin, and p-coumaric acid in the small and large intestine and liver, we did not provide a detailed section on pharmacokinetic data, nor did we include information comparing plasma and urine levels after oral dosing in humans or animals, metabolite profiles, renal-specific transport, the influence of gut bacteria, or bioactivity. The study neglected some factors, such as physiological reactions in the mouth and gut [162], where quercetin and hesperidin may be glycosylated and hydrolyzed, which should be addressed in a future review. Additionally, whether these compounds are present at pharmacologically relevant levels for the inhibition of intra-/extracellular AGE-related cytotoxicity requires experimental verification.

7. Conclusions

This study indicated that orally administered quercetin, hesperidin, and p-coumaric acid may suppress intra-extracellular AGE-mediated cytotoxicity in PTE cells. The non-metabolites of quercetin and p-coumaric acid and the metabolites of quercetin and hesperidin inhibit intracellular AGE generation via the carbonyl trap system. Although various types of AGEs are generated in PTE cells, the carbonyl trap system targets important source components. The non-metabolites of quercetin and p-coumaric acid modulate AGEs-RAGE/TLR4 signaling. However, the functions of the metabolites of quercetin, hesperidin, and p-coumaric acid in AGEs-RAGE/TLR4 signaling remain unclear. This study provides a useful basis on which to design future preclinical and clinical research on the performance and mechanisms of Q. salicina/Q. stenophylla leaf extract in suppressing intra-/extracellular AGE-induced cytotoxicity and treating related urinary disorders.

8. Future Directions

To advance the field, future in vitro and in vivo studies should consider how and to what extent quercetin, hesperidin, p-coumaric acid, and the other components of Q. salicina/Q. stenophylla, along with their metabolites, inhibit the generation/accumulation of intracellular AGEs and suppress the signaling of extracellular AGEs-RAGE/TLR4 in PTE cells. Subsequently, the effects of orally administered Q. salicina/Q. stenophylla leaf extract should be determined in animal models presenting with intra-/extracellular AGE-induced cytotoxicity in PTE cells.

Author Contributions

Conceptualization, T.T., J.M., K.M. and Y.M.; methodology, T.T., J.M., K.M., S.Y., J.H., Q.Y., X.G., T.N., S.M., S.I., T.M. and Y.M.; writing—original draft preparation, T.T., J.M., K.M., S.Y., J.H., Q.Y., X.G., T.N., S.M., S.I., T.M. and Y.M. Writing—review and editing, T.T., J.M., K.M., S.Y., J.H., Q.Y., X.G., T.N., S.M., S.I., T.M. and Y.M. Visualization for Figure 7, T.T., J.M., S.Y., J.H., Q.Y., X.G. and Y.M. Visualization for Figures without Figure 7, T.T. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by Japan Society for the Promotion of Science (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 research and reference checking were performed 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
CELNε-carboxyethyl-lysine
CMLNε-carboxymethyl-lysine
CSECaffeoyl shikimic esterase
CVDCardiovascular disease
DODIC3-Deoxyglucosone-derived imidazolium cross-link
DMDiabetes mellites
ELISAEnzyme-linked immuno-sorbent assay
ESIElectrospray ionization
ESI-MSElectrospray ionization mass-spectrometry
GAGlyceraldehyde
GCGas chromatography
GC–MSGas chromatography-mass spectrometry
GLAPGlyceraldehyde-derived pyridinium
GODICGlyoxal-derived imidazolium cross-link
G-H1Glyoxal-hydro-imidazolone
HCTHydroxycinnamoyl transferase
HPLCHigh-performance liquid chromatography
HSP90Heat shock protein 90
ILInterleukin
LSRDLifestyle-related disease
LPHLactase phlorizin hydrolase
MAGEMelibiose-derived advanced glycation-end product
MALDIMatrix-assisted laser desorption/ionization
MASLDMetabolic dysfunction–associated steatotic liver disease
MG-H1Methylglyoxal-hydro-imidazolone
MODICMethylglyoxal-derived imidazolium cross-link
MOLDMethylglyoxal-lysine dimer
MSMass spectrometry
MyD88Myeloid differentiation factor
NF-κβNuclear factor-κβ
NMRNuclear magnetic resonance
PTEProximal tubular epithelial
PVDFPolyvinylidene fluoride
RAGEReceptor for advanced glycation end-product
ROSReactive oxygen species
RyR2Ryanodine receptor 2
sRAGESoluble receptor for advanced glycation end-product
SGLT1Sodium-glucose cotransporter 1
SLDSteatotic liver disease
SODSuperoxide dismutase
SULTSulfotransferase
TAGEToxic advanced glycation end-product
TCMTraditional Chinese medicine
TLR4Toll-like receptor 4
TJMTraditional Japanese medicine
TNF-αTumor necrosis factor alpha
UGTGlucuronosyltransferase

