Next Article in Journal
Effects of a Carbohydrate Meal on Lipolysis
Next Article in Special Issue
Effect of Whole Blood Dietary Mineral Concentrations on Erythrocytes: Selenium, Manganese, and Chromium: NHANES Data
Previous Article in Journal
Subacute Effects of Moderate-Intensity Aerobic Exercise in the Fasted State on Cell Metabolism and Signaling in Sedentary Rats
Previous Article in Special Issue
Effect of Selol on Tumor Morphology and Biochemical Parameters Associated with Oxidative Stress in a Prostate Tumor-Bearing Mice Model
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Renal Health Through Medicine–Food Homology: A Comprehensive Review of Botanical Micronutrients and Their Mechanisms

by
Yi Zhao
1,
Jian-Ye Song
1,
Ru Feng
1,
Jia-Chun Hu
1,
Hui Xu
1,
Meng-Liang Ye
1,
Jian-Dong Jiang
1,
Li-Meng Chen
2,* and
Yan Wang
1,*
1
State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
2
Department of Nephrology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
*
Authors to whom correspondence should be addressed.
Nutrients 2024, 16(20), 3530; https://doi.org/10.3390/nu16203530
Submission received: 30 August 2024 / Revised: 18 September 2024 / Accepted: 14 October 2024 / Published: 18 October 2024
(This article belongs to the Special Issue Effects of Selenium and Other Micronutrient Intake on Human Health)

Abstract

:
Background: As an ancient concept and practice, “food as medicine” or “medicine–food homology” is receiving more and more attention these days. It is a tradition in many regions to intake medicinal herbal food for potential health benefits to various organs and systems including the kidney. Kidney diseases usually lack targeted therapy and face irreversible loss of function, leading to dialysis dependence. As the most important organ for endogenous metabolite and exogenous nutrient excretion, the status of the kidney could be closely related to daily diet. Therefore, medicinal herbal food rich in antioxidative, anti-inflammation micronutrients are ideal supplements for kidney protection. Recent studies have also discovered its impact on the “gut–kidney” axis. Methods: Here, we review and highlight the kidney-protective effects of botanicals with medicine–food homology including the most frequently used Astragalus membranaceus and Angelica sinensis (Oliv.) Diels, concerning their micronutrients and mechanism, offering a basis and perspective for utilizing and exploring the key substances in medicinal herbal food to protect the kidney. Results: The index for medicine–food homology in China contains mostly botanicals while many of them are also consumed by people in other regions. Micronutrients including flavonoids, polysaccharides and others present powerful activities towards renal diseases. Conclusions: Botanicals with medicine–food homology are widely speeded over multiple regions and incorporating these natural compounds into dietary habits or as supplements shows promising future for renal health.

1. Introduction

The kidneys are indispensable for human physiological functions, including the excretion of drugs and nutrients. The kidneys maintain the balance of water, electrolytes, and endogenous metabolites throughout the body through the processes of glomerular filtration, tubular reabsorption, and tubular secretion. Additionally, they serve as the predominant organ responsible for the excretion of exogenous substances, including drugs and food nutrients [1]. However, this feature also renders the kidneys highly susceptible to acute or chronic injuries caused by these exogenous substances [2]. For example, drug-induced nephrotoxicity (DIN) accounts for up to 60% of cases of acute kidney injury (AKI) and the correlation increases in elder patients [1,3]. Dietary-resourced substances could also lead to abnormal renal status, for instance, oxalate and oxalate precursors as key determinants of urinary oxalate excretion and stone disease. In addition to certain substances that induce injury, the kidney is also affected by metabolic disorders (e.g., diabetic kidney disease) and immune abnormalities (e.g., membranous glomerulonephritis and immunoglobulin A nephropathy). The underlying molecular mechanism of kidney disorders is a complex phenomenon involving oxidative stress, inflammation, fibrosis, and mitochondrial dysfunction. Recently, the vital role of the “gut–kidney” axis, which is closely related to daily diet, has been identified and investigated [4]. However, contemporary renal disease therapy is predominantly supportive, comprising diuretics, renin–angiotensin–aldosterone system inhibitors, and immunosuppressants with a paucity of targeted pharmaceutical agents [2]. Meanwhile, these treatments could hardly reverse the continuous decline in renal function and the formation of end-stage renal disease (ESRD) [5]. Compared to drug treatment, certain daily diet patterns may also have an impact on renal function [6,7]. Natural food plants are extremely wealthy in nutrients and biological active compounds worth exploring and utilizing. Therefore, there is a promising future to explore botanicals with medicine–food homology to alleviate or prevent kidney disease.
In fact, the concept of food as medicine, or “Yao Shi Tong Yuan” in Chinese, is a fundamental tenet of Traditional Chinese Medicine (TCM). It underscores the dual function of specific foods in offering both nutritional and therapeutic advantages. The use of herbal food for medical purposes has a long and illustrious history in numerous other regions, including Japan, Korea, South America, and Europe. Despite the complexity of its ingredients, preparation methods, and active components, medicinal herbal food has been shown to have significant effects on a number of disorders, including diabetes mellitus, allergic diseases, pancreatitis, depression, and renal disease [8,9,10,11]. In the context of renal disease, a considerable number of botanical foods have been observed to exert a protective effect, as evidenced by a reduction in serum creatinine, blood urea nitrogen, histopathological injury, and fibrosis degree [12,13,14]. Early in 2002 [15], the National Health Commission (NHC) of the People‘s Republic of China had published the list of natural medical food including mostly different kinds of herbal ones. Being up to date, the list has expanded to 102 kinds of natural food including Astragalus membranaceus, Codonopsis tangshen Oliv., and Poria cocos (Schw.) Wolf. These herbal foods are frequently consumed by Chinese people either for their flavors or their potential health benefits. Many of them had also demonstrated renal-protective effects through anti-inflammation [16], antioxidation [17,18], and regulating mitochondria [19] and gut microbiota [20] with various micronutrients in them. The Food and Drug Administration (FDA) of the US have also established guidelines for the safety and efficacy of dietary supplements for medical use [21]. The 2020–2030 strategic plan for nutrition research by National Institutes of Health (NIH) had even placed “food as medicine” as one of the strategic goals [22], indicating the potential of medicine–food homology.
Here, we reviewed the frequently used medicinal herbal food in China, the US, and Europe, and summarized the potential renal-protective effects and mechanism of these medicinal herbal foods as shown in Figure 1, providing a better basis for utilizing and exploring the key substances in botanicals with medicine–food homology.

2. Botanical Ingredients with Medicine–Food Homology in China, the US, and Europe

The use of botanical ingredients for dietary supplements or even for food is a widespread tradition around the world. In addition to China, the popularity of botanical dietary supplements is also evident in various European countries. Dietary supplements represent 15–20% of the total botanical market [23]. However, the complex compounds present in botanical ingredients also pose a potential risk. One of the most illustrative examples of a botanical-induced health risk is the aristolochic acid nephropathy (AAN) incident that occurred in the early 1990s [24]. The ingestion of aristolochic acids present in certain herbs had resulted in the development of AAN in numerous patients, leading to chronic renal failure and ESRD in Belgium, China, and Korea [24,25,26]. Considering the widespread and covert use of herbs, some researchers estimated that the AAN was still underestimated and called for a stronger and systematic supervision system of herbal medicine or ingredients [25]. To ensure the safety and efficiency of botanical ingredients and supplements, many regulations and standards have been published to restrict the addition of botanical ingredients in dietary supplements. In 2002, the NHC of China had published the first index of ingredients that could be both food and medication [15]. The initial version contained 92 unique edible parts including mostly botanical ingredients. Many of them have long been thought to have kidney-protective effects like Astragalus membranaceus and Lycium barbarum L. [16,27,28]. In recent years, there has been a growing interest in botanical dietary supplements, leading the NHC to edit the index in 2019 [29] and 2023 [30]. This resulted in the addition of 15 new botanical ingredients. In 2021, the NHC had also published related regulations for this index including the criteria for botanical ingredients as both food and medication [31]. The fundamental criteria consist of the following requirements: (i) have been traditionally consumed as food; (ii) have been listed in the Pharmacopoeia of the PRC; (iii) no food safety problems have been found in the safety assessment; (iv) comply with relevant laws and regulations on the protection of Chinese herbal medicines, wild animals, plants, and ecological resources. For the first rule, it also requires evidence for the ingredients to have been consumed as food for more than 30 years. Concurrently, all of these ingredients are also included in the Pharmacopoeia of the People’s Republic of China, which demonstrates their efficacy as medicinal agents. In addition to botanicals that are regarded as both food and medicine, the NHC had also delineated a list of medicinal herbs that may be included in dietary supplements (Table S1) [15]. With regard to the European countries, the regulation of botanical ingredients is also divided into two distinct categories: medicine and food supplements. The European Medicines Agency (EMA) bears the responsibility of assessing the safety and efficacy of herbal preparations when utilized as medicines, while the European Food Safety Authority (EFSA) is tasked with the oversight of herbal preparations employed as food supplements. The evaluation of botanicals used in food products was initiated in 2004, when the EFSA mandated its Scientific Committee to develop a science-based toolkit for the safety assessment of botanicals and botanical preparations [32]. Subsequently, in 2009, the EFSA published the inaugural edition of the Compendium of Botanicals, which provided information regarding the safety risks associated with botanical ingredients in dietary supplements [32]. In the United States, the Dietary Supplement Health and Education Act (DSHEA) of 1994 initially delineated the status of herbs in dietary supplements, alongside that of vitamins, minerals, amino acids, and other substances [33]. While there is no comprehensive list of ingredients used in dietary supplements by the FDA, the NIH has established a Dietary Supplement Label Database (DSLD) [34] that includes as many of these supplements as possible.
Table 1 shows a full list of botanicals in the China index for botanicals with medicine–food homology. It contains a total of 91 botanicals, the majority of which are also included in dietary supplements in the United States and the European Union, as indicated by the EFSA Compendium of Botanicals and the DSLD. However, only a limited number of the listed botanicals are regarded as herbal medicines in accordance with the EMA standard. This is a markedly different situation from that in China, where all the botanicals in Table 1 are also included in the country’s Pharmacopoeia. Additionally, botanicals such as clove, ginkgo, and ginger are present in all four lists or databases, which suggests their widespread popularity and efficacy as both food and medicine.

3. Nutrients from These Botanical Food Ingredients with Kidney-Protective Effects

3.1. Flavonoids

Flavonoids are polyphenolic compounds that are widely distributed in a variety of botanical foods. The basic chemistry structure of flavonoids consists of two benzene rings with a phenolic hydroxyl group and a heterocyclic ring forming a C6-C3-C6 basic carbon framework [35]. The structure can be further categorized into flavanones, flavones, flavonols, flavanols, dihydroflavonols, and anthocyanins based on different functional groups and their positions [36]. It is one of the most bioactive and common compounds in food resources, especially botanicals. As demonstrated in Table 2, flavonoids that have been validated to have kidney-protective effects include quercetin [37,38,39], kaempferol [40], myricetin [41,42], isorhamnetin [43,44], fisetin [45,46], icariin [47,48], apigenin [49,50], baicalein [51,52], baicalin [53,54], nobiletin [55], vitexin [56,57], hesperidin [58,59], and hesperetin [60,61]. Quercetin and kaempferol appear in the majority of vegetables and fruits, which accounts for their status as the most prevalent dietary supplement ingredients. A review of the DSLD revealed that dietary supplements containing quercetin constituted 2.3% of the total products [29]. As illustrated in Table 2, citrus botanicals such as Citrus medica L. var. sarcodactylis Swingle and Citrus reticulata Blanco exhibit a notable content of flavonoids, including hesperidin, hesperetin, naringin, quercetin, and kaempferol [62,63,64]. The flavonoids present in citrus fruits have been demonstrated to possess potent antioxidant properties, making them a promising avenue of research for the development of novel therapeutic agents for the treatment of diabetes, neurodegenerative disorders, and kidney diseases such as AKI and diabetic nephropathy (DN) [65,66,67,68,69]. In addition to citrus, Glycine max (L.) Merr. (soybean) in Table 1 contains a unique kind of isoflavones called soy isoflavones, or “phytoestrogens” due to their structural similarity with 17-β-estradiol [70]. Though most of the studies of soy isoflavones fell into their endocrine regulation impacts, one recent study had discovered that the intake of soy isoflavones exhibited favorable effects on renal function and kidney morphology [71]. Puerarin, derived from Pueraria lobata (Willd.) Ohwi, is also an isoflavone. Recent studies have demonstrated its influence on the toll-like receptor (TLR) 4/MyD88 pathway and the M1 macrophage differential [72]. Later, flavonoids in Pueraria thomsonii Benth. were further found to decrease inflammation in the kidney by regulating the level of Clostridium in the gut [73]. Hesperidin from citrus botanicals could also alter gut microbiota against Zn-induced nephrotoxicity [58], indicating the potential of the gut–kidney axis.

3.2. Polysaccharides

Natural polysaccharides are a class of complex macromolecules that are formed from a variety of monosaccharide units and diverse glycosidic linkages. The monosaccharide units can be classified into several categories, including hexose, which includes glucose, galactose, and mannose; N-acetyl-hexose (HexNac), which encompasses GalNac and GluNac; pentose (e.g., xylose and arabinose); deoxyhexose (e.g., fucose); and numerous terminal modifications, such as phosphorylation, sulfation, and sialylation. Up to the present date, the identification and isolation still remain a critical challenge. However, the polysaccharides were found to be the principal compounds in some of the botanicals in Table 1 and the pharmacological effects of polysaccharides have also long been confirmed. Table 2 illustrates the representative polysaccharides from botanicals in Table 1 and Table S1, along with their impact on kidney diseases.
Laminaria japonica Aresch. is a widespread food ingredient in China with a significant amount of polysaccharides. It contains polysaccharides weighing from 5 kDa to 104 kDa [231] with great potential in anti-fibrosis effects, which revealed its capacity to treat CKD [106]. In many kidney diseases including the DN model [104,105], DIN model [103,106], and IRI model [102], polysaccharides from Laminaria japonica Aresch. showed strong impact on fibrosis- and apoptosis-related molecules like TGF-β, α-SMA, and Bax/Bcl-2. Polysaccharides are also identified to be the primary compounds in Polygonatum sibiricum Red [232]. Polygonat Rhizomai polysaccharides were discovered to be protective against uranium-induced AKI with 141 kDa polysaccharides in them [17]. The polysaccharides from other traditional kidney-protective botanical food like Lycium barbarum L. [101] and Astragalus membranaceus [19,96,97] showed protective effects against DIN as well.
In consideration of the gut–kidney axis, polysaccharides derived from these botanical ingredients also play a significant role in the intricate interplay of gut–kidney crosstalk. The polysaccharides derived from Phyllostachys nigra [20], Paeonia × suffruticosa (Moutan Cortex) [111], and Astragalus membranaceus (Fisch.) Bge. [97] have been observed to possess the capacity to regulate the gut–kidney axis, thereby conferring benefit to the kidney. The mechanisms involved in remodeling the gut microbiota composition include an increase in the abundance of probiotics, such as Lactobacillales, and an upregulation of SCFAs.

3.3. Terpenoids

Terpenoids, which are derived from isoprene, encompass a diverse range of chemical compounds, including monoterpenoids, sesquiterpenoids, diterpenoids, sesterterpenes, triterpenoids, and polyterpenoids [233]. Considering the ingredients in Table 1 and Table S1, Panax ginseng C. A. Mey. (ginseng), Glycyrrhiza uralensis Fisch. (Liquorice), Alisma orientatle (Sam.) Juzep., and Ganoderma lucidum (Leyss. ex Fr.) Karst. (Ganoderma) are representative for their great amount and variety of terpenoids.
Ganoderma triterpenoids isolated from Ganoderma are some of the major chemical constituents in Ganoderma and could be further divided into C30 (ganoderic acid, ganodermanontriol, applanoxidic acids, and their derivatives), C27 (lucidenic acid, ganolactone, and their derivatives), and C24 (lucidones and their derivatives), and others according to their skeleton carbons [234]. Among the compounds identified, ganoderic acids had been proven to possess protective properties in the context of ADPKD [159], renal fibrosis [160], and IRI [161]. Licorice is another ingredient that has been demonstrated to possess renal-protective terpenoids, the most prominent of which are glycyrrhizic acid and glycyrrhetinic acid [235]. Glycyrrhizic acid possesses surprising protective effects in DN [162,163] and DIN including tacrolimus [164], contrasts [165], and triptolide [166]-induced kidney injury. Similar effects were also found in glycyrrhetinic acid in DN [162] and IRI [167]. Alisma orientatle (Sam.) Juzep. and Panax ginseng C. A. Mey. are two botanicals in Table S1, which means that they are not considered daily food but could be added to dietary supplements. However, they have been traditionally used to treat renal disorders in TCM, especially Alisma orientatle (Sam.) Juzep. Alisols and their derivatives are tetracyclic triterpene alcohols isolated from Alisma orientatle (Sam.) Juzep. and play a significant role in its pharmacological effects [236]. Recent studies had demonstrated the potential of alisols in protecting the kidney from DPP-induced injury and IRI by targeting the farnesoid X receptor and soluble epoxide hydrolase [148,149]. Additionally, alisol B 23-acetate was observed to regulate the gut microbiota, thereby improving renal function in CKD mice [150]. Ginsenosides, the representative compounds in ginseng, are a big family of triterpenoid saponins with a four-ring rigid steroid skeleton [237]. Early in 1998, a study revealed that ginsenoside Rd could protect against IRI by affecting proximal tubule cells [238]. Later, numerous studies had demonstrated that ginsenosides could alleviate AKI [151,152], renal carcinoma [153,154], DN [155,156], and renal fibrosis [157,158].
Iridoids constitute a large and distinctive class of monoterpenoids that are ubiquitous in botanical sources and have been demonstrated to exhibit notable bioactivity [239]. Iridoids are receiving increasing attention for their great pharmacological effects in the liver, kidney, and nervous disorders [240,241,242,243]. Catalpol, one of the most common iridoids, for example, showed strong antioxidation and anti-inflammation effects in DN-, CKD-, and DIN-induced AKI models [115,116,117,118,119,120,121]. Cornus officinalis Sieb. et Zucc. was found to be abundant in iridoids [244], in which loganin and morroniside were shown to be protective for the kidney [122,123,124,125,133]. The representative terpenoids including triterpenoids and iridoids from botanicals with medicine–food homology are also shown in Table 2.