References

  1. Plotuna, I.S.; Balas, M.; Golu, I.; Amzar, D.; Popescu, R.; Petrica, L.; Vlad, A.; Luches, D.; Vlad, D.C.; Vlad, M. The Use of Kidney Biomarkers, Nephrin and KIM-1, for the Detection of Early Glomerular and Tubular Damage in Patients with Acromegaly: A Case-Control Pilot Study. Diseases 2024, 12, 211. [Google Scholar] [CrossRef] [PubMed]
  2. Acharya, P.; Acharya, C.; Thongprayoon, C.; Hansrivijit, P.; Kanduri, S.R.; Kovvura, K.; Medaura, J.; Vaitla, P.; Garcia Anton, D.F.; Mekraksakit, P.; et al. Incidence and Characteristics of Kidney Stones in Patients on Ketogenic Diet: A Systematic Review and Meta-Analysis. Diseases 2021, 9, 39. [Google Scholar] [CrossRef] [PubMed]
  3. Dika, Ž.; Živko, M.; Kljaić, M.; Jelaković, B. SGLT2 Inhibitors and Their Effect on Urolithiasis: Current Evidence and Future Directions. J. Clin. Med. 2024, 13, 6017. [Google Scholar] [CrossRef] [PubMed]
  4. Heng, B.L.; Wu, F.Y.; Tong, X.Y.; Zou, G.J.; Ouyang, J.M. Corn Silk Polysaccharide Reduces the Risk of Kidney Stone Formation by Reducing Oxidative Stress and Inhibiting COM Crystal Adhesion and Aggregation. ACS Omega 2024, 9, 19236–19249. [Google Scholar] [CrossRef]
  5. Han, J.; Tong, X.Y.; Zheng, Y.Y.; Cheng, J.H.; Ouyang, J.M.; Li, K. Corn Silk Polysaccharides Before and After Selenization Reduced Calcium Oxalate Crystal-Induced HK-2 Cells Pyroptosis by Inhibiting the NLRP3-GSDMD Signaling Pathway. J. Inflamm. Res. 2025, 18, 3623–3638. [Google Scholar] [CrossRef]
  6. 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]
  7. Takahashi, A.; Takabatake, Y.; Kimura, T.; Maejima, I.; Namba, T.; Yamamoto, 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]
  8. 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]
  9. 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]
  10. 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]
  11. Takata, T.; Masauji, 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] [CrossRef]
  12. Takata, T.; Inoue, S.; Masauji, T.; Miyazawa, K.; 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] [PubMed]
  13. Zhang, J.; Gu, L.; Jiang, Y.; Ma, Y.; Zhang, Z.; Shen, S.; Shen, S.; Peng, Q.; Xiao, W. Artesunate-Nanoliposome-TPP, a Novel Drug Delivery System That Targets the Mitochondria, Attenuates Cisplatin-Induced Acute Kidney Injury by Suppressing Oxidative Stress and Inflammatory Effects. Int. J. Nanomed. 2024, 19, 1385–1408. [Google Scholar] [CrossRef] [PubMed]
  14. Chen, J.; Peng, H.; Chen, C.; Wang, Y.; Sang, T.; Cai, Z.; Zhao, Q.; Chen, S.; Lin, X.; Eling, T.; et al. NAG-1/GDF15 inhibits diabetic nephropathy via inhibiting AGE/RAGE-mediated inflammation signaling pathways in C57BL/6 mice and HK-2 cells. Life Sci. 2022, 311, 121142. [Google Scholar] [CrossRef] [PubMed]
  15. Li, R.; Guo, Y.; Zhang, Y.; Zhang, X.; Zhu, L.; Yan, T. Salidroside Ameliorates Renal Interstitial Fibrosis by Inhibiting the TLR4/NF-κB and MAPK Signaling Pathways. Int. J. Mol. Sci. 2019, 20, 1103. [Google Scholar] [CrossRef]
  16. Hu, M.; Wei, J.; Yang, L.; Xu, J.; He, Z.; Li, H.; Ning, C.; Lu, S. Linc-KIAA1737-2 promoted LPS-induced HK-2 cell apoptosis by regulating miR-27a-3p/TLR4/NF-κB axis. J. Bioenerg. Biomembr. 2021, 53, 393–403. [Google Scholar] [CrossRef]
  17. Oroojalian, F.; Rezayan, A.H.; Shier, W.T.; Abnous, K.; Ramezani, M. Megalin-targeted enhanced transfection efficiency in cultured human HK-2 renal tubular proximal cells using aminoglycoside-carboxyalkyl- polyethylenimine -containing nanoplexes. Int. J. Pharm. 2017, 523, 102–120. [Google Scholar] [CrossRef]
  18. Sawada, T.; Nagai, J.; Okada, Y.; Yumoto, R.; Takano, M. Gadolinium modulates gentamicin uptake via an endocytosis-independent pathway in HK-2 human renal proximal tubular cell line. Eur. J. Pharmacol. 2012, 684, 146–153. [Google Scholar] [CrossRef]
  19. Saito, A.; Takeda, T.; Sato, K.; Hama, H.; Tanuma, A.; Kaseda, R.; Suzuki, Y.; Gejyo, F. Significance of proximal tubular metabolism of advanced glycation end products in kidney diseases. Ann. N. Y. Acad. Sci. 2005, 1043, 637–643. [Google Scholar] [CrossRef]
  20. Goto, S.; Hosojima, M.; Kabasawa, H.; Saito, A. The endocytosis receptor megalin: From bench to bedside. Int. J. Biochem. Cell Biol. 2023, 157, 106393. [Google Scholar] [CrossRef]
  21. Bednarska, K.; Fecka, I. Aspalathin and Other Rooibos Flavonoids Trapped α-Dicarbonyls and Inhibited Formation of Advanced Glycation End Products In Vitro. Int. J. Mol. Sci. 2022, 23, 14738. [Google Scholar] [CrossRef]
  22. 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]
  23. 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]
  24. Yadav, N.; Palkhede, J.D.; Kim, S.Y. Anti-Glucotoxicity Effect of Phytoconstituents via Inhibiting MGO-AGEs Formation and Breaking MGO-AGEs. Int. J. Mol. Sci. 2023, 24, 7672. [Google Scholar] [CrossRef] [PubMed]
  25. 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] [PubMed]
  26. Maejima, I.; Takahashi, A.; Omori, H.; Kimura, T.; Takabatake, Y.; Saitoh, T.; Yamamoto, A.; Hamasaki, M.; Noda, T.; Isaka, Y.; et al. Autophagy sequesters damaged lysosomes to control lysosomal biogenesis and kidney injury. EMBO J. 2013, 32, 2336–2347. [Google Scholar] [CrossRef]
  27. 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]
  28. 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]
  29. Moriyama, M.T.; Suga, K.; Miyazawa, K.; Tanaka, T.; Higashioka, M.; Noda, K.; Oka, M.; Tanaka, M.; Suzuki, K. Inhibitions of urinary oxidative stress and renal calcium level by an extract of Quercus salicina Blume/Quercus stenophylla Makino in a rat calcium oxalate urolithiasis model. Int. J. Urol. 2009, 16, 397–401. [Google Scholar] [CrossRef]
  30. Yin, S.H.; Zhang, W.J.; Jiang, L.L.; Wang, G.Y.; Jeon, Y.J.; Ding, Y.; Li, Y. Protective effects of the secondary metabolites from Quercus salicina Blume against gentamicin-induced nephrotoxicity in zebrafish (Danio rerio) model. Comp. Biochem. Physiol. Part C Toxicol. Pharmacol. 2024, 283, 109952. [Google Scholar] [CrossRef]
  31. Kim, J.I.; Kim, H.H.; Kim, S.; Lee, K.T.; Ham, I.H.; Whang, W.K. Antioxidative compounds from Quercus salicina Blume stem. Arch. Pharm. Res. 2008, 31, 274–278. [Google Scholar] [CrossRef]
  32. Aung, T.; Bibat, M.A.D.; Zhao, C.C.; Eun, J.B. Bioactive compounds and antioxidant activities of Quercus salicina Blume extract. Food Sci. Biotechnol. 2020, 29, 449–458. [Google Scholar] [CrossRef]
  33. Chung, H.Y.; Yuasa, M.; Chen, F.S.; Yukawa, K.; Motoo, Y.; Arai, I. The status of education for integrative medicine in Japanese medical universities with special reference to Kampo medicines. Tradit. Kampo Med. 2023, 10, 123–131. [Google Scholar] [CrossRef]
  34. Chung, H.Y.; Moroi, M.; Hojo, Y.; Chen, F.S.; Yukawa, K.; Motoo, Y.; Arai, I. The Status of Nationwide Implementation of Integrative Medicine Programs by Japanese Local Government from a “Social Model” Viewpoint. Perspect. Integr. Med. 2024, 3, 98–105. [Google Scholar] [CrossRef]
  35. 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] [PubMed]
  36. Bhuiyan, M.N.I.; Mitsuhashi, S.; Sigetomi, 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]
  37. Lee, S.M.; Zheng, L.W.; Jung, Y.; Hwang, G.S.; Kim, Y.S. Effects of hydroxycinnamic acids on the reduction of furan and α-dicarbonyl compounds. Food Chem. 2020, 312, 126085. [Google Scholar] [CrossRef]
  38. 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]
  39. Hu, Q.; Qu, C.; Xiao, X.; Zhang, W.; Jiang, Y.; Wu, Z.; Song, D.; Peng, X.; Ma, X.; Zhao, Y. Flavonoids on diabetic nephropathy: Advances and therapeutic opportunities. Chin. Med. 2021, 16, 74. [Google Scholar] [CrossRef]
  40. Yu, X.D.; Zhang, D.; Xiao, C.L.; Zhou, Y.; Li, X.; Wang, L.; He, Z.; Reilly, J.; Xiao, Z.Y.; 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]
  41. 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]