3.4. Alkaloids

Isolated from natural herbs, alkaloids are a diverse group of naturally occurring organic compounds that primarily contain basic nitrogen atoms [245]. Structurally, they could be characterized by a heterocyclic ring that incorporates nitrogen, and they often possess complex and varied molecular frameworks, for example, isoquinoline, pyrrolidines, pyridines, indole, and others. Dietary botanicals also contain variable bioactive alkaloids for renal protection. Table 2 provides a list of the principal representative alkaloids with kidney-protective effects derived from the botanicals included in Table 1 and Table S1.
Tetramethylpyrazine, the characteristic alkaloid of Ligusticum sinense Chuanxiong, has been clinically utilized to inhibit platelet aggregation and reduce blood viscosity [246]. Its renal-protective effects were also validated in multiple models including DIN-induced AKI [168,169], IRI [170], and DN [171,172] with classical mechanisms targeting inflammation and oxidation markers. Leonurine from Leonurus japonicus Houtt. also showed protective effects in lipopolysaccharide-, DDP-, and vancomycin-induced AKI [173,174,175,176,177]. Besides AKI, trigonelline, the characteristic alkaloid in Trigonella foenum-graecum L. (Fenugreek) and Coffea arabica (coffee) was demonstrated to maintain a strong protective effect on oxalate nephropathy (ON) [191,192], which is closely related to diet oxalate. Nelumbo nucifera Gaertn. is well known for its abundant alkaloids, especially in its seeds. Liensinine, isoliensinine, neferine, and nuciferine are all alkaloids with kidney-protective effects isolated from Nelumbo nucifera Gaertn. Besides AKI and DN, neferine was also revealed to alleviate hyperuricemic nephropathy by targeting the inflammasome pathway [208]. Another alkaloid that has been widely studied for its protective effects on the kidneys is berberine. This isoquinoline alkaloid was initially isolated from Hydrastis canadensis and is primarily found in Coptis chinensis. Although these botanicals are not typically included in dietary regimens, berberine itself is a frequently utilized compound in dietary supplements. In DSLD, dietary supplements containing berberine reached 865 records. The renal-protective effects and mechanism of berberine have long been studied since 2011 in the DN model [247,248]. Later, the extraordinary effects of berberine were found in multiple nephropathy models including DIN-induced AKI [178,179,180,181,182], IRI-induced AKI [183,184,185], DN [186,187,188], and UUO-induced renal fibrosis [189]. The mechanisms involved in the treatment of berberine contained traditional anti-inflammation, oxidation, apoptosis, and also the regulation of gut bacteria. With the increasing interest in the gut microbiota, the interaction between the gut microbiota and berberine in the treatment of renal diseases has shown great promise. Recent studies demonstrated that berberine could improve chronic kidney disease by inhibiting the production of enterogenous toxins such as trimethylamine oxide (TMAO) and p-cresol (pCS), which are metabolites from gut microbiota [190]. Berberine could also increase the abundance of Coprococcus, Bacteroides, Akkermansia, and Prevotella to alleviate UAN [249,250,251]. A growing body of evidence suggests that the mechanisms of renoprotective effects through the gut–renal axis deserve further investigation.
Though many alkaloids possess renal-protective effects, a few studies have yielded contradictory results for the same compounds, for example, matrine from Sophora japonica L. Some studies [252,253] found that matrine could lead to kidney injury with mitochondria dysfunction and oxidation while another study demonstrated its protective effect on the DDP-induced AKI model [198]. Its derivate, oxymatrine, was also found to alleviate gentamicin-induced AKI [199], IRI [200], and renal fibrosis [254]. It again reinforces the necessity for the scientific research and regulation of the dietary utilization of botanicals with medicine–food homology.

3.5. Others

In addition to the aforementioned classes of compounds, botanicals with medicine–food homology also contain many other abundant bioactive nutrients including various kinds of polyphenols and anthraquinones, among which some showed great potential in benefiting the kidney as shown in Table 1. Simple phenylpropanoids are a kind of natural polyphenols with a three-carbon side chain linked to a phenyl ring, forming a C6-C3 skeleton. The primary structure also makes them precursors to many other natural compounds, such as flavonoids, lignans, and polyphenols [255]. Simple phenylpropanoids, including the widely distributed ferulic acid and chlorogenic acid, were both found to remodel the composition of the gut microbiota, thereby alleviating UAN [227,228,229]. Curcumin, the most bioactive and abundant polyphenol [256] in Curcuma longa L., has been extensively investigated for its potent antioxidant effects [213]. Together with its derivatives, curcumin displayed potential in treating LN [212], DIN-induced AKI [211], DN [215], and even focal segmental glomerulosclerosis (FSGS) [214]. Additionally, research has indicated that curcumin may mitigate renal impairment by modulating the microbiota in conditions such as CKD and UAN [257,258]. Sesamin, a lignan from dietary sesame, was also demonstrated to decrease uremic toxins by inhibiting the gut microbiota indole pathway [223], indicating importance of the gut–kidney axis. Anthraquinone compounds, for example, emodin and its derivatives, are the most important chemical compounds in Rheum palmatum L., which has long been used in TCM and added to dietary supplements [259]. The anthraquinone compounds in Rheum palmatum L. also possess renal-protective effects on DN, CKD, and UUO models, mainly targeting TGF-β secretion [216,217,218,219,220]. Interestingly, emodin was found to decrease uremic toxins as well by targeting gut microbiota [260]. An increasing body of evidence is emerging that demonstrates the significance of the gut–kidney axis in relation to dietary nutrients.

4. Mechanisms Involved in the Kidney-Protective Effects of Botanical Ingredients with Medicine–Food Homology

4.1. Antioxidation, Anti-Inflammation, and Anti-Fibrosis

Previous studies had focused on the inflammation/oxidation-oriented mechanisms, which are key pathological changes in kidney diseases. Oxidative stress is characterized by an initial injury in the kidney due to the activities of intra- and extracellular oxygen-derived radicals and the resultant inflammatory response [261]. When the kidney is exposed to harmful stimuli, ROS such as superoxides and hydroxyl radicals are produced in excess and interact with the molecular components and functions of a nephron such as the cell membrane, DNA methylation, histone modifications, and micro-RNAs, which are crucial mechanisms for fetal programming [262]. The production and scavenging of oxygen-derived radicals are in a dynamic balance through enzymatic (SOD, CAT, HO-1) and non-enzymatic (vitamin C, vitamin E, glutathione) antioxidant systems [263]. To deal with increased oxidative stress, several redox signaling response cascades, including Nrf2, could be activated to regulate this antioxidative gene expression [263]. Due to the existence of the oxygen shunt diffusion, the kidney is highly susceptible to oxidative stress and hypoxia [264]. Abnormalities in oxidative stress have been observed in a range of kidney diseases, including CKD, DN, and DIN-induced AKI and CKD [265,266,267,268].
The critical role of inflammation in the development of AKI, CKD, and DN was also widely confirmed in animal and clinical studies. The inflammation in the kidney is activated by various mechanisms including endothelial injury, drugs, and infection and causes glomerular, interstitial, and vascular damage [269]. The process of neutrophil infiltration is accompanied by an increase in multiple pro-inflammatory chemotaxes, adhesion factors, and chemokines, including ICAM-1, P- and E-selectin, and IL-1β, IL-6, IL-18, and TNF-α. This process is strongly correlated with fibrosis, autophagy, oxidative stress, and mitochondrial dysfunction [270]. In primary glomerular disease including acute glomerulonephritis, IgA nephropathy, and nephrotic syndrome, complement activation and immune complex deposition, which elicit strong inflammation, were also identified as key pathogenesis [271,272]. Pro-inflammatory signaling pathways include the TLR4/MyD88 signaling pathway, which is also closely related to gut microbiota due to its recognition of pathogen-associated molecular patterns [273]. The activation of TLR4 could lead to the upregulation of another key pro-inflammatory signal, the NF-κB-related pathway [273], which is also the target for most of the anti-inflammation effects in Table 2.
Fibrosis represents another pivotal pathological process following inflammation, especially chronic inflammation. With continuous inflammatory stimulation, various stromal cells in the kidney are transformed to myofibroblasts, which contribute to excessive extracellular matrix production and deposition in the renal parenchyma, eventually leading to loss of renal function [274]. The increase in the secretion of TGF-β and the fibroblast growth factor (EGF) and the secretion of type I collagen, fibronectin, chondroitin sulfate, and other components of the extracellular matrix (ECM) are also directly involved in the epithelial–mesenchymal transition (EMT) and fibrosis [275]. Recent studies had discovered that MMP also contributes to renal fibrosis [276]. There are several severe molecules and pathways involved in the complicated process of renal fibrosis, for instance, the Wnt/β-catenin and TGF-β1/Smad pathways, which are also key targets for botanicals. As the most vital target, Smad3-mediated TGF-β stimulation not only induces collagen production and the inhibition of ECM degradation, but also depresses fatty acid oxidation, leading to a profibrotic phenotype [275].
Botanicals with medicine–food homology happen to be abandoned in natural antioxidant, anti-inflammation, and anti-fibrosis compounds regulating the above pathways and inflammatory cytokines as shown in Table 2. The majority of the mechanisms underlying the protective effects are associated with these mechanisms, underscoring the significant potential and importance of incorporating daily dietary botanicals and supplements into one’s routine.

4.2. Regulating the “Gut–Kidney Axis”

In addition to the classical molecular pathological processes centered on inflammation, oxidative stress, and cellular autophagy and apoptosis, a large number of studies have recently revealed a relationship between intestinal disorders and renal disease, a link that has been increasingly emphasized and investigated under the term “gut–kidney axis” [4]. It has been suggested that the gut–renal axis can be subdivided into metabolism-dependent and immunologic pathways [277]. Metabolism-dependent pathways are mainly mediated by metabolites produced by the gut microbiota that have the ability to modulate host physiological functions. For example, alterations in the gut microbiota can lead to intestinal metabolism and dysfunction, thereby increasing uremic toxin production and renal dysfunction [190,278]. On the other hand, kidney disease also affects the composition of gut bacteria and could cause gastrointestinal dysfunction [279,280]. In addition to uremic toxins like pCS, the gut microbiota produces a large number of physiologically active substances such as SCFAs, and bile acids (BAs), which play an important role in the regulation of renal disease and function [4]. Studies have found a reduction in SCFA-producing gut bacteria including Lactobacillaceae and Prevotella species in ESRD patients compared to healthy individuals [281,282]. However, in some models of kidney disease, a high-fiber diet that promotes the growth of SCFA-producing bacteria and direct supplementation with SCFAs attenuated renal fibrosis [283]. As for the immune pathway, components of the immune system (e.g., lymphocytes, monocytes, and cytokines) play a key role in the communication between the gut and the kidney [284]. A growing number of studies have demonstrated that gut flora play a crucial role in mucosal immunity and systemic inflammation, and that changes in gut flora induce changes in inflammatory cytokine profiles in the bone marrow [285], which are further associated with the development of autoimmune nephropathy [286]. Lymphocytes could even migrate from the gut to the injured kidney via the chemokine pathway [284]. Recent studies have also found that the aberrant galactose-deficient IgA, which is key in the pathogenesis of IgA nephropathy, is associated with aberrant hydrolysis by intestinal bacteria, and is recognized in vivo through the metabolism of the intestinal bacteria exposing aberrant antigenic epitopes, which leads to the formation of immune complexes and renal deposition [286].
As attention is increasingly focused on the gut microbiota, their interaction with botanicals has been discovered and showed a promising future [287,288,289,290,291,292]. As shown in Table 2, almost all kinds of compounds have the potential in regulating gut microbiota. Panax notoginseng saponins, the bioactive components of a well-known and widely used botanical, Panax notoginseng, in Table S1, could regulate intestinal microorganisms and therefore ameliorate inflammation and fibrosis. Similarly to terpenoids, the renal protection by oleanic acid was also found to be associated with its regulation on gut microbiota [147]. Berberine had also been found to ameliorate chronic kidney disease through inhibiting the production of gut-derived uremic toxins, for example, TMAO and pCS in the gut microbiota [190]. Astragalus membranaceus, of which the anti-inflammation and fibrosis effects were confirmed, was also found to influence the gut microbiota, with Akkermansia muciniphila and Lactobacillus being the main driving bacteria [97]. More and more renal-protective effects of botanicals are found to be associated with the gut–kidney axis. Meanwhile, the gut microbiota exhibit rapid alterations in response to daily diets [293,294], indicating the potential for dietary nutrients to intervene in the gut–kidney axis.

5. Conclusions and Perspective

The concept of “food as medicine” underpins the traditional use of these botanicals, especially within the framework of TCM. This approach, which integrates dietary and medicinal uses, provides a unique and holistic perspective on disease prevention and health maintenance. The review emphasizes that many botanicals have been traditionally used for both food and therapeutic purposes, demonstrating their dual benefits. The diverse bioactive compounds found in these botanicals, such as flavonoids, polysaccharides, saponins, alkaloids, and polyphenols, demonstrate a range of protective effects on renal health. These effects are primarily mediated through antioxidation, anti-inflammation, anti-fibrosis, the modulation of the immune response, and enhancement in mitochondrial function as shown in Figure 2. The integration of these botanicals into modern therapeutic regimes could offer a complementary approach to conventional treatments for kidney diseases, which often focus on symptom management rather than disease reversal. The promising results from experimental studies suggest that incorporating these natural compounds into dietary habits or as supplements could contribute to better renal health outcomes.
A particularly notable aspect highlighted in this review is the interplay between these botanicals and the gut–kidney axis. This emerging area of research emphasizes the complex relationship between gut microbiota and renal function. Botanical compounds can influence the composition and activity of gut microbiota, which in turn affects systemic inflammation, immune responses, and metabolic processes relevant to kidney health. For instance, the modulation of gut microbiota by polysaccharides and polyphenols can lead to the production of beneficial metabolites, such as short-chain fatty acids, which have protective effects on the kidneys. Considering the close relation between daily diets and gut microbiota, the potential for dietary nutrients to intervene in the gut–kidney axis is worth deeper exploration.
In summary, botanical ingredients with medicine–food homology present a valuable resource for developing new strategies in the prevention and management of kidney diseases. By leveraging their effects on the gut–kidney axis and other pathways, these botanicals hold promise for enhancing renal health and improving patient outcomes. Continued research and clinical validation are essential to fully realize their potential in this domain.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/nu16203530/s1, Table S1: Botanicals that are allowed to be added to dietary supplements in China.

Author Contributions

Conceptualization and writing—original draft preparation, Y.Z.; resources, R.F.; methodology, M.-L.Y.; data curation, J.-C.H.; visualization, H.X.; writing—review and editing, J.-Y.S.; funding acquisition, Y.W.; supervision, L.-M.C., J.-D.J., and Y.W. All authors have read and agreed to the published version of the manuscript.

Funding

We acknowledge financial support from the National Key R&D Program of China (Grant No.: 2022YFA0806400), the National Natural Science Foundation of China (Nos. 82173888 and 81973290), the Medical and Health Technology Innovation Project of Chinese Academy of Medical Sciences (2021-I2M-1–007, 2023-12M-2-006, 2021-I2M-1–028), and the Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD study (Z141102004414062).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

We express our gratitude to Bin Zhao (Department of Pharmacy, Peking Union Medical College Hospital) for his help. We would like to thank Shimadzu Co., Ltd. (Shanghai, China) for technological support. Special thanks to Siyu Ou for the help with the illustration.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Wu, H.; Huang, J. Drug-Induced Nephrotoxicity: Pathogenic Mechanisms, Biomarkers and Prevention Strategies. Curr. Drug Metab. 2018, 19, 559–567. [Google Scholar] [CrossRef] [PubMed]
  2. Mody, H.; Ramakrishnan, V.; Chaar, M.; Lezeau, J.; Rump, A.; Taha, K.; Lesko, L.; Ait-Oudhia, S. A Review on Drug-Induced Nephrotoxicity: Pathophysiological Mechanisms, Drug Classes, Clinical Management, and Recent Advances in Mathematical Modeling and Simulation Approaches. Clin. Pharmacol. Drug Dev. 2020, 9, 896–909. [Google Scholar] [CrossRef]
  3. Schetz, M.; Dasta, J.; Goldstein, S.; Golper, T. Drug-Induced Acute Kidney Injury. Curr. Opin. Crit. Care 2005, 11, 555–565. [Google Scholar] [CrossRef]
  4. Yang, T.; Richards, E.M.; Pepine, C.J.; Raizada, M.K. The Gut Microbiota and the Brain–Gut–Kidney Axis in Hypertension and Chronic Kidney Disease. Nat. Rev. Nephrol. 2018, 14, 442–456. [Google Scholar] [CrossRef]
  5. Gaitonde, D.Y.; Cook, D.L.; Rivera, I.M. Chronic Kidney Disease: Detection and Evaluation. Am. Fam. Physician 2017, 96, 776–783. [Google Scholar]
  6. Tain, Y.L.; Chang, C.I.; Hou, C.Y.; Chang-Chien, G.P.; Lin, S.F.; Hsu, C.N. Resveratrol Propionate Ester Supplement Exerts Antihypertensive Effect in Juvenile Rats Exposed to an Adenine Diet Via Gut Microbiota Modulation. Nutrients 2024, 16, 2131. [Google Scholar] [CrossRef] [PubMed]
  7. Bellomo, F.; Pugliese, S.; Cairoli, S.; Krohn, P.; De Stefanis, C.; Raso, R.; Rega, L.R.; Taranta, A.; De Leo, E.; Ciolfi, A.; et al. Ketogenic Diet and Progression of Kidney Disease in Animal Models of Nephropathic Cystinosis. J. Am. Soc. Nephrol. 2024, 10-1681. [Google Scholar] [CrossRef] [PubMed]
  8. Yang, C.; Wang, T.; Chen, J.; He, J.; Li, Y.; Chen, C.; Lu, G.; Chen, W. Traditional Chinese Medicine Formulas Alleviate Acute Pancreatitis: Pharmacological Activities and Mechanisms. Pancreas 2021, 50, 1348–1356. [Google Scholar] [CrossRef]
  9. Wang, Y.; Li, M.; Liang, Y.; Yang, Y.; Liu, Z.; Yao, K.; Chen, Z.; Zhai, S. Chinese Herbal Medicine for the Treatment of Depression: Applications, Efficacies and Mechanisms. Curr. Pharm. Des. 2017, 23, 5180–5190. [Google Scholar] [CrossRef]
  10. Chan, H.H.L.; Ng, T. Traditional Chinese Medicine (Tcm) and Allergic Diseases. Curr. Allergy Asthma Rep. 2020, 20, 67. [Google Scholar] [CrossRef]
  11. Wojcikowski, K.; Wohlmuth, H.; Johnson, D.W.; Rolfe, M.; Gobe, G. An In Vitro Investigation of Herbs Traditionally Used for Kidney and Urinary System Disorders: Potential Therapeutic and Toxic Effects. Nephrology 2009, 14, 70–79. [Google Scholar] [CrossRef] [PubMed]
  12. Li, C.; Huang, H.; Wang, R.; Zhang, C.; Huang, S.; Wu, J.; Mo, P.; Yu, H.; Li, S.; Chen, J.; et al. Formula Restores Iron Metabolism from Dysregulation in Anemic Rats with Adenine-Induced Nephropathy. J. Ethnopharmacol. 2023, 312, 116526. [Google Scholar] [CrossRef] [PubMed]
  13. Ll, J.; Yu, D.R.; Chen, H.Y.; Zhu, C.F.; Cheng, X.X.; Wang, Y.H.; Ni, J.; Wang, X.J.; Jinag, F. Long-Term Effect of the Treatment of Iga Nephropathy by Tonifying Shen, Activating Blood Stasis, Dispelling Wind-Dampness Combined with Western Medicine. Zhongguo Zhong Xi Yi Jie He Za Zhi 2017, 37, 28–33. [Google Scholar] [PubMed]
  14. Peng, M.; Cai, P.; Ma, H.; Meng, H.; Xu, Y.; Zhang, X.; Si, G. Chinese Herbal Medicine Shenqi Detoxification Granule Inhibits Fibrosis in Adenine Induced Chronic Renal Failure Rats. Afr. J. Tradit. Complement. Altern. Med. 2014, 11, 194–204. [Google Scholar] [PubMed]
  15. Notice of the National Health Commission on Further Regulating the Management of Health Food Raw Materials (No. 51 of 2002); National Health Commission of the People’s Republic of China: Beijing, China, 2002.
  16. Wan, F.; Ma, F.; Wu, J.; Qiao, X.; Chen, M.; Li, W.; Ma, L. Effect of Lycium barbarum Polysaccharide on Decreasing Serum Amyloid A3 Expression through Inhibiting Nf-Κb Activation in a Mouse Model of Diabetic Nephropathy. Anal. Cell. Pathol. 2022, 2022, 7847135. [Google Scholar] [CrossRef]
  17. Li, W.; Yu, L.; Fu, B.; Chu, J.; Chen, C.; Li, X.; Ma, J.; Tang, W. Protective Effects of Polygonatum kingianum Polysaccharides and Aqueous Extract on Uranium-Induced Toxicity in Human Kidney (Hk-2) Cells. Int. J. Biol. Macromol. 2022, 202, 68–79. [Google Scholar] [CrossRef]
  18. Yue, L.; Wang, W.; Wang, Y.; Du, T.; Shen, W.; Tang, H.; Wang, Y.; Yin, H. Bletilla striata Polysaccharide Inhibits Angiotensin Ii-Induced Ros and Inflammation Via Nox4 and Tlr2 Pathways. Int. J. Biol. Macromol. 2016, 89, 376–388. [Google Scholar] [CrossRef]
  19. Ma, Q.; Xu, Y.; Tang, L.; Yang, X.; Chen, Z.; Wei, Y.; Shao, X.; Shao, X.; Xin, Z.; Cai, B.; et al. Astragalus Polysaccharide Attenuates Cisplatin-Induced Acute Kidney Injury by Suppressing Oxidative Damage and Mitochondrial Dysfunction. BioMed Res. Int. 2020, 2020, 2851349. [Google Scholar] [CrossRef]
  20. Zhao, K.; Wu, X.; Han, G.; Sun, L.; Zheng, C.; Hou, H.; Xu, B.B.; El-Bahy, Z.M.; Qian, C.; Kallel, M.; et al. Phyllostachys nigra (Lodd. Ex Lindl.) Derived Polysaccharide with Enhanced Glycolipid Metabolism Regulation and Mice Gut Microbiome. Int. J. Biol. Macromol. 2024, 257 Pt 1, 128588. [Google Scholar] [CrossRef]
  21. Dwyer, J.T.; Coates, P.M.; Smith, M.J. Dietary Supplements: Regulatory Challenges and Research Resources. Nutrients 2018, 10, 41. [Google Scholar] [CrossRef]
  22. Nicastro, H.L.; Vorkoper, S.; Sterling, R.; Korn, A.R.; Brown, A.G.M.; Maruvada, P.; Oh, A.Y. Opportunities to Advance Implementation Science and Nutrition Research: A Commentary on the Strategic Plan for Nih Nutrition Research. Transl. Behav. Med. 2023, 13, 1–6. [Google Scholar] [CrossRef] [PubMed]
  23. Bilia, A.R. Herbal Medicinal Products Versus Botanical-Food Supplements in the European Market: State of Art and Perspectives. Nat. Prod. Commun. 2015, 10, 125–131. [Google Scholar] [CrossRef] [PubMed]
  24. Luciano, R.L.; Perazella, M.A. Aristolochic Acid Nephropathy: Epidemiology, Clinical Presentation, and Treatment. Drug Saf. 2015, 38, 55–64. [Google Scholar] [CrossRef] [PubMed]
  25. Ban, T.H.; Min, J.W.; Seo, C.; Kim, D.R.; Lee, Y.H.; Chung, B.H.; Jeong, K.H.; Lee, J.W.; Kim, B.S.; Lee, S.H.; et al. Update of Aristolochic Acid Nephropathy in Korea. Korean J. Intern. Med. 2018, 33, 961–969. [Google Scholar] [CrossRef] [PubMed]
  26. Dugo, M.; Gatto, R.; Zagatti, R.; Gatti, P.; Cascone, C. Herbal Remedies: Nephrotoxicity and Drug Interactions. G. Ital. Nefrol. 2010, 27 (Suppl. S52), S5–S9. [Google Scholar]
  27. Yang, H.; Zhao, Y.; Ren, B.; Wu, Y.; Qiu, Z.; Cheng, Y.; Qiu, B. Poria Acid Inhibit the Growth and Metastasis of Renal Cell Carcinoma by Inhibiting the Pi3k/Akt/Nf-Κb Signaling Pathway. Heliyon 2024, 10, e31106. [Google Scholar] [CrossRef]
  28. Lu, M.; Yin, J.; Xu, T.; Dai, X.; Liu, T.; Zhang, Y.; Wang, S.; Liu, Y.; Shi, H.; Zhang, Y.; et al. Fuling-Zexie Formula Attenuates Hyperuricemia-Induced Nephropathy and Inhibits Jak2/Stat3 Signaling and Nlrp3 Inflammasome Activation in Mice. J. Ethnopharmacol. 2024, 319 Pt 2, 117262. [Google Scholar] [CrossRef]
  29. Notice on Six New Substances Including Angelica Sinensis and Other Substances Which Are Both Food and Chinese Herbal Medicine According to Tradition (No. 8 of 2019); Food Safety Standards and Evaluation Division (Ed.) National Health Commission of the People’s Republic of China: Beijing, China, 2019.
  30. Notice on Nine New Substances Including Codonopsis Pilosula and Other Substances Which Are Both Food and Chinese Herbal Medicine According to Tradition (No. 9 of 2023); Food Safety Standards and Evaluation Division (Ed.) National Health Commission of the People’s Republic of China: Beijing, China, 2023.
  31. Regulations on Catalog of Substances That Are Traditionally Used as Both Food and Herbal Medicine; Food Safety Standards and Evaluation Division (Ed.) National Health Commission of the People’s Republic of China: Beijing, China, 2021.
  32. Vettorazzi, A.; de Cerain, A.L.; Sanz-Serrano, J.; Gil, A.G.; Azqueta, A. European Regulatory Framework and Safety Assessment of Food-Related Bioactive Compounds. Nutrients 2020, 12, 613. [Google Scholar] [CrossRef]
  33. Bailey, R.L. Current Regulatory Guidelines and Resources to Support Research of Dietary Supplements in the United States. Crit. Rev. Food Sci. Nutr. 2018, 60, 298–309. [Google Scholar] [CrossRef]
  34. US Department of Health and Human Services, National Institutes of Health, Office of Dietary Supplements. Dietary Supplement Label Database (Dsld); US Department of Health and Human Services, National Institutes of Health, Office of Dietary Supplements: Bethesda, MD, USA, 2013.
  35. Liu, Y.; Luo, J.; Peng, L.; Zhang, Q.; Rong, X.; Luo, Y.; Li, J. Flavonoids: Potential Therapeutic Agents for Cardiovascular Disease. Heliyon 2024, 10, e32563. [Google Scholar] [CrossRef]
  36. Frydman, A.; Weisshaus, O.; Bar-Peled, M.; Huhman, D.V.; Sumner, L.W.; Marin, F.R.; Lewinsohn, E.; Fluhr, R.; Gressel, J.; Eyal, Y. Citrus Fruit Bitter Flavors: Isolation and Functional Characterization of the Gene Cm1,2rhat Encoding a 1,2 Rhamnosyltransferase, a Key Enzyme in the Biosynthesis of the Bitter Flavonoids of Citrus. Plant J. 2004, 40, 88–100. [Google Scholar] [CrossRef]
  37. Zhang, L.; Wang, X.; Chang, L.; Ren, Y.; Sui, M.; Fu, Y.; Zhang, L.; Hao, L. Quercetin Improves Diabetic Kidney Disease by Inhibiting Ferroptosis and Regulating the Nrf2 in Streptozotocin-Induced Diabetic Rats. Ren. Fail. 2024, 46, 2327495. [Google Scholar] [CrossRef] [PubMed]
  38. Guo, X.; Wen, S.; Wang, J.; Zeng, X.; Yu, H.; Chen, Y.; Zhu, X.; Xu, L. Senolytic Combination of Dasatinib and Quercetin Attenuates Renal Damage in Diabetic Kidney Disease. Phytomedicine 2024, 130, 155705. [Google Scholar] [CrossRef] [PubMed]
  39. Zeng, Y.F.; Li, J.Y.; Wei, X.Y.; Ma, S.Q.; Wang, Q.G.; Qi, Z.; Duan, Z.C.; Tan, L.; Tang, H. Preclinical Evidence of Reno-Protective Effect of Quercetin on Acute Kidney Injury: A Meta-Analysis of Animal Studies. Front. Pharmacol. 2023, 14, 1310023. [Google Scholar] [CrossRef]
  40. Xu, Z.; Wang, X.; Kuang, W.; Wang, S.; Zhao, Y. Kaempferol Improves Acute Kidney Injury Via Inhibition of Macrophage Infiltration in Septic Mice. Biosci. Rep. 2023, 43, BSR20230873. [Google Scholar] [CrossRef] [PubMed]
  41. Yuan, N.; Diao, J.; Dong, J.; Yan, Y.; Chen, Y.; Yan, S.; Liu, C.; He, Z.; He, J.; Zhang, C.; et al. Targeting Rock1 in Diabetic Kidney Disease: Unraveling Mesangial Fibrosis Mechanisms and Introducing Myricetin as a Novel Antagonist. Biomed. Pharmacother. 2024, 171, 116208. [Google Scholar] [CrossRef]
  42. Yang, Z.J.; Wang, H.R.; Wang, Y.I.; Zhai, Z.H.; Wang, L.W.; Li, L.; Zhang, C.; Tang, L. Myricetin Attenuated Diabetes-Associated Kidney Injuries and Dysfunction Via Regulating Nuclear Factor (Erythroid Derived 2)-Like 2 and Nuclear Factor-Κb Signaling. Front. Pharmacol. 2019, 10, 647. [Google Scholar] [CrossRef]
  43. Liu, P.; Tang, L.; Li, G.; Wu, X.; Hu, F.; Peng, W. Association between Consumption of Flavonol and Its Subclasses and Chronic Kidney Disease in Us Adults: An Analysis Based on National Health and Nutrition Examination Survey Data from 2007–2008, 2009–2010, and 2017–2018. Front. Nutr. 2024, 11, 1399251. [Google Scholar] [CrossRef]
  44. Jian, J.; Li, Y.-Q.; Han, R.-Y.; Zhong, X.; Xie, K.-H.; Yan, Y.; Wang, L.; Tan, R.-Z. Isorhamnetin Ameliorates Cisplatin-Induced Acute Kidney Injury in Mice by Activating Slpi-Mediated Anti-Inflammatory Effect in Macrophage. Immunopharmacol. Immunotoxicol. 2024, 46, 319–329. [Google Scholar] [CrossRef] [PubMed]
  45. Wang, B.; Yang, L.N.; Yang, L.T.; Liang, Y.; Guo, F.; Fu, P.; Ma, L. Fisetin Ameliorates Fibrotic Kidney Disease in Mice Via Inhibiting Acsl4-Mediated Tubular Ferroptosis. Acta Pharmacol. Sin. 2024, 45, 150–165. [Google Scholar] [CrossRef]
  46. Zou, T.F.; Liu, Z.G.; Cao, P.C.; Zheng, S.H.; Guo, W.T.; Wang, T.X.; Chen, Y.L.; Duan, Y.J.; Li, Q.S.; Liao, C.Z.; et al. Fisetin Treatment Alleviates Kidney Injury in Mice with Diabetes-Exacerbated Atherosclerosis through Inhibiting Cd36/Fibrosis Pathway. Acta Pharmacol. Sin. 2023, 44, 2065–2074. [Google Scholar] [CrossRef]
  47. Zhang, D.; Liu, S.; Jiang, H.; Liu, S.; Kong, F. Dia Proteomics Analysis Reveals the Mechanism of Folic Acid-Induced Acute Kidney Injury and the Effects of Icariin. Chem. Biol. Interact. 2024, 390, 110878. [Google Scholar] [CrossRef] [PubMed]
  48. Zhao, Y.; Yang, W.; Zhang, X.; Lv, C.; Lu, J. Icariin, the Main Prenylflavonoid of Epimedii Folium, Ameliorated Chronic Kidney Disease by Modulating Energy Metabolism Via Ampk Activation. J. Ethnopharmacol. 2023, 312, 116543. [Google Scholar] [CrossRef] [PubMed]
  49. Owumi, S.E.; Kazeem, A.I.; Wu, B.; Ishokare, L.O.; Arunsi, U.O.; Oyelere, A.K. Apigeninidin-Rich Sorghum bicolor (L. Moench) Extracts Suppress A549 Cells Proliferation and Ameliorate Toxicity of Aflatoxin B1-Mediated Liver and Kidney Derangement in Rats. Sci. Rep. 2022, 12, 7438. [Google Scholar] [CrossRef] [PubMed]
  50. Amorim, J.M.; de Souza, L.C.R.; de Souza, R.A.L.; da Silva Filha, R.; de Oliveira Silva, J.; de Almeida Araújo, S.; Tagliti, C.A.; Simões, E.S.A.C.; Castilho, R.O. Costus Spiralis Extract Restores Kidney Function in Cisplatin-Induced Nephrotoxicity Model: Ethnopharmacological Use, Chemical and Toxicological Investigation. J. Ethnopharmacol. 2022, 299, 115510. [Google Scholar] [CrossRef] [PubMed]
  51. Wang, X.; Kim, C.S.; Adams, B.C.; Wilkinson, R.; Hill, M.M.; Shah, A.K.; Mohamed, A.; Dutt, M.; Ng, M.S.Y.; Ungerer, J.P.J.; et al. Human Proximal Tubular Epithelial Cell-Derived Small Extracellular Vesicles Mediate Synchronized Tubular Ferroptosis in Hypoxic Kidney Injury. Redox Biol. 2024, 70, 103042. [Google Scholar] [CrossRef]