  42. Sesink, A.L.; O’Leary, K.A.; Hollman, P.C. Quercetin glucuronides but not glucosides are present in human plasma after consumption of quercetin-3-glucoside or quercetin-4′-glucoside. J. Nutr. 2001, 131, 1938–1941. [Google Scholar] [CrossRef]
  43. Holder, C.L.; Churchwell, M.I.; Doerge, D.R. Quantification of soy isoflavones, genistein and daidzein, and conjugates in rat blood using LC/ES-MS. J. Agric. Food Chem. 1999, 47, 3764–3770. [Google Scholar] [CrossRef]
  44. Li, J.; Zhao, Z.; Deng, Y.; Li, X.; Zhu, L.; Wang, X.; Li, L.; Li, X. Regulatory Roles of Quercetin in Alleviating Fructose-Induced Hepatic Steatosis: Targeting Gut Microbiota and Inflammatory Metabolites. Food Sci. Nutr. 2024, 13, e4612. [Google Scholar] [CrossRef]
  45. Amaretti, A.; Raimondi, S.; Leonardi, A.; Quartieri, A.; Rossi, M. Hydrolysis of the rutinose-conjugates flavonoids rutin and hesperidin by the gut microbiota and bifidobacteria. Nutrients 2015, 7, 2788–2800. [Google Scholar] [CrossRef]
  46. Song, C.; Wang, Z.; Cao, J.; Dong, Y.; Chen, Y. Hesperetin alleviates aflatoxin B1 induced liver toxicity in mice: Modulating lipid peroxidation and ferritin autophagy. Ecotoxicol. Environ. Saf. 2024, 284, 116854. [Google Scholar] [CrossRef] [PubMed]
  47. Garrait, G.; Jarrige, J.F.; Blanquet, S.; Beyssac, E.; Cardot, J.M.; Alric, M. Gastrointestinal absorption and urinary excretion of trans-cinnamic and p-coumaric acids in rats. J. Agric. Food Chem. 2006, 54, 2944–2950. [Google Scholar] [CrossRef] [PubMed]
  48. Konishi, Y.; Hitomi, Y.; Yoshioka, E. Intestinal absorption of p-coumaric and gallic acids in rats after oral administration. J. Agric. Food Chem. 2004, 52, 2527–2532. [Google Scholar] [CrossRef] [PubMed]
  49. Nakazawa, Y.; Inoue, S.; Nakamura, Y.; Iida, Y.; Ishigaki, Y.; Miyazawa, K. High-salt diet promotes crystal deposition through hypertension in Dahl salt-sensitive rat model. Int. J. Urol. 2019, 26, 839–846. [Google Scholar] [CrossRef]
  50. Sakamoto, S.; Miyazawa, K.; Yasui, T.; Iguchi, T.; Fujita, M.; Nishimatsu, H.; Masaki, T.; Hasegawa, T.; Hibi, H.; Arakawa, T.; et al. Chronological changes in epidemiological characteristics of lower urinary tract urolithiasis in Japan. Int. J. Urol. 2019, 26, 96–101. [Google Scholar] [CrossRef]
  51. Sholan, R.; Aliyev, R.; Hashimova, U.; Karimov, S.; Bayramov, E. Urinary Stone Composition Analysis of 1465 Patients: The First Series from Azerbaijan. Arch. Iran. Med. 2024, 27, 618–623. [Google Scholar] [CrossRef]
  52. Ushimoto, C.; Sugiki, S.; Kunii, K.; Inoue, S.; Kuroda, E.; Akai, R.; Iwawaki, T.; Miyazawa, K. Dynamic change and preventive role of stress response via Keap1-Nrf2 during renal crystal formation. Free Radic. Biol. Med. 2023, 207, 120–132. [Google Scholar] [CrossRef]
  53. Sun, H.; Chen, J.; Hua, Y.; Zhang, Y.; Liu, Z. New insights into the role of empagliflozin on diabetic renal tubular lipid accumulation. Diabetol. Metab. Syndr. 2022, 14, 121. [Google Scholar] [CrossRef] [PubMed]
  54. Qin, Y.; Wu, S.; Zhang, F.; Zhou, X.; You, C.; Tan, F. N6-methyladenosine methylation regulator RBM15 promotes the progression of diabetic nephropathy by regulating cell proliferation, inflammation, oxidative stress, and pyroptosis through activating the AGE-RAGE pathway. Environ. Toxicol. 2023, 38, 2772–2782. [Google Scholar] [CrossRef] [PubMed]
  55. Li, T.; Sun, H.; Li, Y.; Su, L.; Jiang, J.; Liu, Y.; Jiang, N.; Huang, R.; Zhang, J.; Peng, Z. Downregulation of macrophage migration inhibitory factor attenuates NLRP3 inflammasome mediated pyroptosis in sepsis-induced AKI. Cell Death Discov. 2022, 8, 61. [Google Scholar] [CrossRef] [PubMed]
  56. Ali, A.I.; Abdelfadel, A.; Rohiem, M.F.; Hassan, A. Semirigid ureteroscopy and tamsulosin therapy as dilatation methods before flexible ureteroscopy: Evaluation and benefits. World J. Urol. 2024, 42, 75. [Google Scholar] [CrossRef]
  57. Gupta, M.; Gallante, B.; Bamberger, J.N.; Khusid, J.A.; Parkhomenko, E.; Chandhoke, R.; Capodice, J.; Atallah, W. Prospective Randomized Evaluation of Idiopathic Hyperoxaluria Treatments. J. Endourol. 2021, 35, 1844–1851. [Google Scholar] [CrossRef]
  58. Prezioso, D.; Strazzullo, P.; Lotti, T.; Bianchi, G.; Borghi, L.; Caione, P.; Carini, M.; Caudarella, R.; Ferraro, M.; Gambaro, G.; et al. Dietary treatment of urinary risk factors for renal stone formation. A review of CLU Working Group. Arch. Ital. Urol. Androl. 2015, 87, 105–120. [Google Scholar] [CrossRef]
  59. Mikel, C.C.; Goldfarb, D.S.; Ponte, A.; Steigelman, K.; Latyshev, S. Accurate 24-h urine cystine quantification for patients on cystine-binding thiol drugs. Urolithiasis 2022, 50, 721–727. [Google Scholar] [CrossRef]
  60. Bargagli, M.; Trelle, S.; Bonny, O.; Fuster, D.G. Thiazides for kidney stone recurrence prevention. Curr. Opin. Nephrol. Hypertens. 2024, 33, 427–432. [Google Scholar] [CrossRef]
  61. 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] [PubMed]
  62. Kraev, K.I.; Geneva-Popova, M.G.; Hristov, B.K.; Uchikov, P.A.; Popova-Belova, S.D.; Kraeva, M.I.; Basheva-Kraeva, Y.M.; Stoyanova, N.S.; Mitkova-Hristova, V.T. Celebrating Versatility: Febuxostat’s Multifaceted Therapeutic Application. Life 2023, 13, 2199. [Google Scholar] [CrossRef] [PubMed]
  63. Moriyama, M.T.; Miyazawa, K.; Noda, K.; Oka, M.; Tanaka, M.; Suzuki, K. Reduction in oxalate-induced renal tubular epithelial cell injury by an extract from Quercus salicina Blume/Quercus stenophylla Makino. Urol. Res. 2007, 35, 295–300. [Google Scholar] [CrossRef] [PubMed]
  64. Kawashima, N.; Ito, Y.; Sekiya, Y.; Narita, A.; Okuno, Y.; Muramatsu, H.; Irie, M.; Hama, A.; Takahashi, Y.; Kojima, S. Choreito formula for BK virus-associated hemorrhagic cystitis after allogeneic hematopoietic stem cell transplantation. Biol. Blood Marrow Transplant. 2015, 21, 319–325. [Google Scholar] [CrossRef]
  65. Sugihara, T.; Kamei, J.; Yasunaga, H.; Sasabuchi, Y.; Fujimura, T. Prescription of Choreito, a Japanese Kampo Medicine, with Antimicrobials for Treatment of Acute Cystitis: A Retrospective Cohort Study. Antibiotics 2022, 11, 1840. [Google Scholar] [CrossRef]
  66. Sharahili, A.Y.; Mir, S.A.; Aldosari, S.; Manzar, M.D.; Alshehri, B.; Al Othaim, A.A.; Alghofaili, F.; Madkhali, Y.; Albenasy, K.S.; Alotaibi, J.S. Correlation of HbA1c Level with Lipid Profile in Type 2 Diabetes Mellitus Patients Visiting a Primary Healthcare Center in Jeddah City, Saudi Arabia: A Retrospective Cross-Sectional Study. Diseases 2023, 11, 154. [Google Scholar] [CrossRef]
  67. Ajabnoor, G.M.A.; Bahijri, S.M.; Enani, S.M.; Alsheikh, L.; Ahmed, M.; Alhozali, A.; Al-Shali, K.; Eldakhakhny, B.M.; Alamoudi, A.A.; Al Ahmadi, J.; et al. Exploring the Validity of Available Markers and Indices in the Diagnosis of Nonalcoholic Fatty Liver Disease (NAFLD) in People with Type 2 Diabetes in Saudi Arabia. Diseases 2023, 11, 10. [Google Scholar] [CrossRef]
  68. Mengucci, T.; Chagas, E.F.B.; de Olivera Zanuso, B.; Quesada, K.; Dos Santis Haber, J.F.; Menegucci Zutin, T.L.; Felipe Pimenta, L.; Cressoni Araújo, A.; Landgraf Guiguer, E.; Rucco P Detregiachi, C.; et al. The Influence of Body Fat and Lean Mass on HbA1c and Lipid Profile in Children and Adolescents with Type 1 Diabetes Mellitus. Diseases 2023, 11, 125. [Google Scholar] [CrossRef]
  69. Maritin-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]
  70. 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]
  71. 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]
  72. 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]
  73. Maresch, C.C.; Stute, D.C.; Fleming, T.; Lin, J.; Hammes, H.P.; Linn, T. Hyperglycemia induces spermatogenic disruption via major pathways of diabetes pathogenesis. Sci. Rep. 2019, 9, 13074. [Google Scholar] [CrossRef] [PubMed]
  74. Tessier, F.J.; Monnier, V.M.; Sayre, L.M.; Kornfield, J.A. Triosidines: Novel Maillard reaction products and cross-links from the reaction of triose sugars with lysine and arginine residues. Biochem. J. 2003, 369, 705–719. [Google Scholar] [CrossRef] [PubMed]