  52. Guo, S.; Zhou, L.; Liu, X.; Gao, L.; Li, Y.; Wu, Y. Baicalein Alleviates Cisplatin-Induced Acute Kidney Injury by Inhibiting Alox12-Dependent Ferroptosis. Phytomedicine 2024, 130, 155757. [Google Scholar] [CrossRef]
  53. Hu, H.; Li, W.; Hao, Y.; Peng, Z.; Zou, Z.; Liang, W. Baicalin Ameliorates Renal Fibrosis by Upregulating Cpt1α-Mediated Fatty Acid Oxidation in Diabetic Kidney Disease. Phytomedicine 2024, 122, 155162. [Google Scholar] [CrossRef]
  54. Miguel, V.; Rey-Serra, C.; Tituaña, J.; Sirera, B.; Alcalde-Estévez, E.; Herrero, J.I.; Ranz, I.; Fernández, L.; Castillo, C.; Sevilla, L.; et al. Enhanced Fatty Acid Oxidation through Metformin and Baicalin as Therapy for COVID-19 and Associated Inflammatory States in Lung and Kidney. Redox Biol. 2023, 68, 102957. [Google Scholar] [CrossRef]
  55. Liu, B.; Deng, Q.; Zhang, L.; Zhu, W. Nobiletin Alleviates Ischemia/Reperfusion Injury in the Kidney by Activating Pi3k/Akt Pathway. Mol. Med. Rep. 2020, 22, 4655–4662. [Google Scholar] [CrossRef]
  56. Song, J.; Wang, H.; Sheng, J.; Zhang, W.; Lei, J.; Gan, W.; Cai, F.; Yang, Y. Vitexin Attenuates Chronic Kidney Disease by Inhibiting Renal Tubular Epithelial Cell Ferroptosis Via Nrf2 Activation. Mol. Med. 2023, 29, 147. [Google Scholar] [CrossRef]
  57. Ding, T.; Zhao, T.; Li, Y.; Liu, Z.; Ding, J.; Ji, B.; Wang, Y.; Guo, Z. Vitexin Exerts Protective Effects against Calcium Oxalate Crystal-Induced Kidney Pyroptosis In Vivo and In Vitro. Phytomedicine 2021, 86, 153562. [Google Scholar] [CrossRef] [PubMed]
  58. Yang, Q.; Qian, L.; He, S.; Zhang, C. Hesperidin Alleviates Zinc-Induced Nephrotoxicity Via the Gut-Kidney Axis in Swine. Front. Cell Infect. Microbiol. 2024, 14, 1390104. [Google Scholar] [CrossRef] [PubMed]
  59. Chen, J.; Fan, X.; Chen, J.; Luo, X.; Huang, X.; Zhou, Z.; He, Y.; Feng, S.; Jiao, Y.; Wang, R.; et al. Effects of Hesperidin on the Histological Structure, Oxidative Stress, and Apoptosis in the Liver and Kidney Induced by Nicl(2). Front. Vet. Sci. 2024, 11, 1424711. [Google Scholar] [CrossRef] [PubMed]
  60. Yang, A.Y.; Choi, H.J.; Kim, K.; Leem, J. Antioxidant, Antiapoptotic, and Anti-Inflammatory Effects of Hesperetin in a Mouse Model of Lipopolysaccharide-Induced Acute Kidney Injury. Molecules 2023, 28, 2759. [Google Scholar] [CrossRef]
  61. Chen, X.; Wei, W.; Li, Y.; Huang, J.; Ci, X. Hesperetin Relieves Cisplatin-Induced Acute Kidney Injury by Mitigating Oxidative Stress, Inflammation and Apoptosis. Chem. Biol. Interact. 2019, 308, 269–278. [Google Scholar] [CrossRef]
  62. Chen, C.; Feng, F.; Qi, M.; Chen, Q.; Tang, W.; Diao, H.; Hu, Z.; Qiu, Y.; Li, Z.; Chu, Y.; et al. Dietary Citrus Flavonoids Improved Growth Performance and Intestinal Microbiota of Weaned Piglets Via Immune Function Mediated by Tlr2/Nf-Κb Signaling Pathway. J. Agric. Food Chem. 2024, 72, 16761–16776. [Google Scholar] [CrossRef]
  63. Chang, Y.W.; Chen, Y.L.; Park, S.H.; Yap, E.E.S.; Sung, W.C. Characterization of Functional Ingredients Extracted with Ethanol Solvents from Ponkan (Citrus reticulata) by-Products Using the Microwave Vacuum Drying Method Combined with Ultrasound-Assisted Extraction. Foods 2024, 13, 2129. [Google Scholar] [CrossRef]
  64. Chen, Y.; Pan, H.; Hao, S.; Pan, D.; Wang, G.; Yu, W. Evaluation of Phenolic Composition and Antioxidant Properties of Different Varieties of Chinese Citrus. Food Chem. 2021, 364, 130413. [Google Scholar] [CrossRef]
  65. Kamel, K.M.; El-Raouf, O.M.A.; Metwally, S.A.; El-Latif, H.A.A.; El-sayed, M.E. Hesperidin and Rutin, Antioxidant Citrus Flavonoids, Attenuate Cisplatin-Induced Nephrotoxicity in Rats. J. Biochem. Mol. Toxicol. 2014, 28, 312–319. [Google Scholar] [CrossRef]
  66. Jung, H.A.; Jung, M.J.; Kim, J.Y.; Chung, H.Y.; Choi, J.S. Inhibitory Activity of Flavonoids from Prunus Davidiana and Other Flavonoids on Total Ros and Hydroxyl Radical Generation. Arch. Pharm. Res. 2003, 26, 809–815. [Google Scholar] [CrossRef]
  67. Mbara, K.C.; Fotsing, M.C.D.; Ndinteh, D.T.; Mbeb, C.N.; Nwagwu, C.S.; Khan, R.; Mokhetho, K.C.; Baijnath, H.; Nlooto, M.; Mokhele, S.; et al. Endoplasmic Reticulum Stress in Pancreatic Β-Cell Dysfunction: The Potential Therapeutic Role of Dietary Flavonoids. Curr. Res. Pharmacol. Drug Discov. 2024, 6, 100184. [Google Scholar] [CrossRef] [PubMed]
  68. Ramos, F.M.M.; Ribeiro, C.B.; Cesar, T.B.; Milenkovic, D.; Cabral, L.; Noronha, M.F.; Sivieri, K. Lemon Flavonoids Nutraceutical (Eriomin®) Attenuates Prediabetes Intestinal Dysbiosis: A Double-Blind Randomized Controlled Trial. Food Sci. Nutr. 2023, 11, 7283–7295. [Google Scholar] [CrossRef] [PubMed]
  69. Bellavite, P. Neuroprotective Potentials of Flavonoids: Experimental Studies and Mechanisms of Action. Antioxidants 2023, 12, 280. [Google Scholar] [CrossRef] [PubMed]
  70. Pabich, M.; Materska, M. Biological Effect of Soy Isoflavones in the Prevention of Civilization Diseases. Nutrients 2019, 11, 1660. [Google Scholar] [CrossRef]
  71. Misiakiewicz-Has, K.; Maciejewska-Markiewicz, D.; Szypulska-Koziarska, D.; Kolasa, A.; Wiszniewska, B. The Influence of Soy Isoflavones and Soy Isoflavones with Inulin on Kidney Morphology, Fatty Acids, and Associated Parameters in Rats with and without Induced Diabetes Type 2. Int. J. Mol. Sci. 2024, 25, 5418. [Google Scholar] [CrossRef]
  72. Hu, Z.; Chen, D.; Yan, P.; Zheng, F.; Zhu, H.; Yuan, Z.; Yang, X.; Zuo, Y.; Chen, C.; Lu, H.; et al. Puerarin Suppresses Macrophage M1 Polarization to Alleviate Renal Inflammatory Injury through Antagonizing Tlr4/Myd88-Mediated Nf-Κb P65 and Jnk/Foxo1 Activation. Phytomedicine 2024, 132, 155813. [Google Scholar] [CrossRef]
  73. Zhang, S.S.; Zhang, N.N.; Guo, S.; Liu, S.J.; Hou, Y.F.; Li, S.; Ho, C.T.; Bai, N.S. Glycosides and Flavonoids from the Extract of Pueraria Thomsonii Benth Leaf Alleviate Type 2 Diabetes in High-Fat Diet Plus Streptozotocin-Induced Mice by Modulating the Gut Microbiota. Food Funct. 2022, 13, 3931–3945. [Google Scholar] [CrossRef]
  74. Zhou, Y.; Lu, W.; Huang, K.; Gan, F. Ferroptosis Is Involved in Quercetin-Mediated Alleviation of Ochratoxin a-Induced Kidney Damage. Food Chem. Toxicol. 2024, 191, 114877. [Google Scholar] [CrossRef] [PubMed]
  75. Liu, F.; Feng, Q.; Yang, M.; Yang, Y.; Nie, J.; Wang, S. Quercetin Prevented Diabetic Nephropathy by Inhibiting Renal Tubular Epithelial Cell Apoptosis Via the Pi3k/Akt Pathway. Phytother. Res. 2024, 38, 3594–3606. [Google Scholar] [CrossRef]
  76. Sheng, H.; Zhang, D.; Zhang, J.; Zhang, Y.; Lu, Z.; Mao, W.; Liu, X.; Zhang, L. Kaempferol Attenuated Diabetic Nephropathy by Reducing Apoptosis and Promoting Autophagy through Ampk/Mtor Pathways. Front. Med. 2022, 9, 986825. [Google Scholar] [CrossRef]
  77. Wu, Q.; Chen, J.; Zheng, X.; Song, J.; Yin, L.; Guo, H.; Chen, Q.; Liu, Y.; Ma, Q.; Zhang, H.; et al. Kaempferol Attenuates Doxorubicin-Induced Renal Tubular Injury by Inhibiting Ros/Ask1-Mediated Activation of the Mapk Signaling Pathway. Biomed. Pharmacother. 2023, 157, 114087. [Google Scholar] [CrossRef] [PubMed]
  78. Guan, Y.; Quan, D.; Chen, K.; Kang, L.; Yang, D.; Wu, H.; Yan, M.; Wu, S.; Lv, L.; Zhang, G. Kaempferol Inhibits Renal Fibrosis by Suppression of the Sonic Hedgehog Signaling Pathway. Phytomedicine 2023, 108, 154246. [Google Scholar] [CrossRef] [PubMed]
  79. Yang, X.; Zhang, P.; Jiang, J.; Almoallim, H.S.; Alharbi, S.A.; Li, Y. Myricetin Attenuates Ethylene Glycol-Induced Nephrolithiasis in Rats Via Mitigating Oxidative Stress and Inflammatory Markers. Appl. Biochem. Biotechnol. 2023, 196, 5419–5434. [Google Scholar] [CrossRef] [PubMed]
  80. Liu, J.; Xie, L.; Zhai, H.; Wang, D.; Li, X.; Wang, Y.; Song, M.; Xu, C. Exploration of the Protective Mechanisms of Icariin against Cisplatin-Induced Renal Cell Damage in Canines. Front. Vet. Sci. 2024, 11, 1331409. [Google Scholar] [CrossRef] [PubMed]
  81. Ding, N.; Sun, S.; Zhou, S.; Lv, Z.; Wang, R. Icariin Alleviates Renal Inflammation and Tubulointerstitial Fibrosis Via Nrf2-Mediated Attenuation of Mitochondrial Damage. Cell Biochem. Funct. 2024, 42, e4005. [Google Scholar] [CrossRef]
  82. Duan, S.; Ding, Z.; Liu, C.; Wang, X.; Dai, E. Icariin Suppresses Nephrotic Syndrome by Inhibiting Pyroptosis and Epithelial-to-Mesenchymal Transition. PLoS ONE 2024, 19, e0298353. [Google Scholar] [CrossRef]
  83. Wu, Q.; Li, W.; Zhao, J.; Sun, W.; Yang, Q.; Chen, C.; Xia, P.; Zhu, J.; Zhou, Y.; Huang, G.; et al. Apigenin Ameliorates Doxorubicin-Induced Renal Injury Via Inhibition of Oxidative Stress and Inflammation. Biomed. Pharmacother. 2021, 137, 111308. [Google Scholar] [CrossRef]
  84. Li, Y.; Zhao, Z.; Luo, J.; Jiang, Y.; Li, L.; Chen, Y.; Zhang, L.; Huang, Q.; Cao, Y.; Zhou, P.; et al. Apigenin Ameliorates Hyperuricemic Nephropathy by Inhibiting Urat1 and Glut9 and Relieving Renal Fibrosis Via the Wnt/Β-Catenin Pathway. Phytomedicine 2021, 87, 153585. [Google Scholar] [CrossRef]
  85. Zamani, F.; Samiei, F.; Mousavi, Z.; Azari, M.R.; Seydi, E.; Pourahmad, J. Apigenin Ameliorates Oxidative Stress and Mitochondrial Damage Induced by Multiwall Carbon Nanotubes in Rat Kidney Mitochondria. J. Biochem. Mol. Toxicol. 2021, 35, 1–7. [Google Scholar] [CrossRef]
  86. Wang, T.; Zhang, Z.; Xie, M.; Li, S.; Zhang, J.; Zhou, J. Apigenin Attenuates Mesoporous Silica Nanoparticles-Induced Nephrotoxicity by Activating Foxo3a. Biol. Trace Elem. Res. 2022, 200, 2793–2806. [Google Scholar] [CrossRef]
  87. Azimi, A.; Eidi, A.; Mortazavi, P.; Rohani, A.H. Protective Effect of Apigenin on Ethylene Glycol-Induced Urolithiasis Via Attenuating Oxidative Stress and Inflammatory Parameters in Adult Male Wistar Rats. Life Sci. 2021, 279, 119641. [Google Scholar] [CrossRef] [PubMed]
  88. Zhang, K.; Li, J.; Dong, W.; Huang, Q.; Wang, X.; Deng, K.; Ali, W.; Song, R.; Zou, H.; Ran, D.; et al. Luteolin Alleviates Cadmium-Induced Kidney Injury by Inhibiting Oxidative DNA Damage and Repairing Autophagic Flux Blockade in Chickens. Antioxidants 2024, 13, 525. [Google Scholar] [CrossRef]
  89. Xu, X.; Yu, Z.; Han, B.; Li, S.; Sun, Y.; Du, Y.; Wang, Z.; Gao, D.; Zhang, Z. Luteolin Alleviates Inorganic Mercury-Induced Kidney Injury Via Activation of the Ampk/Mtor Autophagy Pathway. J. Inorg. Biochem. 2021, 224, 111583. [Google Scholar] [CrossRef] [PubMed]
  90. Ding, T.; Yi, T.; Li, Y.; Zhang, W.; Wang, X.; Liu, J.; Fan, Y.; Ji, J.; Xu, L. Luteolin Attenuates Lupus Nephritis by Regulating Macrophage Oxidative Stress Via Hif-1α Pathway. Eur. J. Pharmacol. 2023, 953, 175823. [Google Scholar] [CrossRef]
  91. Li, F.; Wei, R.; Huang, M.; Chen, J.; Li, P.; Ma, Y.; Chen, X. Luteolin Can Ameliorate Renal Interstitial Fibrosis-Induced Renal Anaemia through the Sirt1/Foxo3 Pathway. Food Funct. 2022, 13, 11896–11914. [Google Scholar] [CrossRef]
  92. Awoyomi, O.V.; Adeoye, Y.D.; Oyagbemi, A.A.; Ajibade, T.O.; Asenuga, E.R.; Gbadamosi, I.T.; Ogunpolu, B.S.; Falayi, O.O.; Hassan, F.O.; Omobowale, T.O.; et al. Luteolin Mitigates Potassium Dichromate-Induced Nephrotoxicity, Cardiotoxicity and Genotoxicity through Modulation of Kim-1/Nrf2 Signaling Pathways. Environ. Toxicol. 2021, 36, 2146–2160. [Google Scholar] [CrossRef] [PubMed]
  93. Zhu, Q.; Yang, S.; Wei, C.; Lu, G.; Lee, K.; He, J.C.; Liu, R.; Zhong, Y. Puerarin Attenuates Diabetic Kidney Injury through Interaction with Guanidine Nucleotide-Binding Protein Gi Subunit Alpha-1 (Gnai1) Subunit. J. Cell Mol. Med. 2022, 26, 3816–3827. [Google Scholar] [CrossRef]
  94. Song, Q.; Jian, W.; Zhang, Y.; Li, Q.; Zhao, Y.; Liu, R.; Zeng, Y.; Zhang, F.; Duan, J. Puerarin Attenuates Iron Overload-Induced Ferroptosis in Retina through a Nrf2-Mediated Mechanism. Mol. Nutr. Food Res. 2024, 68, e2300123. [Google Scholar] [CrossRef]
  95. Jing, G.H.; Liu, Y.D.; Liu, J.N.; Jin, Y.S.; Yu, S.L.; An, R.H. Puerarin Prevents Calcium Oxalate Crystal-Induced Renal Epithelial Cell Autophagy by Activating the Sirt1-Mediated Signaling Pathway. Urolithiasis 2022, 50, 545–556. [Google Scholar] [CrossRef]
  96. Sun, J.; Wei, S.; Zhang, Y.; Li, J. Protective Effects of Astragalus Polysaccharide on Sepsis-Induced Acute Kidney Injury. Anal. Cell. Pathol. 2021, 2021, 7178253. [Google Scholar] [CrossRef]
  97. Li, J.; Zhao, J.; Chai, Y.; Li, W.; Liu, X.; Chen, Y. Astragalus Polysaccharide Protects Sepsis Model Rats after Cecum Ligation and Puncture. Front. Bioeng. Biotechnol. 2022, 10, 1020300. [Google Scholar] [CrossRef] [PubMed]
  98. Wang, Y.; Liu, D.; Chen, S.; Wang, Y.; Jiang, H.; Yin, H. A New Glucomannan from Bletilla Striata: Structural and Anti-Fibrosis Effects. Fitoterapia 2014, 92, 72–78. [Google Scholar] [CrossRef] [PubMed]
  99. Pan, Y.; Zhang, Y.; Li, J.; Zhang, Z.; He, Y.; Zhao, Q.; Yang, H.; Zhou, P. A Proteoglycan Isolated from Ganoderma Lucidum Attenuates Diabetic Kidney Disease by Inhibiting Oxidative Stress-Induced Renal Fibrosis Both In Vitro and In Vivo. J. Ethnopharmacol. 2023, 310, 116405. [Google Scholar] [CrossRef]
  100. Zhong, D.; Wang, H.; Liu, M.; Li, X.; Huang, M.; Zhou, H.; Lin, S.; Lin, Z.; Yang, B. Ganoderma Lucidum Polysaccharide Peptide Prevents Renal Ischemia Reperfusion Injury Via Counteracting Oxidative Stress. Sci. Rep. 2015, 5, 16910. [Google Scholar] [CrossRef]
  101. Xie, W.; Huang, Y.-Y.; Chen, H.-G.; Zhou, X. Study on the Efficacy and Mechanism of Lycium Barbarum Polysaccharide against Lead-Induced Renal Injury in Mice. Nutrients 2021, 13, 2945. [Google Scholar] [CrossRef]
  102. Chen, J.; Wang, W.; Zhang, Q.; Li, F.; Lei, T.; Luo, D.; Zhou, H.; Yang, B. Low Molecular Weight Fucoidan against Renal Ischemia–Reperfusion Injury Via Inhibition of the Mapk Signaling Pathway. PLoS ONE 2013, 8, e56224. [Google Scholar] [CrossRef]