  75. 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]
  76. 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]
  77. Fujimoto, S.; Murakami, Y.; Miyake, H.; Hayase, F.; Watanabe, H. Identification of a novel advanced glycation end product derived from lactaldehyde. Biosci. Biotechnol. Biochem. 2019, 83, 1136–1145. [Google Scholar] [CrossRef]
  78. Litwnowicz, 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]
  79. Sugawa, H.; Ikeda, T.; Tominaga, Y.; Katsuta, N.; Nagai, R. Rapid formation of Nε-(carboxymethyl)lysine (CML) from ribose depends on glyoxal production by oxidation. RSC Chem. Biol. 2024, 5, 1140–1146. [Google Scholar] [CrossRef]
  80. Ohno, R.I.; Ichimaru, K.; Tanaka, S.; Sugawa, H.; Katsuta, N.; Sakake, S.; Tominaga, Y.K.; Ban, I.; Shirakawa, J.I.; 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]
  81. 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]
  82. 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]
  83. Shen, C.Y.; Lu, C.H.; Cheng, C.F.; Li, K.J.; Kuo, Y.M.; Wu, C.H.; Liu, C.H.; Hsieh, S.C.; Tsai, C.Y.; Yu, C.L. 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]
  84. Lee, H.W.; Gu, M.J.; Lee, J.Y.; Lee, S.; Kim, Y.; Ha, S.K. Methylglyoxal-Lysine Dimer, an Advanced Glycation End Product, Induces Inflammation via Interaction with RAGE in Mesangial Cells. Mol. Nutr. Food Res. 2021, 65, e2000799. [Google Scholar] [CrossRef]
  85. 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] [PubMed]
  86. 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] [PubMed]
  87. 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. Biochim. Biophys. Acta 2011, 1812, 769–781. [Google Scholar] [CrossRef] [PubMed]
  88. 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]
  89. 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]
  90. 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]
  91. Nakashima, K.; Miyashita, H.; Yoshimitsu, H.; Fujiwara, Y.; Nagai, R.; Ikeda, T. Prenylflavonoids isolated from Epimedii Herba show inhibition activity against advanced glycation end-products. Front. Chem. 2024, 12, 1407934. [Google Scholar] [CrossRef]
  92. 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]
  93. Saito, A.; Nagai, R.; Tanuma, A.; Hama, H.; Cho, K.; Takeda, T.; Yoshida, Y.; Toda, T.; Shimizu, F.; Horiuchi, S.; et al. Role of megalin in endocytosis of advanced glycation end products: Implications for a novel protein binding to both megalin and advanced glycation end products. J. Am. Soc. Nephrol. 2003, 14, 1123–1131. [Google Scholar] [CrossRef] [PubMed]
  94. Vianello, E.; Beitrami, A.P.; Aleksova, A.; Janjusevic, M.; Fluca, A.L.; Corsi Romanelli, M.M.; La Sala, L.; Dozio, E. The Advanced Glycation End-Products (AGE)-Receptor for AGE System (RAGE): An Inflammatory Pathway Linking Obesity And Cardiovascular Diseases. Int. J. Mol. Sci. 2025, 26, 3707. [Google Scholar] [CrossRef] [PubMed]
  95. 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]
  96. He, Q.; Sawada, M.; Yamasaki, N.; Akazawa, S.; Furuta, H.; Uenishi, H.; Meng, X.; Nakahashi, T.; Ishigaki, Y.; Moriya, J. Neuroinflammation, Oxidative Stress, and Neurogenesis in a Mouse Model of Chronic Fatigue Syndrome, and the Treatment with Kampo Medicine. Biol. Pharm. Bull. 2020, 43, 110–115. [Google Scholar] [CrossRef]
  97. Chen, H.J.; Chen, D.Y.; Zhou, S.Z.; Chi, K.D.; Wu, J.Z.; Huang, F.L. Multiple tophi deposits in the spine: A case report. World J. Clin. Cases 2022, 10, 10647–10654. [Google Scholar] [CrossRef]
  98. Yuxi, G.; Ze, L.I.; Nan, C.; Xuemei, J.; Jie, W.; Hongyu, M.A.; Runyuan, Z.; Bolin, L.I.; Yucong, X.; Yanru, C.; 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]
  99. 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]
  100. 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]
  101. Han, J.; Guo, X.; Meng, X.J.; Zhang, J.; Yamaguchi, R.; Motoo, Y.; Yamada, S. Acupuncture improved lipid metabolism by regulating intestinal absorption in mice. World J. Gastroenterol. 2020, 26, 5118–5129. [Google Scholar] [CrossRef]
  102. 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]
  103. Marzouk, M.M.; Hegazi, N.M.; El Shabrawy, M.O.A.; Farid, M.M.; Kawashty, S.A.; Hussein, S.R.; Saleh, N.A.M. Discriminative Metabolomics Analysis and Cytotoxic Evaluation of Flowers, Leaves, and Roots Extracts of Matthiola longipetala subsp. livida. Metabolites 2023, 13, 909. [Google Scholar] [CrossRef] [PubMed]
  104. Tarroum, M.; Alfarraj, N.S.; Al-Qurainy, F.; Al-Hashimi, A.; Khan, S.; Nadeem, M.; Salih, A.M.; Shaikhaldein, H.O. Improving the Production of Secondary Metabolites via the Application of Biogenic Zinc Oxide Nanoparticles in the Calli of Delonix elata: A Potential Medicinal Plant. Metabolites 2023, 13, 905. [Google Scholar] [CrossRef] [PubMed]
  105. Nakahigashi, J.; Kurikami, M.; Iwai, S.; Iwamoto, S.; Kobayashi, S.; Kobayashi, E. Exploring the Pharmacokinetics and Gut Microbiota Modulation of Hesperidin and Nobiletin from Mandarin Orange Peel in Experimental Dogs: A Pilot Study. Metabolites 2024, 15, 3. [Google Scholar] [CrossRef]
  106. Fathallah, N.; El Deeb, M.; Rabea, A.A.; Almehmady, A.M.; Alkharobi, H.; Elhady, S.S.; Khalil, N. Ultra-Performance Liquid Chromatography Coupled with Mass Metabolic Profiling of Ammi majus Roots as Waste Product with Isolation and Assessment of Oral Mucosal Toxicity of Its Psoralen Component Xanthotoxin. Metabolites 2023, 13, 1044. [Google Scholar] [CrossRef]
  107. Fujitaka, Y.; Hamada, H.; Uesugi, D.; Kuboki, A.; Shimoda, K.; Iwaki, T.; Kiriake, Y.; Saikawa, T. Synthesis of Daidzein Glycosides, α-Tocopherol Glycosides, Hesperetin Glycosides by Bioconversion and Their Potential for Anti-Allergic Functional-Foods and Cosmetics. Molecules 2019, 24, 2975. [Google Scholar] [CrossRef]
  108. Gajić Umiljendić, J.; Sarić-Krsmanović, M.; Radivojević, L.; Šantrić, L.; Šćepanović, M.; Šoštarčić, V.; Đorđević, T. Phytochemical analysis of Asclepias syriaca L. leaf extract and its potential phytotoxic effect on some invasive weeds. Sci. Rep. 2025, 15, 6752. [Google Scholar] [CrossRef]
  109. Weng, V.; Cardeira, M.; Bento-Silva, A.; Serra, A.T.; Brazinha, C.; Bronze, M.R. Arabinoxylan from Corn Fiber Obtained through Alkaline Extraction and Membrane Purification: Relating Bioactivities with the Phenolic Compounds. Molecules 2023, 28, 5621. [Google Scholar] [CrossRef]
  110. Wang, X.; Ouyang, Y.Y.; Liu, J.; Zhao, G. Flavonoid intake and risk of CVD: A systematic review and meta-analysis of prospective cohort studies. Br. J. Nutr. 2014, 111, 1–11. [Google Scholar] [CrossRef]
  111. Li, T.; Zhao, Y.; Yuan, L.; Zhang, D.; Feng, Y.; Hu, H.; Hu, D.; Liu, J. Total dietary flavonoid intake and risk of cardiometabolic diseases: A dose-response meta-analysis of prospective cohort studies. Crit. Rev. Food Sci. Nutr. 2024, 64, 2760–2772. [Google Scholar] [CrossRef]
  112. Silva, R.M.G.D.; Barbosa, F.C.; Santos, H.H.; 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]
  113. Boojar, M.M.A.; Boojar, M.M.A.; Firouzivand, Y. In vitro Investigation of the Anti-Diabetic Effects of Imperialine on Beta-TC6 Pancreatic and C2C12 Skeletal Muscle Cell Lines. J. Appl. Biotechnol. Rep. 2021, 8, 337–345. [Google Scholar]
  114. Cha, S.H.; Hwang, Y.; Heo, S.J.; Jun, H.S. Diphlorethohydroxycarmalol Attenuates Methylglyoxal-Induced Oxidative Stress and Advanced Glycation End Product Formation in Human Kidney Cells. Oxid. Med. Cell. Longev. 2018, 2018, 3654095. [Google Scholar] [CrossRef] [PubMed]
  115. 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]
  116. 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]
  117. Gong, X.; Huang, Y.; Ma, Q.; Jiang, 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]
  118. Zhang, H.X.; Li, Y.Y.; Liu, Z.J.; Wang, J.F. 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]
  119. 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]
  120. 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]
  121. Duan, Z.L.; Wang, Y.J.; Lu, Z.H.; Tian, L.; Xia, Z.Q.; Wang, K.L.; Chen, T.; Wang, R.; Feng, Z.Y.; Shi, G.P.; et al. Wumei Wan attenuates angiogenesis and inflammation by modulating RAGE signaling pathway in IBD: Network pharmacology analysis and experimental evidence. Phytomedicine 2023, 111, 154658. [Google Scholar] [CrossRef]
  122. Lan, C.Y.; Chen, S.Y.; Kuo, C.W.; Lu, C.C.; Yen, G.C. Quercetin facilitates cell death and chemosensitivity through RAGE/PI3K/AKT/mTOR axis in human pancreatic cancer cells. J. Food Drug Anal. 2019, 27, 887–896. [Google Scholar] [CrossRef] [PubMed]
  123. Abd Elmaaboud, M.A.; Estfanous, R.S.; Atef, A.; Kabel, A.M.; Alnemari, K.A.; Naguib, T.M.; Aisufyani, S.E.; Darwish, H.W.; Arab, H.H. Dapagliflozin/Hesperidin Combination Mitigates Lipopolysaccharide-Induced Alzheimer’s Disease in Rats. Pharmaceuticals 2023, 16, 1370. [Google Scholar] [CrossRef] [PubMed]
  124. Han, X.; Zhang, Y.; Zhang, L.; Zhuang, Y.; Wang, Y. Efficacy and molecular mechanisms of hesperidin in mitigating Alzheimer’s disease: A systematic review. Eur. J. Med. Chem. 2025, 283, 117144. [Google Scholar] [CrossRef] [PubMed]
  125. Hong, Y.; An, Z. Hesperidin attenuates learning and memory deficits in APP/PS1 mice through activation of Akt/Nrf2 signaling and inhibition of RAGE/NF-κB signaling. Arch. Pharm. Res. 2018, 41, 655–663. [Google Scholar] [CrossRef]
  126. 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]
  127. 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]
  128. Setchell, K.D.; Brown, N.M.; Desai, P.; Zimmer-Nechemias, L.; Wolfe, B.E.; Brashear, W.T.; Kirschner, A.S.; Cassidy, A.; Heubi, J.E. Bioavailability of pure isoflavones in healthy humans and analysis of commercial soy isoflavone supplements. J. Nutr. 2001, 131, 1362S–1375S. [Google Scholar] [CrossRef]
  129. Nakamura, T.; Murota, K.; Kumamoto, K.; Kumamoto, S.; Misumi, K.; Bando, N.; Ikushiro, S.; Takahashi, N.; Sekido, K.; Kato, Y.; et al. Plasma metabolites of dietary flavonoids after combination meal consumption with onion and tofu in humans. Mol. Nutr. Food Res. 2014, 58, 310–317. [Google Scholar] [CrossRef]
  130. Murota, K.; Matsuda, N.; Kashino, Y.; Fujikura, Y.; Nakamura, T.; Kato, Y.; Shimizu, R.; Okuyama, S.; Tanaka, H.; Koda, T.; et al. alpha-Oligoglucosylation of a sugar moiety enhances the bioavailability of quercetin glucosides in humans. Arch. Biochem. Biophys. 2010, 501, 91–97. [Google Scholar] [CrossRef]
  131. Walle, T.; Otake, Y.; Walle, U.K.; Wilson, F.A. Quercetin glucosides are completely hydrolyzed in ileostomy patients before absorption. J. Nutr. 2000, 130, 2658–2661. [Google Scholar] [CrossRef]
  132. Wolffram, S.; Blöck, M.; Ader, P. Quercetin-3-glucoside is transported by the glucose carrier SGLT1 across the brush border membrane of rat small intestine. J. Nutr. 2002, 132, 630–635. [Google Scholar] [CrossRef] [PubMed]
  133. Walgren, R.A.; Lin, J.T.; Kinne, R.K.; Walle, T. Cellular uptake of dietary flavonoid quercetin 4′-beta-glucoside by sodium-dependent glucose transporter SGLT1. J. Pharmacol. Exp. Ther. 2000, 294, 837–843. [Google Scholar] [CrossRef] [PubMed]
  134. Day, A.J.; Gee, J.M.; Dupont, M.S.; Johnson, I.T.; Williamson, G. Absorption of quercetin-3-glucoside and quercetin-4′-glucoside in the rat small intestine: The role of lactase phlorizin hydrolase and the sodium-dependent glucose transporter. Biochem. Pharmacol. 2003, 65, 1199–1206. [Google Scholar] [CrossRef] [PubMed]
  135. Sesink, A.L.A.; Arts, I.C.W.; Faassen-Peters, M.; Hollman, P.C.H. Intestinal uptake of quercetin-3-glucoside in rats involves hydrolysis by lactase phlorizin hydrolase. J. Nutr. 2003, 133, 773–776. [Google Scholar] [CrossRef]
  136. Németh, K.; Plumb, G.W.; Berrin, J.G.; Juge, N.; Jacob, R.; Naim, H.Y.; Williamson, G.; Swallow, D.M.; Kroon, P.A. Deglycosylation by small intestinal epithelial cell beta-glucosidases is a critical step in the absorption and metabolism of dietary flavonoid glycosides in humans. Eur. J. Nutr. 2003, 42, 29–42. [Google Scholar] [CrossRef]
  137. Byambaakhuu, N.; Duan, S.; Sa, R.; Yang, Q.L.; Xu, H.Y.; Shan, C.B.; Xu, R.H.; Ma, C.M. Characterization of intestinal bacteria for the production of quercetin and isoquercitrin from rutin. Arch. Microbiol. 2025, 207, 83. [Google Scholar] [CrossRef]
  138. Makino, T.; Shimizu, R.; Kanemaru, M.; Suzuki, Y.; Moriwaki, M.; Mizukami, H. Enzymatically modified isoquercitrin, alpha-oligoglucosyl quercetin 3-O-glucoside, is absorbed more easily than other quercetin glycosides or aglycone after oral administration in rats. Biol. Pharm. Bull. 2009, 32, 2034–2040. [Google Scholar] [CrossRef]
  139. Wang, D.; Jiang, Q.; Li, P.; Yu, C.; Yuan, R.; Dong, Z.; Meng, T.; Hu, F.; Wang, J.; Yuan, H. Orally Administrated Precision Nanomedicine for Restoring the Intestinal Barrier and Alleviating Inflammation in Treatment of Inflammatory Bowel Disease. ACS Appl. Mater. Interfaces 2025, 17, 10986–11001. [Google Scholar] [CrossRef]
  140. Sousa, A.; Kämpfer, A.A.M.; Schins, R.P.F.; Cavalho, F.; Fernandes, E.; Freitas, M. Protective effects of quercetin on intestinal barrier and cellular viability against silver nanoparticle exposure: Insights from an intestinal co-culture model. Nanotoxicology 2025, 19, 141–155. [Google Scholar] [CrossRef]
  141. Jantip, P.A.; Singh, C.K.; Klu, Y.A.K.; Ahmad, N.; Bolling, B.W. Peanut skin polyphenols inhibit proliferation of leukemia cells in vitro, and its A-type procyanidins selectively pass through a Caco-2 intestinal barrier. J. Food Sci. 2025, 90, e70018. [Google Scholar] [CrossRef] [PubMed]
  142. Murota, K.; Shimizu, S.; Chujo, H.; Moon, J.H.; Terao, J. Efficiency of absorption and metabolic conversion of quercetin and its glucosides in human intestinal cell line Caco-2. Arch. Biochem. Biophys. 2000, 384, 391–397. [Google Scholar] [CrossRef] [PubMed]
  143. Zhong, W.; Chen, T.; Chen, L.; Xing, Y.; Lin, H.; Xie, S.; Nawaz, M.; Huang, D.; Huang, Z.; Lu, J.; et al. Crippled Hepatocarcinogenesis Inhibition of Quercetin in Glycolysis Pathway with Hepatic Farnesoid X Receptor Deficiency. Curr. Pharm. Des. 2025, 31, 1800–1815. [Google Scholar] [CrossRef] [PubMed]
  144. Yakubo, S.; Abe, H.; Li, Y.; Kudo, M.; Kimura, A.; Wakabayashi, T.; Watanabe, Y.; Kimura, N.; Setsu, T.; Yokoo, T.; et al. Dasatinib and Quercetin as Senolytic Drugs Improve Fat Deposition and Exhibit Antifibrotic Effects in the Medaka Metabolic Dysfunction-Associated Steatotic Liver Disease Model. Diseases 2024, 12, 317. [Google Scholar] [CrossRef]
  145. Kaci, H.; Bakos, É.; Needs, P.W.; Kroon, P.A.; Valentová, K.; Poór, M.; Özvegy-Laczka, C. The 2-aminoethyl diphenylborinate-based fluorescent method identifies quercetin and luteolin metabolites as substrates of Organic anion transporting polypeptides, OATP1B1 and OATP2B1. Eur. J. Pharm. Sci. 2024, 196, 106740. [Google Scholar] [CrossRef]
  146. Hou, Y.; Chen, C.; Li, Z.; Wu, J.; Lyu, S.; Guo, D.; Liu, Y.; Liu, Y.; Hou, T. Protective effect of quercetin against macrophage-mediated hepatocyte injury via anti-inflammation, anti-apoptosis and inhibition of ferroptosis. Autoimmunity 2024, 57, 2350202. [Google Scholar] [CrossRef]
  147. Michala, A.S.; Pritsa, A. Quercetin: A Molecule of Great Biochemical and Clinical Value and Its Beneficial Effect on Diabetes and Cancer. Diseases 2022, 10, 37. [Google Scholar] [CrossRef]
  148. Kim, G.J.; Jang, Y.; Kwon, K.T.; Kim, J.W.; Kang, S.I.; Ko, H.C.; Lee, J.Y.; Apostolidis, E.; Know, Y.I. Jeju Citrus (Citrus unshiu) Leaf Extract and Hesperidin Inhibit Small Intestinal α-Glucosidase Activities In Vitro and Postprandial Hyperglycemia in Animal Model. Int. J. Mol. Sci. 2024, 25, 13721. [Google Scholar] [CrossRef]
  149. Stevens, Y.; Rymenant, E.V.; Grootaert, C.; Camp, J.V.; Possemiers, S.; Masclee, A.; Jonkers, D. The Intestinal Fate of Citrus Flavanones and Their Effects on Gastrointestinal Health. Nutrients 2019, 11, 1464. [Google Scholar] [CrossRef]