  103. Li, X.-Y.; Chen, H.-R.; Zha, X.-Q.; Chen, S.; Pan, L.-H.; Li, Q.-M.; Luo, J.-P. Prevention and Possible Mechanism of a Purified Laminaria Japonica Polysaccharide on Adriamycin-Induced Acute Kidney Injury in Mice. Int. J. Biol. Macromol. 2020, 148, 591–600. [Google Scholar] [CrossRef] [PubMed]
  104. Wang, Y.; Sun, Y.; Shao, F.; Zhang, B.; Wang, Z.; Li, X. Low Molecular Weight Fucoidan Can Inhibit the Fibrosis of Diabetic Kidneys by Regulating the Kidney Lipid Metabolism. J. Diabetes Res. 2021, 2021, 7618166. [Google Scholar] [CrossRef]
  105. Chen, J.; Cui, W.; Zhang, Q.; Jia, Y.; Sun, Y.; Weng, L.; Luo, D.; Zhou, H.; Yang, B. Low Molecular Weight Fucoidan Ameliorates Diabetic Nephropathy Via Inhibiting Epithelial-Mesenchymal Transition and Fibrotic Processes. Am. J. Transl. Res. 2015, 7, 1553–1563. [Google Scholar]
  106. Li, X.-Y.; Chen, H.-R.; Kuang, D.-D.; Pan, L.-H.; Li, Q.-M.; Luo, J.-P.; Zha, X.-Q. Laminaria Japonica Polysaccharide Attenuates Podocyte Epithelial-Mesenchymal Transformation Via Tgf-Β1-Mediated Smad3 and P38mapk Pathways. Int. J. Biol. Macromol. 2023, 241, 124637. [Google Scholar] [CrossRef]
  107. Wang, J.; Zhang, Q.; Li, S.; Chen, Z.; Tan, J.; Yao, J.; Duan, D. Low Molecular Weight Fucoidan Alleviates Diabetic Nephropathy by Binding Fibronectin and Inhibiting Ecm-Receptor Interaction in Human Renal Mesangial Cells. Int. J. Biol. Macromol. 2020, 150, 304–314. [Google Scholar] [CrossRef] [PubMed]
  108. Li, X.; Li, X.; Zhang, Q.; Zhao, T. Low Molecular Weight Fucoidan and Its Fractions Inhibit Renal Epithelial Mesenchymal Transition Induced by Tgf-Β1 or Fgf-2. Int. J. Biol. Macromol. 2017, 105, 1482–1490. [Google Scholar] [CrossRef] [PubMed]
  109. Wei, X.-M.; Jiang, S.; Li, S.-S.; Sun, Y.-S.; Wang, S.-H.; Liu, W.-C.; Wang, Z.; Wang, Y.-P.; Zhang, R.; Li, W. Endoplasmic Reticulum Stress-Activated Perk-Eif2α-Atf4 Signaling Pathway Is Involved in the Ameliorative Effects of Ginseng Polysaccharides against Cisplatin-Induced Nephrotoxicity in Mice. ACS Omega 2021, 6, 8958–8966. [Google Scholar] [CrossRef] [PubMed]
  110. Lian, Y.; Zhu, M.; Chen, J.; Yang, B.; Lv, Q.; Wang, L.; Guo, S.; Tan, X.; Li, C.; Bu, W.; et al. Characterization of a Novel Polysaccharide from Moutan Cortex and Its Ameliorative Effect on Ages-Induced Diabetic Nephropathy. Int. J. Biol. Macromol. 2021, 176, 589–600. [Google Scholar] [CrossRef]
  111. Zhang, M.; Yang, L.; Zhu, M.; Yang, B.; Yang, Y.; Jia, X.; Feng, L. Moutan Cortex Polysaccharide Ameliorates Diabetic Kidney Disease Via Modulating Gut Microbiota Dynamically in Rats. Int. J. Biol. Macromol. 2022, 206, 849–860. [Google Scholar] [CrossRef]
  112. Zhao, H.; Xu, J.; Wang, R.; Tang, W.; Kong, L.; Wang, W.; Wang, L.; Zhang, Y.; Ma, W. Plantaginis Semen Polysaccharides Ameliorate Renal Damage through Regulating Nlrp3 Inflammasome in Gouty Nephropathy Rats. Food Funct. 2021, 12, 2543–2553. [Google Scholar] [CrossRef]
  113. Huang, C.; Yu, J.; Da, J.; Dong, R.; Dai, L.; Yang, Y.; Deng, Y.; Yuan, J. Dendrobium Officinale Kimura & Migo Polysaccharide Inhibits Hyperglycaemia-Induced Kidney Fibrosis Via the Mirna-34a-5p/Sirt1 Signalling Pathway. J. Ethnopharmacol. 2023, 313, 116601. [Google Scholar]
  114. Wang, X.; Liu, W.; Jin, G.; Wu, Z.; Zhang, D.; Bao, Y.; Shi, W. Salvia Miltiorrhiza Polysaccharides Alleviates Florfenicol Induced Kidney Injury in Chicks Via Inhibiting Oxidative Stress and Apoptosis. Ecotoxicol. Environ. Saf. 2022, 233, 113339. [Google Scholar] [CrossRef]
  115. Zhang, J.; Bi, R.; Meng, Q.; Wang, C.; Huo, X.; Liu, Z.; Wang, C.; Sun, P.; Sun, H.; Ma, X.; et al. Catalpol Alleviates Adriamycin-Induced Nephropathy by Activating the Sirt1 Signalling Pathway In Vivo and In Vitro. Br. J. Pharmacol. 2019, 176, 4558–4573. [Google Scholar] [CrossRef]
  116. Cong, C.; Yuan, X.; Hu, Y.; Chen, W.; Wang, Y.; Tao, L. Catalpol Alleviates Ang Ii-Induced Renal Injury through Nf-Κb Pathway and Tgf-Β1/Smads Pathway. J. Cardiovasc. Pharmacol. 2022, 79, e116–e121. [Google Scholar] [CrossRef]
  117. Chen, Y.; Liu, Q.; Meng, X.; Zhao, L.; Zheng, X.; Feng, W. Catalpol Ameliorates Fructose-Induced Renal Inflammation by Inhibiting Tlr4/Myd88 Signaling and Uric Acid Reabsorption. Eur. J. Pharmacol. 2024, 967, 176356. [Google Scholar] [CrossRef] [PubMed]
  118. Zaaba, N.E.; Al-Salam, S.; Beegam, S.; Elzaki, O.; Yasin, J.; Nemmar, A. Catalpol Attenuates Oxidative Stress and Inflammation Via Mechanisms Involving Sirtuin-1 Activation and Nf-Κb Inhibition in Experimentally-Induced Chronic Kidney Disease. Nutrients 2023, 15, 237. [Google Scholar] [CrossRef] [PubMed]
  119. Zhang, J.; Zhao, T.; Wang, C.; Meng, Q.; Huo, X.; Wang, C.; Sun, P.; Sun, H.; Ma, X.; Wu, J.; et al. Catalpol-Induced Ampk Activation Alleviates Cisplatin-Induced Nephrotoxicity through the Mitochondrial-Dependent Pathway without Compromising Its Anticancer Properties. Oxid. Med. Cell Longev. 2021, 2021, 7467156. [Google Scholar] [CrossRef] [PubMed]
  120. Chen, J.; Yang, Y.; Lv, Z.; Shu, A.; Du, Q.; Wang, W.; Chen, Y.; Xu, H. Study on the Inhibitive Effect of Catalpol on Diabetic Nephropathy. Life Sci. 2020, 257, 118120. [Google Scholar] [CrossRef]
  121. Shu, A.; Du, Q.; Chen, J.; Gao, Y.; Zhu, Y.; Lv, G.; Lu, J.; Chen, Y.; Xu, H. Catalpol Ameliorates Endothelial Dysfunction and Inflammation in Diabetic Nephropathy Via Suppression of Rage/Rhoa/Rock Signaling Pathway. Chem.-Biol. Interact. 2021, 348, 109625. [Google Scholar] [CrossRef]
  122. Zhang, J.; Wang, C.; Kang, K.; Liu, H.; Liu, X.; Jia, X.; Yu, K. Loganin Attenuates Septic Acute Renal Injury with the Participation of Akt and Nrf2/Ho-1 Signaling Pathways. Drug Des. Devel Ther. 2021, 15, 501–513. [Google Scholar] [CrossRef]
  123. Huang, F.; Wang, X.; Xiao, G.; Xiao, J. Loganin Exerts a Protective Effect on Ischemia-Reperfusion-Induced Acute Kidney Injury by Regulating Jak2/Stat3 and Nrf2/Ho-1 Signaling Pathways. Drug Dev. Res. 2022, 83, 150–157. [Google Scholar] [CrossRef]
  124. Kong, X.; Zhao, Y.; Wang, X.; Yu, Y.; Meng, Y.; Yan, G.; Yu, M.; Jiang, L.; Song, W.; Wang, B.; et al. Loganin Reduces Diabetic Kidney Injury by Inhibiting the Activation of Nlrp3 Inflammasome-Mediated Pyroptosis. Chem.-Biol. Interact. 2023, 382, 110640. [Google Scholar] [CrossRef]
  125. Liu, K.; Xu, H.; Lv, G.; Liu, B.; Lee, M.K.K.; Lu, C.; Lv, X.; Wu, Y. Loganin Attenuates Diabetic Nephropathy in C57bl/6j Mice with Diabetes Induced by Streptozotocin and Fed with Diets Containing High Level of Advanced Glycation End Products. Life Sci. 2015, 123, 78–85. [Google Scholar] [CrossRef]
  126. Li, X.; Ma, A.; Liu, K. Geniposide Alleviates Lipopolysaccharide-Caused Apoptosis of Murine Kidney Podocytes by Activating Ras/Raf/Mek/Erk-Mediated Cell Autophagy. Artif. Cells Nanomed. Biotechnol. 2019, 47, 1524–1532. [Google Scholar] [CrossRef]
  127. Liu, J.; Zhao, N.; Shi, G.; Wang, H. Geniposide Ameliorated Sepsis-Induced Acute Kidney Injury by Activating Pparγ. Aging 2020, 12, 22744–22758. [Google Scholar] [CrossRef] [PubMed]
  128. Liu, X.; Qian, N.; Zhu, L.; Fan, L.; Fu, G.; Ma, M.; Bao, J.; Cao, C.; Liang, X. Geniposide Ameliorates Acute Kidney Injury Via Enhancing the Phagocytic Ability of Macrophages Towards Neutrophil Extracellular Traps. Eur. J. Pharmacol. 2023, 957, 176018. [Google Scholar] [CrossRef] [PubMed]
  129. Cheng, W.; Tan, L.; Yu, S.; Song, J.; Li, Z.; Peng, X.; Wei, Q.; He, Z.; Zhang, W.; Yang, X. Geniposide Reduced Oxidative Stress-Induced Apoptosis in Hk-2 Cell through Pi3k/Akt3/Foxo1 by M6a Modification. Int. Immunopharmacol. 2024, 131, 111820. [Google Scholar] [CrossRef]
  130. Hu, X.; Zhang, X.; Jin, G.; Shi, Z.; Sun, W.; Chen, F. Geniposide Reduces Development of Streptozotocin-Induced Diabetic Nephropathy Via Regulating Nuclear Factor-Kappa B Signaling Pathways Geniposide Reduces Development of Streptozotocin-Induced Diabetic Nephropathy Via Regulating Nuclear Factor-Kappa B Signaling Pathways. Fundam. Clin. Pharmacol. 2017, 31, 54–63. [Google Scholar]
  131. Dusabimana, T.; Park, E.J.; Je, J.; Jeong, K.; Yun, S.P.; Kim, H.J.; Kim, H.; Park, S.W. Geniposide Improves Diabetic Nephropathy by Enhancing Ulk1-Mediated Autophagy and Reducing Oxidative Stress through Ampk Activation. Int. J. Mol. Sci. 2021, 22, 1651. [Google Scholar] [CrossRef]
  132. Peng, J.H.; Leng, J.; Tian, H.J.; Yang, T.; Fang, Y.; Feng, Q.; Zhao, Y.; Hu, Y.Y. Geniposide and Chlorogenic Acid Combination Ameliorates Non-Alcoholic Steatohepatitis Involving the Protection on the Gut Barrier Function in Mouse Induced by High-Fat Diet. Front. Pharmacol. 2018, 9, 1399. [Google Scholar] [CrossRef]
  133. Gao, X.; Liu, Y.; Wang, L.; Sai, N.; Liu, Y.; Ni, J. Morroniside Inhibits H2O2-Induced Podocyte Apoptosis by Down-Regulating Nox4 Expression Controlled by Autophagy in Vitro. Front. Pharmacol. 2020, 11, 533809. [Google Scholar] [CrossRef]
  134. Liu, Y.; Dai, W.; Ye, S. The Olive Constituent Oleuropein Exerts Nephritic Protective Effects on Diabetic Nephropathy in Db/Db Mice. Arch. Physiol. Biochem. 2019, 128, 455–462. [Google Scholar] [CrossRef] [PubMed]
  135. Tüfekci, K.K.; Tatar, M. Oleuropein Mitigates Acrylamide-Induced Nephrotoxicity by Affecting Placental Growth Factor Immunoactivity in the Rat Kidney. Eurasian J. Med. 2023, 55, 228–233. [Google Scholar]
  136. Chen, L.; Cao, G.; Wang, M.; Feng, Y.L.; Chen, D.Q.; Vaziri, N.D.; Zhuang, S.; Zhao, Y.Y. The Matrix Metalloproteinase-13 Inhibitor Poricoic Acid Zi Ameliorates Renal Fibrosis by Mitigating Epithelial-Mesenchymal Transition. Mol. Nutr. Food Res. 2019, 63, e1900132. [Google Scholar] [CrossRef]
  137. Wang, M.; Chen, D.Q.; Chen, L.; Cao, G.; Zhao, H.; Liu, D.; Vaziri, N.D.; Guo, Y.; Zhao, Y.Y. Novel Inhibitors of the Cellular Renin-Angiotensin System Components, Poricoic Acids, Target Smad3 Phosphorylation and Wnt/Β-Catenin Pathway against Renal Fibrosis. Br. J. Pharmacol. 2018, 175, 2689–2708. [Google Scholar] [CrossRef] [PubMed]
  138. Chen, D.Q.; Wang, Y.N.; Vaziri, N.D.; Chen, L.; Hu, H.H.; Zhao, Y.Y. Poricoic Acid a Activates Ampk to Attenuate Fibroblast Activation and Abnormal Extracellular Matrix Remodelling in Renal Fibrosis. Phytomedicine 2020, 72, 153232. [Google Scholar] [CrossRef] [PubMed]
  139. Chen, D.Q.; Wu, X.Q.; Chen, L.; Hu, H.H.; Wang, Y.N.; Zhao, Y.Y. Poricoic Acid a as a Modulator of Tph-1 Expression Inhibits Renal Fibrosis Via Modulating Protein Stability of Β-Catenin and Β-Catenin-Mediated Transcription. Ther. Adv. Chronic Dis. 2020, 11, 2040622320962648. [Google Scholar] [CrossRef]
  140. Chen, D.Q.; Feng, Y.L.; Chen, L.; Liu, J.R.; Wang, M.; Vaziri, N.D.; Zhao, Y.Y. Poricoic Acid a Enhances Melatonin Inhibition of Aki-to-Ckd Transition by Regulating Gas6/Axlnfκb/Nrf2 Axis. Free Radic. Biol. Med. 2019, 134, 484–497. [Google Scholar] [CrossRef] [PubMed]
  141. Chen, D.Q.; Chen, L.; Guo, Y.; Wu, X.Q.; Zhao, T.T.; Zhao, H.L.; Zhang, H.J.; Yan, M.H.; Zhang, G.Q.; Li, P. Poricoic Acid a Suppresses Renal Fibroblast Activation and Interstitial Fibrosis in Uuo Rats Via Upregulating Sirt3 and Promoting Β-Catenin K49 Deacetylation. Acta Pharmacol. Sin. 2023, 44, 1038–1050. [Google Scholar] [CrossRef]
  142. Li, Q.; Ming, Y.; Jia, H.; Wang, G. Poricoic Acid a Suppresses Tgf-Β1-Induced Renal Fibrosis and Proliferation Via the Pdgf-C, Smad3 and Mapk Pathways. Exp. Ther. Med. 2021, 21, 289. [Google Scholar] [CrossRef]
  143. Chung, S.; Yoon, H.E.; Kim, S.J.; Kim, S.J.; Koh, E.S.; Hong, Y.A.; Park, C.W.; Chang, Y.S.; Shin, S.J. Oleanolic Acid Attenuates Renal Fibrosis in Mice with Unilateral Ureteral Obstruction Via Facilitating Nuclear Translocation of Nrf2. Nutr. Metab. 2014, 11, 2. [Google Scholar] [CrossRef] [PubMed]
  144. Nataraju, A.; Saini, D.; Ramachandran, S.; Benshoff, N.; Liu, W.; Chapman, W.; Mohanakuma, T.r. Oleanolic Acid, a Plant Triterpenoid, Significantly Improves Survival and Function of Islet Allograft. Transplantation 2009, 88, 987–994. [Google Scholar] [CrossRef]
  145. Alqrad, M.A.I.; El-Agamy, D.S.; Ibrahim, S.R.M.; Sirwi, A.; Abdallah, H.M.; Abdel-Sattar, E.; El-Halawany, A.M.; Elsaed, W.M.; Mohamed, G.A. Sirt1/Nrf2/Nf-Κb Signaling Mediates Anti-Inflammatory and Anti-Apoptotic Activities of Oleanolic Acid in a Mouse Model of Acute Hepatorenal Damage. Medicina 2023, 59, 1351. [Google Scholar] [CrossRef]
  146. Liu, Y.; Zheng, J.Y.; Wei, Z.T.; Liu, S.K.; Sun, J.L.; Mao, Y.H.; Xu, Y.D.; Yang, Y. Therapeutic Effect and Mechanism of Combination Therapy with Ursolic Acid and Insulin on Diabetic Nephropathy in a Type I Diabetic Rat Model. Front. Pharmacol. 2022, 13, 969207. [Google Scholar] [CrossRef]
  147. Wang, K.; Xu, X.; Shan, Q.; Ding, R.; Lyu, Q.; Huang, L.; Chen, X.; Han, X.; Yang, Q.; Sang, X.; et al. Integrated Gut Microbiota and Serum Metabolomics Reveal the Protective Effect of Oleanolic Acid on Liver and Kidney-Injured Rats Induced by Euphorbia Pekinensis. Phytother. Res. 2022. [Google Scholar] [CrossRef] [PubMed]
  148. Zhang, J.; Luan, Z.L.; Huo, X.K.; Zhang, M.; Morisseau, C.; Sun, C.P.; Hammock, B.D.; Ma, X.C. Direct Targeting of Seh with Alisol B Alleviated the Apoptosis, Inflammation, and Oxidative Stress in Cisplatin-Induced Acute Kidney Injury. Int. J. Biol. Sci. 2023, 19, 294–310. [Google Scholar] [CrossRef] [PubMed]
  149. Luan, Z.L.; Ming, W.H.; Sun, X.W.; Zhang, C.; Zhou, Y.; Zheng, F.; Yang, Y.L.; Guan, Y.F.; Zhang, X.Y. A Naturally Occurring Fxr Agonist, Alisol B 23-Acetate, Protects against Renal Ischemia-Reperfusion Injury. Am. J. Physiol. Renal Physiol. 2021, 321, F617–F628. [Google Scholar] [CrossRef] [PubMed]
  150. Chen, H.; Wang, M.C.; Chen, Y.Y.; Chen, L.; Wang, Y.N.; Vaziri, N.D.; Miao, H.; Zhao, Y.Y. Alisol B 23-Acetate Attenuates Ckd Progression by Regulating the Renin-Angiotensin System and Gut-Kidney Axis. Ther. Adv. Chronic Dis. 2020, 11, 2040622320920025. [Google Scholar] [CrossRef] [PubMed]