  150. Yamada, M.; Tanabe, F.; Arai, N.; Mitsuzumi, H.; Miwa, Y.; Kubota, M.; Chaen, H.; Kibata, M. Bioavailability of glucosyl hesperidin in rats. Biosci. Biotechnol. Biochem. 2006, 70, 1386–1394. [Google Scholar] [CrossRef]
  151. Van Rymenant, E.; Salden, B.; Voorspoels, S.; Jacobs, G.; Noten, B.; Pitart, J.; Possemiers, S.; Smagghe, G.; Grootaert, C.; Van Camp, J. A Critical Evaluation of In Vitro Hesperidin 2S Bioavailability in a Model Combining Luminal (Microbial) Digestion and Caco-2 Cell Absorption in Comparison to a Randomized Controlled Human Trial. Mol. Nutr. Food Res. 2018, 62, e1700881. [Google Scholar] [CrossRef]
  152. Brand, W.; Boersma, M.G.; Bik, H.; Hoek-van den Hil, E.F.; Vervoort, J.; Barron, D.; Meinl, W.; Glatt, H.; Williamson, G.; van Bladeren, P.J.; et al. Phase II metabolism of hesperetin by individual UDP-glucuronosyltransferases and sulfotransferases and rat and human tissue samples. Drug Metab. Dispos. 2010, 38, 617–625. [Google Scholar] [CrossRef]
  153. Morshedzadeh, N.; Ramezani Ahmadi, A.; Behrouz, V.; Mir, E. A narrative review on the role of hesperidin on metabolic parameters, liver enzymes, and inflammatory markers in nonalcoholic fatty liver disease. Food Sci. Nutr. 2023, 11, 7523–7533. [Google Scholar] [CrossRef] [PubMed]
  154. Megahed, A.; Gadalla, H.; Filimban, W.A.; Albukhari, T.A.; Sembawa, H.; Bagadood, R.M.; Sindi, G.; Abdelhamid, F.M.; El-Boshy, M.E.; Risha, E.F. Hesperidin ameliorates thioacetamide-induced liver fibrosis via antioxidative and anti-inflammatory mechanisms targeting TGF-β/α-SMA pathways in rats. Int. J. Immunopathol. Pharmacol. 2024, 38, 3946320241309004. [Google Scholar] [CrossRef] [PubMed]
  155. Saponara, I.; Aloisio Caruso, E.; Cofano, M.; De Nunzio, V.; Pinto, G.; Centonze, M.; Notarnicola, M. Anti-Inflammatory and Anti-Fibrotic Effects of a Mixture of Polyphenols Extracted from “Navelina” Orange in Human Hepa-RG and LX-2 Cells Mediated by Cannabinoid Receptor 2. Int. J. Mol. Sci. 2025, 26, 512. [Google Scholar] [CrossRef] [PubMed]
  156. Konishi, Y.; Zhao, Z.; Shimizu, M. Phenolic acids are absorbed from the rat stomach with different absorption rates. J. Agric. Food Chem. 2006, 54, 7539–7543. [Google Scholar] [CrossRef]
  157. Gallego-Lobillo, P.; Lopez-Rodulfo, I.M.; Martinez, M.M. Rat small intestine extract as a source of mammalian α- and β-glycosidases to study polyphenol bioaccessibility and deglycosylation in vitro: A case study with matrix-devoid and matrix-defined apple fractions. Food Res. Int. 2025, 199, 115346. [Google Scholar] [CrossRef]
  158. Vanholme, R.; Cesarino, I.; Rataj, K.; Xiao, Y.; Sundin, L.; Goeminne, G.; Kim, H.; Cross, J.; Morreel, K.; Araujo, P.; et al. Caffeoyl shikimate esterase (CSE) is an enzyme in the lignin biosynthetic pathway in Arabidopsis. Science 2013, 341, 1103–1106. [Google Scholar] [CrossRef]
  159. Barros, J.; Escamilla-Trevino, L.; Song, L.; Rao, X.; Serrani Yarce, J.C.; Palacios, M.D.; Engle, N.; Choudhury, F.K.; Tschaplinski, T.J.; Venables, B.J.; et al. 4-Coumarate 3-hydroxylase in the lignin biosynthesis pathway is a cytosolic ascorbate peroxidase. Nat. Commun. 2019, 10, 1994. [Google Scholar] [CrossRef]
  160. Zielińska, D.; Zieliński, H.; Laparra-Llopis, J.M.; Szawara-Nowak, D.; Honke, J.; Giménez-Bastida, J.A. Caffeic Acid Modulates Processes Associated with Intestinal Inflammation. Nutrients 2021, 13, 554. [Google Scholar] [CrossRef]
  161. Rashedinia, M.; Rasti Arbabi, Z.R.; Sabet, R.; Emami, L.; Poustforoosh, A.; Sabahi, Z. Comparison of Protective Effects of Phenolic Acids on Protein Glycation of BSA Supported by In Vitro and Docking Studies. Biochem. Res. Int. 2023, 2023, 9984618. [Google Scholar] [CrossRef]
  162. Zhao, Y.; Zhao, J.; Ma, H.; Han, Y.; Xu, W.; Wang, J.; Cai, Y.; Jia, X.; Jia, Q.; Yang, Q. High Hepcidin Levels Promote Abnormal Iron Metabolism and Ferroptosis in Chronic Atrophic Gastritis. Biomedicines 2023, 11, 2338. [Google Scholar] [CrossRef]
Figure 1. Proximal tubular epithelial (PTE) cells and mechanisms of dysfunction. Upper and lower cells form the proximal tubular walls, and the middle space is the urinary tract. Closed squares indicate PTE cells. Closed pink circles indicate cell nuclei. Closed red polygon indicates reactive oxygen species (ROS). Closed blue polygons indicate urinary stones. Yellow cells indicate the normal cells. Gray cell indicates a senescent cell. Green cells indicate cells with inflammation.
Figure 1. Proximal tubular epithelial (PTE) cells and mechanisms of dysfunction. Upper and lower cells form the proximal tubular walls, and the middle space is the urinary tract. Closed squares indicate PTE cells. Closed pink circles indicate cell nuclei. Closed red polygon indicates reactive oxygen species (ROS). Closed blue polygons indicate urinary stones. Yellow cells indicate the normal cells. Gray cell indicates a senescent cell. Green cells indicate cells with inflammation.
Ijms 26 09216 g001
Figure 2. Origins of AGEs [6,9,11,12]. Glucose is a hexose, and glyceraldehyde is a triose. Glyceraldehyde, glycolaldehyde, methylglyoxal, glyoxal, and 3-deoxyglucosone can be generated from glucose via enzymatic and non-enzymatic (e.g., oxidation) reactions.
Figure 2. Origins of AGEs [6,9,11,12]. Glucose is a hexose, and glyceraldehyde is a triose. Glyceraldehyde, glycolaldehyde, methylglyoxal, glyoxal, and 3-deoxyglucosone can be generated from glucose via enzymatic and non-enzymatic (e.g., oxidation) reactions.
Ijms 26 09216 g002
Figure 3. Free types of AGEs [6,9,11,12]. (a) AGEs containing one amino acid. CML, Nε-carboxymethyl-lysine; CEL, Nε-carboxyethyl-lysine; MG-H1, methylglyoxal-hydro-imidazolone; GLAP, glyceraldehyde-derived pyridinium; GH-1, glyoxal-hydro-imidazolone. (b) AGEs containing two amino acids. MOLD, methylglyoxal-derived imidazolium cross-link; GODIC, glyoxal-derived imidazolium cross-link; MODIC, methylglyoxal-derived imidazolium cross-link; DODIC, 3-deoxyglucosone-derived imidazolium cross-link.
Figure 3. Free types of AGEs [6,9,11,12]. (a) AGEs containing one amino acid. CML, Nε-carboxymethyl-lysine; CEL, Nε-carboxyethyl-lysine; MG-H1, methylglyoxal-hydro-imidazolone; GLAP, glyceraldehyde-derived pyridinium; GH-1, glyoxal-hydro-imidazolone. (b) AGEs containing two amino acids. MOLD, methylglyoxal-derived imidazolium cross-link; GODIC, glyoxal-derived imidazolium cross-link; MODIC, methylglyoxal-derived imidazolium cross-link; DODIC, 3-deoxyglucosone-derived imidazolium cross-link.
Ijms 26 09216 g003
Figure 4. Crude AGE patterns [6,11,12]. MG-H1, argpyrimidine, and GLAP can be generated from glyceraldehyde, though MG-H1 and argpyrimidine can also be generated from methylglyoxal. Black arrow shows a step in enzymatic/non-enzymatic reactions. MG-H1, methylglyoxal-hydro-imidazolone; GLAP, glyceraldehyde-derived pyridinium.
Figure 4. Crude AGE patterns [6,11,12]. MG-H1, argpyrimidine, and GLAP can be generated from glyceraldehyde, though MG-H1 and argpyrimidine can also be generated from methylglyoxal. Black arrow shows a step in enzymatic/non-enzymatic reactions. MG-H1, methylglyoxal-hydro-imidazolone; GLAP, glyceraldehyde-derived pyridinium.
Ijms 26 09216 g004
Figure 5. Type 1 and 2 diverse AGE patterns [11,12,88]. Closed blue circles indicate amino acids. Black number indicates the number of amino acid residues. (a) Type 1 diverse AGE pattern. Left and middle, middle and right, and left and right protein A structures indicate type 1A, 1B, and 1C diverse AGE patterns. MG-H1, methylglyoxal-hydro-imidazolone; Arg-P, argpyrimidine. (b) Type 2 diverse AGE pattern. CML; Nε-carboxymethyl-lysine.