  151. Wang, L.; Hao, X.; Li, X.; Li, Q.; Fang, X. Effects of Ginsenoside Rh2 on Cisplatin-Induced Nephrotoxicity in Renal Tubular Epithelial Cells by Inhibiting Endoplasmic Reticulum Stress. J. Biochem. Mol. Toxicol. 2024, 38, e23768. [Google Scholar] [CrossRef]
  152. Guo, J.; Chen, L.; Ma, M. Ginsenoside Rg1 Suppresses Ferroptosis of Renal Tubular Epithelial Cells in Sepsis-Induced Acute Kidney Injury Via the Fsp1-Coq(10)-Nad(P)H Pathway. Curr. Med. Chem. 2024, 31, 2119–2132. [Google Scholar] [CrossRef]
  153. Zhao, H.; Ding, R.; Han, J. Ginsenoside Rh4 Facilitates the Sensitivity of Renal Cell Carcinoma to Ferroptosis Via the Nrf2 Pathway. Arch. Esp. Urol. 2024, 77, 119–128. [Google Scholar] [CrossRef]
  154. Hwang, H.J.; Hong, S.H.; Moon, H.S.; Yoon, Y.E.; Park, S.Y. Ginsenoside Rh2 Sensitizes the Anti-Cancer Effects of Sunitinib by Inducing Cell Cycle Arrest in Renal Cell Carcinoma. Sci. Rep. 2022, 12, 19752. [Google Scholar] [CrossRef]
  155. Chen, Y.; Peng, Y.; Li, P.; Jiang, Y.; Song, D. Ginsenoside Rg3 Induces Mesangial Cells Proliferation and Attenuates Apoptosis by Mir-216a-5p/Mapk Pathway in Diabetic Kidney Disease. Aging 2024, 16, 9933–9943. [Google Scholar] [CrossRef]
  156. Sui, Z.; Sui, D.; Li, M.; Yu, Q.; Li, H.; Jiang, Y. Ginsenoside Rg3 Has Effects Comparable to Those of Ginsenoside Re on Diabetic Kidney Disease Prevention in Db/Db Mice by Regulating Inflammation, Fibrosis and Pparγ. Mol. Med. Rep. 2023, 27, 84. [Google Scholar] [CrossRef]
  157. Liu, Y.; Mou, L.; Yi, Z.; Lin, Q.; Banu, K.; Wei, C.; Yu, X. Integrative Informatics Analysis Identifies That Ginsenoside Re Improves Renal Fibrosis through Regulation of Autophagy. J. Nat. Med. 2024, 78, 722–731. [Google Scholar] [CrossRef] [PubMed]
  158. Ji, P.; Shi, Q.; Liu, Y.; Han, M.; Su, Y.; Sun, R.; Zhou, H.; Li, W.; Li, W. Ginsenoside Rg1 Treatment Alleviates Renal Fibrosis by Inhibiting the Nox4-Mapk Pathway in T2dm Mice. Ren. Fail. 2023, 45, 2197075. [Google Scholar] [CrossRef] [PubMed]
  159. Su, L.; Liu, L.; Jia, Y.; Lei, L.; Liu, J.; Zhu, S.; Zhou, H.; Chen, R.; Lu, H.A.J.; Yang, B. Ganoderma Triterpenes Retard Renal Cyst Development by Downregulating Ras/Mapk Signaling and Promoting Cell Differentiation. Kidney Int. 2017, 92, 1404–1418. [Google Scholar] [CrossRef]
  160. Geng, X.Q.; Ma, A.; He, J.Z.; Wang, L.; Jia, Y.L.; Shao, G.Y.; Li, M.; Zhou, H.; Lin, S.Q.; Ran, J.H.; et al. Ganoderic Acid Hinders Renal Fibrosis Via Suppressing the Tgf-Β/Smad and Mapk Signaling Pathways. Acta Pharmacol. Sin. 2020, 41, 670–677. [Google Scholar] [CrossRef]
  161. Shao, G.; He, J.; Meng, J.; Ma, A.; Geng, X.; Zhang, S.; Qiu, Z.; Lin, D.; Li, M.; Zhou, H.; et al. Ganoderic Acids Prevent Renal Ischemia Reperfusion Injury by Inhibiting Inflammation and Apoptosis. Int. J. Mol. Sci. 2021, 22, 10229. [Google Scholar] [CrossRef]
  162. Cai, L.; Horowitz, M.; Islam, M.S. Potential Therapeutic Targets for the Prevention of Diabetic Nephropathy: Glycyrrhetinic Acid. World J. Diabetes 2023, 14, 1717–1720. [Google Scholar] [CrossRef]
  163. Hou, S.; Zhang, T.; Li, Y.; Guo, F.; Jin, X. Glycyrrhizic Acid Prevents Diabetic Nephropathy by Activating Ampk/Sirt1/Pgc-1α Signaling in Db/Db Mice. J. Diabetes Res. 2017, 2017, 2865912. [Google Scholar] [CrossRef] [PubMed]
  164. Cao, R.; Li, Y.; Hu, X.; Qiu, Y.; Li, S.; Xie, Y.; Xu, C.; Lu, C.; Chen, G.; Yang, J. Glycyrrhizic Acid Improves Tacrolimus-Induced Renal Injury by Regulating Autophagy. Faseb J. 2023, 37, e22749. [Google Scholar] [CrossRef] [PubMed]
  165. Oh, H.; Choi, A.; Seo, N.; Lim, J.S.; You, J.S.; Chung, Y.E. Protective Effect of Glycyrrhizin, a Direct Hmgb1 Inhibitor, on Post-Contrast Acute Kidney Injury. Sci. Rep. 2021, 11, 15625. [Google Scholar] [CrossRef]
  166. Li, Z.; Yan, M.; Cao, L.; Fang, P.; Guo, Z.; Hou, Z.; Zhang, B. Glycyrrhetinic Acid Accelerates the Clearance of Triptolide through P-Gp in Vitro. Phytother. Res. 2017, 31, 1090–1096. [Google Scholar] [CrossRef]
  167. Jiang, Y.; Cai, C.; Zhang, P.; Luo, Y.; Guo, J.; Li, J.; Rong, R.; Zhang, Y.; Zhu, T. Transcriptional Profile Changes after Treatment of Ischemia Reperfusion Injury-Induced Kidney Fibrosis with 18β-Glycyrrhetinic Acid. Ren. Fail. 2022, 44, 660–671. [Google Scholar] [CrossRef] [PubMed]
  168. Zhu, Z.; Li, J.; Song, Z.; Li, T.; Li, Z.; Gong, X. Tetramethylpyrazine Attenuates Renal Tubular Epithelial Cell Ferroptosis in Contrast-Induced Nephropathy by Inhibiting Transferrin Receptor and Intracellular Reactive Oxygen Species. Clin. Sci. 2024, 138, 235–249. [Google Scholar] [CrossRef] [PubMed]
  169. Gong, X.; Duan, Y.; Zheng, J.; Ye, Z.; Hei, T.K. Tetramethylpyrazine Prevents Contrast-Induced Nephropathy Via Modulating Tubular Cell Mitophagy and Suppressing Mitochondrial Fragmentation, Ccl2/Ccr2-Mediated Inflammation, and Intestinal Injury. Oxid. Med. Cell Longev. 2019, 2019, 7096912. [Google Scholar] [CrossRef] [PubMed]
  170. Sun, W.; Li, A.; Wang, Z.; Sun, X.; Dong, M.; Qi, F.; Wang, L.; Zhang, Y.; Du, P. Tetramethylpyrazine Alleviates Acute Kidney Injury by Inhibiting Nlrp3/Hif-1α and Apoptosis. Mol. Med. Rep. 2020, 22, 2655–2664. [Google Scholar] [CrossRef]
  171. Rai, U.; Kosuru, R.; Prakash, S.; Tiwari, V.; Singh, S. Tetramethylpyrazine Alleviates Diabetic Nephropathy through the Activation of Akt Signalling Pathway in Rats. Eur. J. Pharmacol. 2019, 865, 172763. [Google Scholar] [CrossRef]
  172. Jing, M.; Cen, Y.; Gao, F.; Wang, T.; Jiang, J.; Jian, Q.; Wu, L.; Guo, B.; Luo, F.; Zhang, G.; et al. Nephroprotective Effects of Tetramethylpyrazine Nitrone Tbn in Diabetic Kidney Disease. Front. Pharmacol. 2021, 12, 680336. [Google Scholar] [CrossRef]
  173. Hu, J.; Gu, W.; Ma, N.; Fan, X.; Ci, X. Leonurine Alleviates Ferroptosis in Cisplatin-Induced Acute Kidney Injury by Activating the Nrf2 Signalling Pathway. Br. J. Pharmacol. 2022, 179, 3991–4009. [Google Scholar] [CrossRef]
  174. Yin, X.; Gao, Q.; Li, C.; Yang, Q.; Dong, H.; Li, Z. Leonurine Alleviates Vancomycin Nephrotoxicity Via Activating Pparγ and Inhibiting the Tlr4/Nf-Κb/Tnf-A Pathway. Int. Immunopharmacol. 2024, 131, 111898. [Google Scholar] [CrossRef]
  175. Cheng, H.; Bo, Y.; Shen, W.; Tan, J.; Jia, Z.; Xu, C.; Li, F. Leonurine Ameliorates Kidney Fibrosis Via Suppressing Tgf-Β and Nf-Κb Signaling Pathway in Uuo Mice. Int. Immunopharmacol. 2015, 25, 406–415. [Google Scholar] [CrossRef]
  176. Xu, D.; Chen, M.; Ren, X.; Ren, X.; Wu, Y. Leonurine Ameliorates Lps-Induced Acute Kidney Injury Via Suppressing Ros-Mediated Nf-Κb Signaling Pathway. Fitoterapia 2014, 97, 148–155. [Google Scholar] [CrossRef]
  177. Cheng, R.; Wang, X.; Huang, L.; Lu, Z.; Wu, A.; Guo, S.; Li, C.; Mao, W.; Xie, Y.; Xu, P.; et al. Novel Insights into the Protective Effects of Leonurine against Acute Kidney Injury: Inhibition of Er Stress-Associated Ferroptosis Via Regulating Atf4/Chop/Acsl4 Pathway. Chem. Biol. Interact. 2024, 395, 111016. [Google Scholar] [CrossRef] [PubMed]
  178. Hassanein, E.H.M.; Shalkami, A.-G.S.; Khalaf, M.M.; Mohamed, W.R.; Hemeida, R.A.M. The Impact of Keap1/Nrf2, P38Mapk/Nf-Κb and Bax/Bcl2/Caspase-3 Signaling Pathways in the Protective Effects of Berberine against Methotrexate-Induced Nephrotoxicity. Biomed. Pharmacother. 2019, 109, 47–56. [Google Scholar] [CrossRef]
  179. Fouad, G.I.; Ahmed, K.A. The Protective Impact of Berberine against Doxorubicin-Induced Nephrotoxicity in Rats. Tissue Cell 2021, 73, 101612. [Google Scholar] [CrossRef]
  180. Malaviya, A.N. Landmark Papers on the Discovery of Methotrexate for the Treatment of Rheumatoid Arthritis and Other Systemic Inflammatory Rheumatic Diseases: A Fascinating Story. Int. J. Rheum. Dis. 2016, 19, 844–851. [Google Scholar] [CrossRef] [PubMed]
  181. Domitrovic, R.; Cvijanovic, O.; Pernjak-Pugel, E.; Skoda, M.; Mikelic, L.; Crncevic-Orlic, Z. Berberine Exerts Nephroprotective Effect against Cisplatin-Induced Kidney Damage through Inhibition of Oxidative/Nitrosative Stress, Inflammation, Autophagy and Apoptosis. Food Chem. Toxicol. 2013, 62, 397–406. [Google Scholar] [CrossRef]
  182. Adil, M.; Kandhare, A.D.; Dalvi, G.; Ghosh, P.; Venkata, S.; Raygude, K.S.; Bodhankar, S.L. Ameliorative Effect of Berberine against Gentamicin-Induced Nephrotoxicity in Rats Via Attenuation of Oxidative Stress, Inflammation, Apoptosis and Mitochondrial Dysfunction. Ren. Fail. 2016, 38, 996–1006. [Google Scholar] [CrossRef] [PubMed]
  183. Kumas, M.; Esrefoglu, M.; Karatas, E.; Duymac, N.; Kanbay, S.; Ergun, I.S.; Uyuklu, M.; Kocyigit, A. Investigation of Dose-Dependent Effects of Berberine against Renal Ischemia/Reperfusion Injury in Experimental Diabetic Rats. Nefrologia 2019, 39, 411–423. [Google Scholar] [CrossRef]
  184. Lu, J.; Yi, Y.; Pan, R.; Zhang, C.; Han, H.; Chen, J.; Liu, W. Berberine Protects Hk-2 Cells from Hypoxia/Reoxygenation Induced Apoptosis Via Inhibiting Sphk1 Expression. J. Nat. Med. 2018, 72, 390–398. [Google Scholar] [CrossRef]
  185. Visnagri, A.; Kandhare, A.D.; Bodhankar, S.L. Renoprotective Effect of Berberine Via Intonation on Apoptosis and Mitochondrial-Dependent Pathway in Renal Ischemia Reperfusion-Induced Mutilation. Ren. Fail. 2015, 37, 482–493. [Google Scholar] [CrossRef]
  186. Zhang, X.; He, H.; Liang, D.; Jiang, Y.; Liang, W.; Chi, Z.-H.; Ma, J. Protective Effects of Berberine on Renal Injury in Streptozotocin (Stz)-Induced Diabetic Mice. Int. J. Mol. Sci. 2016, 17, 1327. [Google Scholar] [CrossRef]
  187. Yang, G.; Zhao, Z.; Zhang, X.; Wu, A.; Huang, Y.; Miao, Y.; Yang, M. Effect of Berberine on the Renal Tubular Epithelial-to- Mesenchymal Transition by Inhibition of the Notch/Snail Pathway in Diabetic Nephropathy Model Kkay Mice. Drug Des. Dev. Ther. 2017, 11, 1065–1079. [Google Scholar] [CrossRef] [PubMed]
  188. Zhang, X.; Guan, T.; Yang, B.; Chi, Z.; Wan, Q.; Gu, H.F. Protective Effect of Berberine on High Glucose and Hypoxia-Induced Apoptosis Via the Modulation of Hif-1α in Renal Tubular Epithelial Cells. Am. J. Transl. Res. 2019, 11, 669–682. [Google Scholar] [PubMed]
  189. Wang, F.-M.; Yang, Y.-J.; Ma, L.-L.; Tian, X.-J.; He, Y.-Q. Berberine Ameliorates Renal Interstitial Fibrosis Induced by Unilateral Ureteral Obstruction in Rats. Nephrology 2014, 19, 542–551. [Google Scholar] [CrossRef] [PubMed]
  190. Pan, L.; Yu, H.; Fu, J.; Hu, J.; Xu, H.; Zhang, Z.; Bu, M.; Yang, X.; Zhang, H.; Lu, J.; et al. Berberine Ameliorates Chronic Kidney Disease through Inhibiting the Production of Gut-Derived Uremic Toxins in the Gut Microbiota. Acta Pharm. Sin. B 2023, 13, 1537–1553. [Google Scholar] [CrossRef]
  191. Peerapen, P.; Thongboonkerd, V. Protective Roles of Trigonelline against Oxalate-Induced Epithelial-to-Mesenchymal Transition in Renal Tubular Epithelial Cells: An in Vitro Study. Food Chem. Toxicol. 2020, 135, 110915. [Google Scholar] [CrossRef]
  192. Peerapen, P.; Boonmark, W.; Putpeerawit, P.; Sassanarakkit, S.; Thongboonkerd, V. Proteomic and Computational Analyses Followed by Functional Validation of Protective Effects of Trigonelline against Calcium Oxalate-Induced Renal Cell Deteriorations. Comput. Struct. Biotechnol. J. 2023, 21, 5851–5867. [Google Scholar] [CrossRef]
  193. Sheweita, S.A.; ElHady, S.A.; Hammoda, H.M. Trigonella Stellata Reduced the Deleterious Effects of Diabetes Mellitus through Alleviation of Oxidative Stress, Antioxidant- and Drug-Metabolizing Enzymes Activities. J. Ethnopharmacol. 2020, 256, 112821. [Google Scholar] [CrossRef]
  194. Chen, C.; Ma, J.; Miao, C.S.; Zhang, H.; Zhang, M.; Cao, X.; Shi, Y. Trigonelline Induces Autophagy to Protect Mesangial Cells in Response to High. Glucose Via Activating the Mir-5189-5p-Ampk Pathway. Phytomedicine 2021, 92, 153614. [Google Scholar] [CrossRef]
  195. Gong, M.; Guo, Y.; Dong, H.; Wu, W.; Wu, F.; Lu, F. Trigonelline Inhibits Tubular Epithelial-Mesenchymal Transformation in Diabetic Kidney Disease Via Targeting Smad7. Biomed. Pharmacother. 2023, 168, 115747. [Google Scholar] [CrossRef]
  196. Peerapen, P.; Boonmark, W.; Thongboonkerd, V. Trigonelline Prevents Kidney Stone Formation Processes by Inhibiting Calcium Oxalate Crystallization, Growth and Crystal-Cell Adhesion, and Downregulating Crystal Receptors. Biomed. Pharmacother. 2022, 149, 112876. [Google Scholar] [CrossRef]
  197. Zhou, L.; Wu, K.; Gao, Y.; Qiao, R.; Tang, N.; Dong, D.; Li, X.Q.; Nong, Q.; Luo, D.Q.; Xiao, Q.; et al. Piperlonguminine Attenuates Renal Fibrosis by Inhibiting Trpc6. J. Ethnopharmacol. 2023, 313, 116561. [Google Scholar] [CrossRef] [PubMed]
  198. Yuan, L.; Yang, J.; Li, Y.; Yuan, L.; Liu, F.; Yuan, Y.; Tang, X. Matrine Alleviates Cisplatin-Induced Acute Kidney Injury by Inhibiting Mitochondrial Dysfunction and Inflammation Via Sirt3/Opa1 Pathway. J. Cell Mol. Med. 2022, 26, 3702–3715. [Google Scholar] [CrossRef] [PubMed]
  199. Kang, S.; Chen, T.; Hao, Z.; Yang, X.; Wang, M.; Zhang, Z.; Hao, S.; Lang, F.; Hao, H. Oxymatrine Alleviates Gentamicin-Induced Renal Injury in Rats. Molecules 2022, 27, 6209. [Google Scholar] [CrossRef] [PubMed]
  200. Jiang, G.; Liu, X.; Wang, M.; Chen, H.; Chen, Z.; Qiu, T. Oxymatrine Ameliorates Renal Ischemia-Reperfusion Injury from Oxidative Stress through Nrf2/Ho-1 Pathway. Acta Cir. Bras. 2015, 30, 422–429. [Google Scholar] [CrossRef]
  201. Yao, M.; Lian, D.; Wu, M.; Zhou, Y.; Fang, Y.; Zhang, S.; Zhang, W.; Yang, Y.; Li, R.; Chen, H.; et al. Isoliensinine Attenuates Renal Fibrosis and Inhibits Tgf-Β1/Smad2/3 Signaling Pathway in Spontaneously Hypertensive Rats. Drug Des. Devel Ther. 2023, 17, 2749–2762. [Google Scholar] [CrossRef]
  202. Zhang, W.; Chen, H.; Xu, Z.; Zhang, X.; Tan, X.; He, N.; Shen, J.; Dong, J. Liensinine Pretreatment Reduces Inflammation, Oxidative Stress, Apoptosis, and Autophagy to Alleviate Sepsis Acute Kidney Injury. Int. Immunopharmacol. 2023, 122, 110563. [Google Scholar] [CrossRef]
  203. Xiong, Y.; Zhong, J.; Chen, W.; Li, X.; Liu, H.; Li, Y.; Xiong, W.; Li, H. Neferine Alleviates Acute Kidney Injury by Regulating the Ppar-A/Nf-Κb Pathway. Clin. Exp. Nephrol. 2024, 1–19. [Google Scholar] [CrossRef]