Figure 5. Type 1 and 2 diverse AGE patterns [11,12,88]. Closed blue circles indicate amino acids. Black number indicates the number of amino acid residues. (a) Type 1 diverse AGE pattern. Left and middle, middle and right, and left and right protein A structures indicate type 1A, 1B, and 1C diverse AGE patterns. MG-H1, methylglyoxal-hydro-imidazolone; Arg-P, argpyrimidine. (b) Type 2 diverse AGE pattern. CML; Nε-carboxymethyl-lysine.
Ijms 26 09216 g005
Figure 6. Type 1 and 2 multiple AGE patterns. Closed blue circles indicate amino acids. Black or red number indicates the number of amino acid residues [6,11,12,87,88]. (a) Type 1 multiple AGE pattern. CEL, Nε-carboxyethyl-lysine; Arg-P, argpyrimidine; CML, Nε-carboxymethyl-lysine; MG-H1, methylglyoxal-hydro-imidazolone. (b) Type 2 multiple AGE pattern. Proteins Y and Z are linked with D1—free AGE—involving two amino acids (the 5th amino acid residue in protein Y and the 6th amino acid residue in protein Z).
Figure 6. Type 1 and 2 multiple AGE patterns. Closed blue circles indicate amino acids. Black or red number indicates the number of amino acid residues [6,11,12,87,88]. (a) Type 1 multiple AGE pattern. CEL, Nε-carboxyethyl-lysine; Arg-P, argpyrimidine; CML, Nε-carboxymethyl-lysine; MG-H1, methylglyoxal-hydro-imidazolone. (b) Type 2 multiple AGE pattern. Proteins Y and Z are linked with D1—free AGE—involving two amino acids (the 5th amino acid residue in protein Y and the 6th amino acid residue in protein Z).
Ijms 26 09216 g006
Figure 7. Traditional Japanese medical science (TJMS) comprises Kampo, Japanese folk medical science (JFMS), and traditional Chinese medical science (TCMS).
Figure 7. Traditional Japanese medical science (TJMS) comprises Kampo, Japanese folk medical science (JFMS), and traditional Chinese medical science (TCMS).
Ijms 26 09216 g007
Figure 8. Chemical structures of study molecules. (a) Quercetin, hesperidin, and p-coumaric acid. (b) The structure of the flavonoid skeleton. Numbers indicate the location of the functional group (e.g., hydroxyl, carbonyl).
Figure 8. Chemical structures of study molecules. (a) Quercetin, hesperidin, and p-coumaric acid. (b) The structure of the flavonoid skeleton. Numbers indicate the location of the functional group (e.g., hydroxyl, carbonyl).
Ijms 26 09216 g008
Figure 9. Metabolites of quercetin in the blood [129,130,131]. Q3G, quercetin-3-O-glucoside; Q3GA, quercetin 3-O-glucuronide; 3′-O-methyl-Q3GA, 3′-O-methylquercetin-O-glucuronide; Q4′G, quercetin-4′-O-glucoside; Q4′GA, quercetin-4′-O-glucuronide; 3′-O-methyl-Q4′GA, 3′-O-methylquercetin-4-O-glucuronide; Q3′GA, quercetin-3′-glucuronide; Q3′S, quercetin-3′-O-sulfate.
Figure 9. Metabolites of quercetin in the blood [129,130,131]. Q3G, quercetin-3-O-glucoside; Q3GA, quercetin 3-O-glucuronide; 3′-O-methyl-Q3GA, 3′-O-methylquercetin-O-glucuronide; Q4′G, quercetin-4′-O-glucoside; Q4′GA, quercetin-4′-O-glucuronide; 3′-O-methyl-Q4′GA, 3′-O-methylquercetin-4-O-glucuronide; Q3′GA, quercetin-3′-glucuronide; Q3′S, quercetin-3′-O-sulfate.
Ijms 26 09216 g009
Figure 10. Model of Q3G, Q4′G, and quercetin absorption into the small intestine, and leaking of glucuronide-quercetin and quercetin into the blood or lymph vessels. Closed blue square indicates small-intestinal epithelial cells. Open pink area indicates blood or lymph vessel. Closed yellow square indicates sodium-glucose cotransporter 1 (SGLT1). Closed pink hexagon indicates lactase phlorizin hydrolase (LPH). Closed green square indicates cytosolic β-glucosidase. Closed red square indicates phase II enzyme system. Q3G, quercetin-3-O-glucoside; Q4′GA, quercetin-4′-O-glucuronide.
Figure 10. Model of Q3G, Q4′G, and quercetin absorption into the small intestine, and leaking of glucuronide-quercetin and quercetin into the blood or lymph vessels. Closed blue square indicates small-intestinal epithelial cells. Open pink area indicates blood or lymph vessel. Closed yellow square indicates sodium-glucose cotransporter 1 (SGLT1). Closed pink hexagon indicates lactase phlorizin hydrolase (LPH). Closed green square indicates cytosolic β-glucosidase. Closed red square indicates phase II enzyme system. Q3G, quercetin-3-O-glucoside; Q4′GA, quercetin-4′-O-glucuronide.
Ijms 26 09216 g010
Figure 11. Model of Q3G, Q4′G, and quercetin absorption into the large intestine, and leaking of quercetin-glucuronide and quercetin into the blood or lymph vessels [143,144,145,146,147]. Closed blue square indicates large-intestinal epithelial cells. Open pink area indicates blood or lymph vessel. Closed orange square indicates β-glucosidase of internal bacteria. Closed yellow square indicates sodium-glucose cotransporter 1 (SGLT1). Closed green square indicates cytosolic β-glucosidase. Closed red square indicates phase II enzyme system. Q3G, quercetin-3-O-glucoside; Q4′GA, quercetin-4′-O-glucuronide.
Figure 11. Model of Q3G, Q4′G, and quercetin absorption into the large intestine, and leaking of quercetin-glucuronide and quercetin into the blood or lymph vessels [143,144,145,146,147]. Closed blue square indicates large-intestinal epithelial cells. Open pink area indicates blood or lymph vessel. Closed orange square indicates β-glucosidase of internal bacteria. Closed yellow square indicates sodium-glucose cotransporter 1 (SGLT1). Closed green square indicates cytosolic β-glucosidase. Closed red square indicates phase II enzyme system. Q3G, quercetin-3-O-glucoside; Q4′GA, quercetin-4′-O-glucuronide.
Ijms 26 09216 g011
Figure 12. Metabolites detected in HepG2 cells treated with quercetin [147]. Q7GA, quercetin-7-O-glucuronide.
Figure 12. Metabolites detected in HepG2 cells treated with quercetin [147]. Q7GA, quercetin-7-O-glucuronide.
Ijms 26 09216 g012
Figure 13. Model of the absorption of hesperidin and hesperetin in the small/large intestine, and leaking of hesperidin-glucuronide and hesperetin into the blood or lymph vessels [148,149,150,151]. Closed blue square indicates small/large intestine epithelial cells. Open pink area indicates blood or lymph vessel. Closed orange square indicates β-glucosidase of intestinal bacteria. Closed yellow circle indicates α-glucosidase. Closed red trapezoid indicates UDP-UGT/SULT. UGT, glucuronosyltransferase; SULT, sulfotransferase. Closed red square indicates phase II enzyme system.
Figure 13. Model of the absorption of hesperidin and hesperetin in the small/large intestine, and leaking of hesperidin-glucuronide and hesperetin into the blood or lymph vessels [148,149,150,151]. Closed blue square indicates small/large intestine epithelial cells. Open pink area indicates blood or lymph vessel. Closed orange square indicates β-glucosidase of intestinal bacteria. Closed yellow circle indicates α-glucosidase. Closed red trapezoid indicates UDP-UGT/SULT. UGT, glucuronosyltransferase; SULT, sulfotransferase. Closed red square indicates phase II enzyme system.
Ijms 26 09216 g013
Figure 14. Metabolites of hesperidin detected in cytosolic fractions of small intestine, large intestine, and hepatic tissue [151,152].
Figure 14. Metabolites of hesperidin detected in cytosolic fractions of small intestine, large intestine, and hepatic tissue [151,152].
Ijms 26 09216 g014
Figure 15. Metabolic pathway of p-coumaric acid in plants [158,159]. 4CL, 4-coumaric CoA-ligase; HCT, hydroxycinnamoyl transferase; C3′H, p-coumaroyl-shikimic acid-3-hydroxytransferase; CSE, caffeoyl shikimic esterase.
Figure 15. Metabolic pathway of p-coumaric acid in plants [158,159]. 4CL, 4-coumaric CoA-ligase; HCT, hydroxycinnamoyl transferase; C3′H, p-coumaroyl-shikimic acid-3-hydroxytransferase; CSE, caffeoyl shikimic esterase.
Ijms 26 09216 g015
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.; Yamada, S.; Han, J.; Yang, Q.; Guo, X.; Nakahashi, T.; Mizuta, S.; Inoue, S.; et al. Potential of Orally Administered Quercetin, Hesperidin, and p-Coumaric Acid in Suppressing Intra-/Extracellular Advanced Glycation End-Product-Induced Cytotoxicity in Proximal Tubular Epithelial Cells. Int. J. Mol. Sci. 2025, 26, 9216. https://doi.org/10.3390/ijms26189216

AMA Style

Takata T, Moriya J, Miyazawa K, Yamada S, Han J, Yang Q, Guo X, Nakahashi T, Mizuta S, Inoue S, et al. Potential of Orally Administered Quercetin, Hesperidin, and p-Coumaric Acid in Suppressing Intra-/Extracellular Advanced Glycation End-Product-Induced Cytotoxicity in Proximal Tubular Epithelial Cells. International Journal of Molecular Sciences. 2025; 26(18):9216. https://doi.org/10.3390/ijms26189216

Chicago/Turabian Style

Takata, Takanobu, Junji Moriya, Katsuhito Miyazawa, Sohsuke Yamada, Jia Han, Qian Yang, Xin Guo, Takeshi Nakahashi, Shuichi Mizuta, Shinya Inoue, and et al. 2025. "Potential of Orally Administered Quercetin, Hesperidin, and p-Coumaric Acid in Suppressing Intra-/Extracellular Advanced Glycation End-Product-Induced Cytotoxicity in Proximal Tubular Epithelial Cells" International Journal of Molecular Sciences 26, no. 18: 9216. https://doi.org/10.3390/ijms26189216

APA Style

Takata, T., Moriya, J., Miyazawa, K., Yamada, S., Han, J., Yang, Q., Guo, X., Nakahashi, T., Mizuta, S., Inoue, S., Masauji, T., & Motoo, Y. (2025). Potential of Orally Administered Quercetin, Hesperidin, and p-Coumaric Acid in Suppressing Intra-/Extracellular Advanced Glycation End-Product-Induced Cytotoxicity in Proximal Tubular Epithelial Cells. International Journal of Molecular Sciences, 26(18), 9216. https://doi.org/10.3390/ijms26189216

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