  204. Li, H.; Chen, W.; Chen, Y.; Zhou, Q.; Xiao, P.; Tang, R.; Xue, J. Neferine Attenuates Acute Kidney Injury by Inhibiting Nf-Κb Signaling and Upregulating Klotho Expression. Front. Pharmacol. 2019, 10, 1197. [Google Scholar] [CrossRef]
  205. Hongmei, H.; Maojun, Y.; Ting, L.I.; Dandan, W.; Ying, L.I.; Xiaochi, T.; Lu, Y.; Shi, G.U.; Yong, X.U. Neferine Inhibits the Progression of Diabetic Nephropathy by Modulating the Mir-17-5p/Nuclear Factor E2-Related Factor 2 Axis. J. Tradit. Chin. Med. 2024, 44, 44–53. [Google Scholar]
  206. Li, H.; Ge, H.; Song, X.; Tan, X.; Xiong, Q.; Gong, Y.; Zhang, L.; He, Y.; Zhang, W.; Zhu, P.; et al. Neferine Mitigates Cisplatin-Induced Acute Kidney Injury in Mice by Regulating Autophagy and Apoptosis. Clin. Exp. Nephrol. 2023, 27, 122–131. [Google Scholar] [CrossRef]
  207. Li, H.; Tang, Y.; Wen, L.; Kong, X.; Chen, X.; Liu, P.; Zhou, Z.; Chen, W.; Xiao, C.; Xiao, P.; et al. Neferine Reduces Cisplatin-Induced Nephrotoxicity by Enhancing Autophagy Via the Ampk/Mtor Signaling Pathway. Biochem. Biophys. Res. Commun. 2017, 484, 694–701. [Google Scholar] [CrossRef]
  208. Yin, W.; Wang, J.H.; Liang, Y.M.; Liu, K.H.; Chen, Y.; Chen, Y. Neferine Targeted the Nlrc5/Nlrp3 Pathway to Inhibit M1-Type Polarization and Pyroptosis of Macrophages to Improve Hyperuricemic Nephropathy. Curr. Mol. Med. 2024; online ahead of print. [Google Scholar] [CrossRef]
  209. Wang, M.X.; Zhao, X.J.; Chen, T.Y.; Liu, Y.L.; Jiao, R.Q.; Zhang, J.H.; Ma, C.H.; Liu, J.H.; Pan, Y.; Kong, L.D. Nuciferine Alleviates Renal Injury by Inhibiting Inflammatory Responses in Fructose-Fed Rats. J. Agric. Food Chem. 2016, 64, 7899–7910. [Google Scholar] [CrossRef] [PubMed]
  210. Li, D.; Liu, B.; Fan, Y.; Liu, M.; Han, B.; Meng, Y.; Xu, X.; Song, Z.; Liu, X.; Hao, Q.; et al. Nuciferine Protects against Folic Acid-Induced Acute Kidney Injury by Inhibiting Ferroptosis. Br. J. Pharmacol. 2021, 178, 1182–1199. [Google Scholar] [CrossRef] [PubMed]
  211. Zhai, J.; Chen, Z.; Zhu, Q.; Guo, Z.; Sun, X.; Jiang, L.; Li, J.; Wang, N.; Yao, X.; Zhang, C.; et al. Curcumin Inhibits Pat-Induced Renal Ferroptosis Via the P62/Keap1/Nrf2 Signalling Pathway. Toxicology 2024, 506, 153863. [Google Scholar] [CrossRef]
  212. Yang, H.; Zhang, H.; Tian, L.; Guo, P.; Liu, S.; Chen, H.; Sun, L. Curcumin Attenuates Lupus Nephritis by Inhibiting Neutrophil Migration Via Pi3k/Akt/Nf-Κb Signalling Pathway. Lupus Sci. Med. 2024, 11, e001220. [Google Scholar] [CrossRef] [PubMed]
  213. Ghasemzadeh Rahbardar, M.; Hosseinzadeh, H. The Ameliorative Effect of Turmeric (Curcuma longa Linn) Extract and Its Major Constituent, Curcumin, and Its Analogs on Ethanol Toxicity. Phytother. Res. 2024, 38, 2165–2181. [Google Scholar] [CrossRef]
  214. Zhang, H.; Dong, Q.Q.; Shu, H.P.; Tu, Y.C.; Liao, Q.Q.; Yao, L.J. Curcumin Ameliorates Focal Segmental Glomerulosclerosis by Inhibiting Apoptosis and Oxidative Stress in Podocytes. Arch. Biochem. Biophys. 2023, 746, 109728. [Google Scholar] [CrossRef]
  215. Altamimi, J.Z.; AlFaris, N.A.; Al-Farga, A.M.; Alshammari, G.M.; BinMowyna, M.N.; Yahya, M.A. Curcumin Reverses Diabetic Nephropathy in Streptozotocin-Induced Diabetes in Rats by Inhibition of Pkcβ/P(66)Shc Axis and Activation of Foxo-3a. J. Nutr. Biochem. 2021, 87, 108515. [Google Scholar] [CrossRef]
  216. Feng, L.; Lin, Z.; Tang, Z.; Zhu, L.; Xu, S.; Tan, X.; Wang, X.; Mai, J.; Tan, Q. Emodin Improves Renal Fibrosis in Chronic Kidney Disease by Regulating Mitochondrial Homeostasis through the Mediation of Peroxisome Proliferator-Activated Receptor-Gamma Coactivator-1 Alpha (Pgc-1α). Eur. J. Histochem. 2024, 68, 3917. [Google Scholar] [CrossRef]
  217. Dong, X.; Wen, R.; Xiong, Y.; Jia, X.; Zhang, X.; Li, X.; Zhang, L.; Li, Z.; Zhang, S.; Yu, Y.; et al. Emodin Alleviates Crs4-Induced Mitochondrial Damage Via Activation of the Pgc1α Signaling. Phytother. Res. 2024, 38, 1345–1357. [Google Scholar] [CrossRef]
  218. Wang, L.; Wang, X.; Li, G.; Zhou, S.; Wang, R.; Long, Q.; Wang, M.; Li, L.; Huang, H.; Ba, Y. Emodin Ameliorates Renal Injury and Fibrosis Via Regulating the Mir-490-3p/Hmga2 Axis. Front. Pharmacol. 2023, 14, 1042093. [Google Scholar] [CrossRef] [PubMed]
  219. Yang, F.; Deng, L.; Li, J.; Chen, M.; Liu, Y.; Hu, Y.; Zhong, W. Emodin Retarded Renal Fibrosis through Regulating Hgf and Tgfβ-Smad Signaling Pathway. Drug Des. Devel Ther. 2020, 14, 3567–3575. [Google Scholar] [CrossRef] [PubMed]
  220. Tian, N.; Gao, Y.; Wang, X.; Wu, X.; Zou, D.; Zhu, Z.; Han, Z.; Wang, T.; Shi, Y. Emodin Mitigates Podocytes Apoptosis Induced by Endoplasmic Reticulum Stress through the Inhibition of the Perk Pathway in Diabetic Nephropathy. Drug Des. Devel Ther. 2018, 12, 2195–2211. [Google Scholar] [CrossRef] [PubMed]
  221. Kuang, B.C.; Wang, Z.H.; Hou, S.H.; Zhang, J.; Wang, M.Q.; Zhang, J.S.; Sun, K.L.; Ni, H.Q.; Gong, N.Q. Methyl Eugenol Protects the Kidney from Oxidative Damage in Mice by Blocking the Nrf2 Nuclear Export Signal through Activation of the Ampk/Gsk3β Axis. Acta Pharmacol. Sin. 2023, 44, 367–380. [Google Scholar] [CrossRef] [PubMed]
  222. Fathy, M.; Abdel-Latif, R.; Abdelgwad, Y.M.; Othman, O.A.; Abdel-Razik, A.H.; Dandekar, T.; Othman, E.M. Nephroprotective Potential of Eugenol in a Rat Experimental Model of Chronic Kidney Injury; Targeting Nox, Tgf-Β, and Akt Signaling. Life Sci. 2022, 308, 120957. [Google Scholar] [CrossRef]
  223. Oikawa, D.; Yamashita, S.; Takahashi, S.; Waki, T.; Kikuchi, K.; Abe, T.; Katayama, T.; Nakayama, T. (+)-Sesamin, a Sesame Lignan, Is a Potent Inhibitor of Gut Bacterial Tryptophan Indole-Lyase That Is a Key Enzyme in Chronic Kidney Disease Pathogenesis. Biochem. Biophys. Res. Commun. 2022, 590, 158–162. [Google Scholar] [CrossRef]
  224. Altyar, A.E.; Albadrani, G.M.; Farouk, S.M.; Alamoudi, M.K.; Sayed, A.A.; Mohammedsaleh, Z.M.; Al-Ghadi, M.Q.; Saleem, R.M.; Sakr, H.I.; Abdel-Daim, M.M. The Antioxidant, Anti-Inflammatory, and Anti-Apoptotic Effects of Sesamin against Cisplatin-Induced Renal and Testicular Toxicity in Rats. Ren. Fail. 2024, 46, 2378212. [Google Scholar] [CrossRef]
  225. Rousta, A.M.; Mirahmadi, S.M.; Shahmohammadi, A.; Nourabadi, D.; Khajevand-Khazaei, M.R.; Baluchnejadmojarad, T.; Roghani, M. Protective Effect of Sesamin in Lipopolysaccharide-Induced Mouse Model of Acute Kidney Injury Via Attenuation of Oxidative Stress, Inflammation, and Apoptosis. Immunopharmacol. Immunotoxicol. 2018, 40, 423–429. [Google Scholar] [CrossRef]
  226. Zhang, R.; Yu, Y.; Deng, J.; Zhang, C.; Zhang, J.; Cheng, Y.; Luo, X.; Han, B.; Yang, H. Sesamin Ameliorates High-Fat Diet-Induced Dyslipidemia and Kidney Injury by Reducing Oxidative Stress. Nutrients 2016, 8, 276. [Google Scholar] [CrossRef]
  227. Zhang, N.; Zhou, J.; Zhao, L.; Zhao, Z.; Wang, S.; Zhang, L.; Zhou, F. Ferulic Acid Supplementation Alleviates Hyperuricemia in High-Fructose/Fat Diet-Fed Rats Via Promoting Uric Acid Excretion and Mediating the Gut Microbiota. Food Funct. 2023, 14, 1710–1725. [Google Scholar] [CrossRef]
  228. Zhou, X.; Zhang, B.; Zhao, X.; Lin, Y.; Zhuang, Y.; Guo, J.; Wang, S. Chlorogenic Acid Prevents Hyperuricemia Nephropathy Via Regulating Tmao-Related Gut Microbes and Inhibiting the Pi3k/Akt/Mtor Pathway. J. Agric. Food Chem. 2022, 70, 10182–10193. [Google Scholar] [CrossRef] [PubMed]
  229. Zhou, X.; Zhang, B.; Zhao, X.; Lin, Y.; Wang, J.; Wang, X.; Hu, N.; Wang, S. Chlorogenic Acid Supplementation Ameliorates Hyperuricemia, Relieves Renal Inflammation, and Modulates Intestinal Homeostasis. Food Funct. 2021, 12, 5637–5649. [Google Scholar] [CrossRef] [PubMed]
  230. Qu, S.; Dai, C.; Hao, Z.; Tang, Q.; Wang, H.; Wang, J.; Zhao, H. Chlorogenic Acid Prevents Vancomycin-Induced Nephrotoxicity without Compromising Vancomycin Antibacterial Properties. Phytother. Res. 2020, 34, 3189–3199. [Google Scholar] [CrossRef] [PubMed]
  231. Li, H.-Y.; Yi, Y.-L.; Guo, S.; Zhang, F.; Yan, H.; Zhan, Z.-L.; Zhu, Y.; Duan, J.-A. Isolation, Structural Characterization and Bioactivities of Polysaccharides from Laminaria Japonica: A Review. Food Chem. 2022, 370, 131010. [Google Scholar] [CrossRef]
  232. Lu, J.; Yao, J.; Pu, J.; Wang, D.; Liu, J.; Zhang, Y.; Zha, L. Transcriptome Analysis of Three Medicinal Plants of the Genus Polygonatum: Identification of Genes Involved in Polysaccharide and Steroidal Saponins Biosynthesis. Front. Plant Sci. 2023, 14, 1293411. [Google Scholar] [CrossRef]
  233. Ge, J.; Liu, Z.; Zhong, Z.; Wang, L.; Zhuo, X.; Li, J.; Jiang, X.; Ye, X.-Y.; Xie, T.; Bai, R. Natural Terpenoids with Anti-Inflammatory Activities: Potential Leads for Anti-Inflammatory Drug Discovery. Bioorganic Chem. 2022, 124, 105817. [Google Scholar] [CrossRef] [PubMed]
  234. Galappaththi, M.C.A.; Patabendige, N.M.; Premarathne, B.M.; Hapuarachchi, K.K.; Tibpromma, S.; Dai, D.-Q.; Suwannarach, N.; Rapior, S.; Karunarathna, S.C. A Review of Ganoderma Triterpenoids and Their Bioactivities. Biomolecules 2022, 13, 24. [Google Scholar] [CrossRef]
  235. Lu, D.; Yang, Y.; Du, Y.; Zhang, L.; Yang, Y.; Tibenda, J.J.; Nan, Y.; Yuan, L. The Potential of Glycyrrhiza from “Medicine Food Homology” in the Fight against Digestive System Tumors. Molecules 2023, 28, 7719. [Google Scholar] [CrossRef]
  236. Wu, Y.; Wang, X.; Yang, L.; Kang, S.; Yan, G.; Han, Y.; Fang, H.; Sun, H. Potential of Alisols as Cancer Therapeutic Agents: Investigating Molecular Mechanisms, Pharmacokinetics and Metabolism. Biomed. Pharmacother. 2023, 168, 115722. [Google Scholar] [CrossRef]
  237. Lee, C.H.; Kim, J.H. A Review on the Medicinal Potentials of Ginseng and Ginsenosides on Cardiovascular Diseases. J. Ginseng Res. 2014, 38, 161–166. [Google Scholar] [CrossRef]
  238. Yokozawa, T.; Liu, Z.W.; Dong, E. A Study of Ginsenoside-Rd in a Renal Ischemia-Reperfusion Model. Nephron 1998, 78, 201–206. [Google Scholar] [CrossRef] [PubMed]
  239. Kouda, R.; Yakushiji, F. Recent Advances in Iridoid Chemistry: Biosynthesis and Chemical Synthesis. Chem.—Asian J. 2020, 15, 3771–3783. [Google Scholar] [CrossRef] [PubMed]
  240. Danielewski, M.; Matuszewska, A.; Nowak, B.; Kucharska, A.Z.; Sozański, T. The Effects of Natural Iridoids and Anthocyanins on Selected Parameters of Liver and Cardiovascular System Functions. Oxid. Med. Cell Longev. 2020, 2020, 2735790. [Google Scholar] [CrossRef]
  241. Bridi, R.; von Poser, G.L.; de Carvalho Meirelles, G. Iridoids as a Potential Hepatoprotective Class: A Review. Mini Rev. Med. Chem. 2023, 23, 452–479. [Google Scholar] [PubMed]
  242. Zhou, T.Y.; Tian, N.; Li, L.; Yu, R. Iridoids Modulate Inflammation in Diabetic Kidney Disease: A Review. J. Integr. Med. 2024, 22, 210–222. [Google Scholar] [CrossRef]
  243. Kou, Y.; Li, Z.; Yang, T.; Shen, X.; Wang, X.; Li, H.; Zhou, K.; Li, L.; Xia, Z.; Zheng, X.; et al. Therapeutic Potential of Plant Iridoids in Depression: A Review. Pharm. Biol. 2022, 60, 2167–2181. [Google Scholar] [CrossRef]
  244. Zhang, F.; Yan, Y.; Xu, J.K.; Zhang, L.M.; Li, L.; Chen, X.; Li, D.X.; Peng, Y.; Yang, H.; Li, L.Z.; et al. Simultaneous Determination of Thirteen Iridoid Glycosides in Crude and Processed Fructus Corni from Different Areas by Uplc-Ms/Ms Method. J. Chromatogr. Sci. 2024, 62, 562–569. [Google Scholar] [CrossRef]
  245. Cheng, C.; Li, Z.; Zhao, X.; Liao, C.; Quan, J.; Bode, A.M.; Cao, Y.; Luo, X. Natural Alkaloid and Polyphenol Compounds Targeting Lipid Metabolism: Treatment Implications in Metabolic Diseases. Eur. J. Pharmacol. 2020, 870, 172922. [Google Scholar] [CrossRef]
  246. Li, J.; Gong, X. Tetramethylpyrazine: An Active Ingredient of Chinese Herbal Medicine W Ith Therapeutic Potential in Acute Kidney Injury and Renal Fibrosis. Front. Pharmacol. 2022, 13, 820071. [Google Scholar] [CrossRef]
  247. Wu, D.; Wen, W.; Qi, C.L.; Zhao, R.X.; Lü, J.H.; Zhong, C.Y.; Chen, Y.Y. Ameliorative Effect of Berberine on Renal Damage in Rats with Diabetes Induced by High-Fat Diet and Streptozotocin. Phytomedicine 2012, 19, 712–718. [Google Scholar] [CrossRef]
  248. Tang, L.; Lv, F.; Liu, S.; Zhang, S. Effect of Berberine on Expression of Transforming Growth Factor-Beta1 and Type Iv Collagen Proteins in Mesangial Cells of Diabetic Rats with Nephropathy. Zhongguo Zhong Yao Za Zhi = Zhongguo Zhongyao Zazhi = China J. Chin. Mater. Medica 2011, 36, 3494–3497. [Google Scholar]
  249. Chen, Q.; Li, D.; Wu, F.; He, X.; Zhou, Y.; Sun, C.; Wang, H.; Liu, Y. Berberine Regulates the Metabolism of Uric Acid and Modulates Intestinal Flora in Hyperuricemia Rats Model. Comb. Chem. High. Throughput Screen. 2023, 26, 2057–2066. [Google Scholar] [CrossRef] [PubMed]
  250. Shan, B.; Wu, M.; Chen, T.; Tang, W.; Li, P.; Chen, J. Berberine Attenuates Hyperuricemia by Regulating Urate Transporters and Gut Microbiota. Am. J. Chin. Med. 2022, 50, 2199–2221. [Google Scholar] [CrossRef] [PubMed]
  251. Pan, L.; Han, P.; Ma, S.; Peng, R.; Wang, C.; Kong, W.; Cong, L.; Fu, J.; Zhang, Z.; Yu, H.; et al. Abnormal Metabolism of Gut Microbiota Reveals the Possible Molecular Mechanism of Nephropathy Induced by Hyperuricemia. Acta Pharm. Sin. B 2020, 10, 249–261. [Google Scholar] [CrossRef] [PubMed]
  252. Wang, T.; Zhang, J.; Wei, H.; Wang, X.; Xie, M.; Jiang, Y.; Zhou, J. Matrine-Induced Nephrotoxicity Via Gsk-3β/Nrf2-Mediated Mitochondria-Dependent Apoptosis. Chem. Biol. Interact. 2023, 378, 110492. [Google Scholar] [CrossRef]
  253. Wang, X.; Lin, Z.; Li, T.; Zhu, W.; Huang, H.; Hu, J.; Zhou, J. Sodium Selenite Prevents Matrine-Induced Nephrotoxicity by Suppressing Ferroptosis Via the Gsh-Gpx4 Antioxidant System. Biol. Trace Element Res. 2024, 202, 4674–4686. [Google Scholar] [CrossRef]
  254. Wang, H.-W.; Shi, L.; Xu, Y.-P.; Qin, X.-Y.; Wang, Q.-Z. Oxymatrine Inhibits Renal Fibrosis of Obstructive Nephropathy by Downregulating the Tgf-Β1-Smad3 Pathway. Ren. Fail. 2016, 38, 945–951. [Google Scholar] [CrossRef]
  255. Zhu, Z.; Chen, R.; Zhang, L.; Zhu, Z. Simple Phenylpropanoids: Recent Advances in Biological Activities, Biosynthetic Pathways, and Microbial Production. Nat. Prod. Rep. 2024, 41, 6–24. [Google Scholar] [CrossRef]
  256. Zhu, X.; Quan, Y.-Y.; Yin, Z.-J.; Li, M.; Wang, T.; Zheng, L.-Y.; Feng, S.-Q.; Zhao, J.-N.; Li, L. Sources, Morphology, Phytochemistry, Pharmacology of Curcumae Longae Rhizoma, Curcumae Radix, and Curcumae Rhizoma: A Review of the Literature. Front. Pharmacol. 2023, 14, 1229963. [Google Scholar] [CrossRef]
  257. Pivari, F.; Mingione, A.; Piazzini, G.; Ceccarani, C.; Ottaviano, E.; Brasacchio, C.; Cas, M.D.; Vischi, M.; Cozzolino, M.G.; Fogagnolo, P.; et al. Curcumin Supplementation (Meriva®) Modulates Inflammation, Lipid Peroxidation and Gut Microbiota Composition in Chronic Kidney Disease. Nutrients 2022, 14, 231. [Google Scholar] [CrossRef]
  258. Xu, X.; Wang, H.; Guo, D.; Man, X.; Liu, J.; Li, J.; Luo, C.; Zhang, M.; Zhen, L.; Liu, X. Curcumin Modulates Gut Microbiota and Improves Renal Function in Rats with Uric Acid Nephropathy. Ren. Fail. 2021, 43, 1063–1075. [Google Scholar] [CrossRef] [PubMed]
  259. Mohtashami, L.; Amiri, M.S.; Ayati, Z.; Ramezani, M.; Jamialahmadi, T.; Emami, S.A.; Sahebkar, A. Ethnobotanical Uses, Phytochemistry and Pharmacology of Different Rheum Species (Polygonaceae): A Review. Adv. Exp. Med. Biol. 2021, 1308, 309–352. [Google Scholar]
  260. Zeng, Y.Q.; Dai, Z.; Lu, F.; Lu, Z.; Liu, X.; Chen, C.; Qu, P.; Li, D.; Hua, Z.; Qu, Y.; et al. Emodin Via Colonic Irrigation Modulates Gut Microbiota and Reduces Uremic Toxins in Rats with Chronic Kidney Disease. Oncotarget 2016, 7, 17468–17478. [Google Scholar] [CrossRef]
  261. Tucker, P.S.; Scanlan, A.T.; Dalbo, V.J. Chronic Kidney Disease Influences Multiple Systems: Describing the Relationship between Oxidative Stress, Inflammation, Kidney Damage, and Concomitant Disease. Oxidative Med. Cell. Longev. 2015, 2015, 1–8. [Google Scholar] [CrossRef]
  262. Tain, Y.-L.; Hsu, C.-N. Perinatal Oxidative Stress and Kidney Health: Bridging the Gap between Animal Models and Clinical Reality. Antioxidants 2023, 12, 13. [Google Scholar] [CrossRef] [PubMed]
  263. Mapuskar, K.A.; Pulliam, C.F.; Zepeda-Orozco, D.; Griffin, B.R.; Furqan, M.; Spitz, D.R.; Allen, B.G. Redox Regulation of Nrf2 in Cisplatin-Induced Kidney Injury. Antioxidants 2023, 12, 1728. [Google Scholar] [CrossRef]
  264. Honda, T.; Hirakawa, Y.; Nangaku, M. The Role of Oxidative Stress and Hypoxia in Renal Disease. Kidney Res. Clin. Pract. 2019, 38, 414–426. [Google Scholar] [CrossRef] [PubMed]
  265. Allameh, H.; Fatemi, I.; Malayeri, A.R.; Nesari, A.; Mehrzadi, S.; Goudarzi, M. Pretreatment with Berberine Protects against Cisplatin-Induced Renal Injury in Male Wistar Rats. Naunyn-Schmiedebergs Arch. Pharmacol. 2020, 393, 1825–1833. [Google Scholar] [CrossRef]
  266. Verma, V.K.; Malik, S.; Mutneja, E.; Sahu, A.K.; Rupashi, K.; Dinda, A.K.; Arya, D.S.; Bhatia, J. Mechanism Involved in Fortification by Berberine in Cddp-Induced Nephrotoxicity. Curr. Mol. Pharmacol. 2020, 13, 342–352. [Google Scholar] [CrossRef]
  267. El-Horany, H.E.-S.; Gaballah, H.H.; Helal, D.S. Berberine Ameliorates Renal Injury in a Rat Model of D-Galactose-Induced Aging through a Pten/Akt-Dependent Mechanism. Arch. Physiol. Biochem. 2020, 126, 157–165. [Google Scholar] [CrossRef]
  268. Hasanein, P.; Riahi, H. Preventive Use of Berberine in Inhibition of Lead-Induced Renal Injury in Rats. Environ. Sci. Pollut. Res. 2018, 25, 4896–4903. [Google Scholar] [CrossRef] [PubMed]
  269. Basile, D.P.; Anderson, M.D.; Sutton, T.A. Pathophysiology of Acute Kidney Injury. Compr. Physiol. 2012, 2, 1303–1353. [Google Scholar] [PubMed]
  270. Fu, Y.; Xiang, Y.; Li, H.; Chen, A.; Dong, Z. Inflammation in Kidney Repair: Mechanism and Therapeutic Potential. Pharmacol. Ther. 2022, 237, 108240. [Google Scholar] [CrossRef] [PubMed]
  271. Mosquera-Sulbaran, J.A.; Pedreañez, A.; Vargas, R.; Hernandez-Fonseca, J.P. Apoptosis in Post-Streptococcal Glomerulonephritis and Mechanisms for Failed of Inflammation Resolution. Pediatr. Nephrol. 2024, 39, 1709–1724. [Google Scholar] [CrossRef] [PubMed]
  272. Zhang, H.; Deng, Z.; Wang, Y. Molecular Insight in Intrarenal Inflammation Affecting Four Main Types of Cells in Nephrons in Iga Nephropathy. Front. Med. 2023, 10, 1128393. [Google Scholar] [CrossRef]
  273. Vallés, P.G.; Lorenzo, A.F.G.; Garcia, R.D.; Cacciamani, V.; Benardon, M.E.; Costantino, V.V. Toll-Like Receptor 4 in Acute Kidney Injury. Int. J. Mol. Sci. 2023, 24, 1415. [Google Scholar] [CrossRef]
  274. Yeh, T.H.; Tu, K.C.; Wang, H.Y.; Chen, J.Y. From Acute to Chronic: Unraveling the Pathophysiological Mechanisms of the Progression from Acute Kidney Injury to Acute Kidney Disease to Chronic Kidney Disease. Int. J. Mol. Sci. 2024, 25, 1755. [Google Scholar] [CrossRef]
  275. Lovisa, S.; Zeisberg, M.; Kalluri, R. Partial Epithelial-to-Mesenchymal Transition and Other New Mechanisms of Kidney Fibrosis. Trends Endocrinol. Metab. 2016, 27, 681–695. [Google Scholar] [CrossRef]
  276. La Russa, A.; Serra, R.; Faga, T.; Crugliano, G.; Bonelli, A.; Coppolino, G.; Bolignano, D.; Battaglia, Y.; Ielapi, N.; Costa, D.; et al. Kidney Fibrosis and Matrix Metalloproteinases (Mmps). Front. Biosci.-Landmark 2024, 29, 192. [Google Scholar] [CrossRef]
  277. Evenepoel, P.; Poesen, R.; Meijers, B. The Gut-Kidney Axis. Pediatr. Nephrol. 2017, 32, 2005–2014. [Google Scholar] [CrossRef]
  278. Ramya Ranjan Nayak, S.P.; Boopathi, S.; Haridevamuthu, B.; Arockiaraj, J. Toxic Ties: Unraveling the Complex Relationship between Endocrine Disrupting Chemicals and Chronic Kidney Disease. Environ. Pollut. 2023, 338, 122686. [Google Scholar] [CrossRef] [PubMed]
  279. Hayeeawaema, F.; Muangnil, P.; Jiangsakul, J.; Tipbunjong, C.; Huipao, N.; Khuituan, P. A Novel Model of Adenine-Induced Chronic Kidney Disease-Associated Gastrointestinal Dysfunction in Mice: The Gut-Kidney Axis. Saudi J. Biol. Sci. 2023, 30, 103660. [Google Scholar] [CrossRef] [PubMed]
  280. Lu, X.; Ma, J.; Li, R. Alterations of Gut Microbiota in Biopsy-Proven Diabetic Nephropathy and a Long History of Diabetes without Kidney Damage. Sci. Rep. 2023, 13, 12150. [Google Scholar] [CrossRef] [PubMed]
  281. Jiang, S.; Xie, S.; Lv, D.; Zhang, Y.; Deng, J.; Zeng, L.; Chen, Y. A Reduction in the Butyrate Producing Species Roseburia Spp. and Faecalibacterium prausnitzii Is Associated with Chronic Kidney Disease Progression. Antonie Van Leeuwenhoek 2016, 109, 1389–1396. [Google Scholar] [CrossRef]
  282. Wong, J.; Piceno, Y.M.; DeSantis, T.Z.; Pahl, M.; Andersen, G.L.; Vaziri, N.D. Expansion of Urease- and Uricase-Containing, Indole- and P-Cresol-Forming and Contraction of Short-Chain Fatty Acid-Producing Intestinal Microbiota in Esrd. Am. J. Nephrol. 2014, 39, 230–237. [Google Scholar] [CrossRef]
  283. Marques, F.Z.; Nelson, E.; Chu, P.Y.; Horlock, D.; Fiedler, A.; Ziemann, M.; Tan, J.K.; Kuruppu, S.; Rajapakse, N.W.; El-Osta, A.; et al. High-Fiber Diet and Acetate Supplementation Change the Gut Microbiota and Prevent the Development of Hypertension and Heart Failure in Hypertensive Mice. Circulation 2017, 135, 964–977. [Google Scholar] [CrossRef] [PubMed]
  284. Liang, Z.; Tang, Z.; Zhu, C.; Li, F.; Chen, S.; Han, X.; Zheng, R.; Hu, X.; Lin, R.; Pei, Q.; et al. Intestinal Cxcr6+ Ilc3s Migrate to the Kidney and Exacerbate Renal Fibrosis Via Il-23 Receptor Signaling Enhanced by Pd-1 Expression. Immunity 2024, 57, 1306–1323. [Google Scholar] [CrossRef]
  285. Yan, J.; Herzog, J.W.; Tsang, K.; Brennan, C.A.; Bower, M.A.; Garrett, W.S.; Sartor, B.R.; Aliprantis, A.O.; Charles, J.F. Gut Microbiota Induce Igf-1 and Promote Bone Formation and Growth. Proc. Natl. Acad. Sci. USA 2016, 113, E7554–E7563. [Google Scholar] [CrossRef]
  286. Gleeson, P.J.; Benech, N.; Chemouny, J.; Metallinou, E.; Berthelot, L.; da Silva, J.; Bex-Coudrat, J.; Boedec, E.; Canesi, F.; Bounaix, C.; et al. The Gut Microbiota Posttranslationally Modifies Iga1 in Autoimmune Glomerulonephritis. Sci. Transl. Med. 2024, 16, eadl6149. [Google Scholar] [CrossRef]
  287. Lan, T.; Tang, T.; Li, Y.; Duan, Y.; Yuan, Q.; Liu, W.; Ren, Y.; Li, N.; Liu, X.; Zhang, Y.; et al. Ftz Polysaccharides Ameliorate Kidney Injury in Diabetic Mice by Regulating Gut-Kidney Axis. Phytomedicine 2023, 118, 154935. [Google Scholar] [CrossRef]
  288. Kunter, U.; Seikrit, C.; Floege, J. Novel Agents for Treating Iga Nephropathy. Curr. Opin. Nephrol. Hypertens. 2023, 32, 418–426. [Google Scholar] [CrossRef] [PubMed]
  289. Zhang, Z.-W.; Han, P.; Fu, J.; Yu, H.; Xu, H.; Hu, J.-C.; Lu, J.-Y.; Yang, X.-Y.; Zhang, H.-J.; Bu, M.-M.; et al. Gut microbiota-based metabolites of Xiaoyao Pills (a typical Traditional Chinese medicine) ameliorate depression by inhibiting fatty acid amide hydrolase levels in brain. J. Ethnopharmacol. 2023, 313, 116555. [Google Scholar] [CrossRef] [PubMed]
  290. Zhang, Z.-W.; Gao, C.-S.; Zhang, H.; Yang, J.; Wang, Y.-P.; Pan, L.-B.; Yu, H.; He, C.-Y.; Luo, H.-B.; Zhao, Z.-X.; et al. Morinda officinalis oligosaccharides increase serotonin in the brain and ameliorate depression via promoting 5-hydroxytryptophan production in the gut microbiota. Acta Pharm. Sin. B 2022, 12, 3298–3312. [Google Scholar] [CrossRef] [PubMed]
  291. Wang, Y.; Tong, Q.; Ma, S.-R.; Zhao, Z.-X.; Pan, L.-B.; Cong, L.; Han, P.; Peng, R.; Yu, H.; Lin, Y.; et al. Oral berberine improves brain dopa/dopamine levels to ameliorate Parkinson’s disease by regulating gut microbiota. Signal Transduct. Target. Ther. 2021, 6, 77. [Google Scholar] [CrossRef]
  292. Feng, R.; Shou, J.-W.; Zhao, Z.-X.; He, C.-Y.; Ma, C.; Huang, M.; Fu, J.; Tan, X.-S.; Li, X.-Y.; Wen, B.-Y.; et al. Transforming berberine into its intestine-absorbable form by the gut microbiota. Sci. Rep. 2015, 5, 12155. [Google Scholar] [CrossRef]
  293. Wu, G.D.; Chen, J.; Hoffmann, C.; Bittinger, K.; Chen, Y.-Y.; Keilbaugh, S.A.; Bewtra, M.; Knights, D.; Walters, W.A.; Knight, R.; et al. Linking Long-Term Dietary Patterns with Gut Microbial Enterotypes. Science 2011, 334, 105–108. [Google Scholar] [CrossRef]
  294. Turnbaugh, P.J.; Ridaura, V.K.; Faith, J.J.; Rey, F.E.; Knight, R.; Gordon, J.I. The Effect of Diet on the Human Gut Microbiome: A Metagenomic Analysis in Humanized Gnotobiotic Mice. Sci. Transl. Med. 2009, 1, 6ra14. [Google Scholar] [CrossRef]
Figure 1. Illustration abstract drawn by authors.
Figure 1. Illustration abstract drawn by authors.
Nutrients 16 03530 g001
Figure 2. Mechanisms involved in the interplay of dietary medical botanicals and kidney function, drawn by authors using Figdraw (www.figdraw.com).
Figure 2. Mechanisms involved in the interplay of dietary medical botanicals and kidney function, drawn by authors using Figdraw (www.figdraw.com).
Nutrients 16 03530 g002
Table 1. The full list of botanicals in the China index for medicine–food homology.
Table 1. The full list of botanicals in the China index for medicine–food homology.
Latin NameEnglish Common NameEMA Herbal MedicineEFSA
Compendium
NIH DSLD
Eugenia caryophyllata Thunb.Clove
Illicium verum Hook. f.Star anise
Canavalia gladiata (Jacq.) DC.Sword bean
Foeniculum vulgare Mill.Fennel
Cirsium setosum (Willd.) MB.Thistle
Dioscorea opposita Thunb.Chinese yam
Crataegus pinnatifida Bge. var. major N.E.Br./Crataegus pinnatifida Bge.Hawthorn
Portulaca oleracea L.Purslane
Prunus mume (Sieb.) Sieb. et Zucc.Japanese apricot
Chaenomeles speciosa (Sweet) NakaiFlowering quince
Cannabis sativa L.Hemp
Citrus aurantium L. var. amara Engl.Bitter orange
Polygonatum odoratum (Mill.) DruceSolomon’s seal
Glycyrrhiza uralensis Fisch./Glycyrrhiza inflata Bat./Glycyrrhiza glabra L.Licorice
Angelica dahurica (Fisch. ex Hoffm.) Benth. et Hook. f./Angelica dahurica (Fisch. ex Hoffm.) Benth. et Hook. f. var. formosana (Boiss.) Shan et YuanChinese angelica
Ginkgo biloba L.Ginkgo
Dolichos lablab L.Hyacinth bean
Dimocarpus longan Lour.Longan
Cassia obtusifolia L./Cassia tora L.Sicklepod
Lilium lancifolium Thunb./Lilium pumilum DC.Tiger lily
Myristica fragrans Houtt.Nutmeg
Cinnamomum cassia PreslCassia
Phyllanthus emblica L.Amla
Citrus medica L. var. sarcodactylis SwingleFingered citron
Prunus armeniaca L. var. ansu Maxim/Prunus sibirica L./Prunus mandshurica (Maxim) Koehne/Prunus armeniaca L.Chinese apricot
Hippophae rhamnoides L.Sea buckthorn
Euryale ferox Salisb.Euryale ferox
Zanthoxylum schinifolium Sieb. et Zucc./Zanthoxylum bungeanum Maxim.Sichuan pepper
Vigna umbeuata Ohwi et Ohashi/Vigna angularis Ohwi et OhashiAzuki bean
Hordeum vulgare L.Barley
Laminaria japonica Aresch./Ecklonia kurome Okam.Kunbu
Ziziphus jujuba Mill.Jujube
Siraitia grosvenorii (Swingle.) C.Jeffrey ex A. M. Lu et Z. Y.ZhangLuohanguo siraitia fruit
Prunus japonica Thunb.Pruni semen
Lonicera japonica Thunb.Japanese honeysuckle
Canarium album Raeusch.White canarium
Houttuynia cordata Thunb.Fish mint
Zingiber officinale Rosc.Ginger
Hovenia dulcis Thunnb./Hovenia acerba Lindl./Hovenia trichocarpa Chun et TsiangJapanese raisin tree
Lycium barbarum L.Goji berry
Gardenia jasminoides EllisCape jasmine
Amomum villosum Lour./Amomum villosum Lour. var. xanthioides T. L. Wu et Senjen/Amomum longiligulare T. L. WuVillus amomum
Sterculia lychnophora HanceMalva nut tree
Poria cocos (Schw.) WolfPoria, fu ling
Citrus medica L./Citrus wilsonii TanakaCitron
Mosla chinensis Maxim./Mosla chinensis Maxim. cv. JiangxiangruChinese mosla
Prunus persica (L.) Batsch/Prunus davidiana (Carr.) Franch.Peach
Morus alba L.White mulberry
Citrus reticulata BlancoMandarin orange
Platycodon grandiflorum (Jacq.) A. DC.Balloon flower
Alpinia oxyphylla Miq.Sharp-leaf galangal
Nelumbo nucifera Gaertn.Lotus leaf
Raphanus sativus L.Radish seed
Alpinia officinarum HanceLesser galangal
Lophatherum gracile Brongn.Lophatherum
Glycine max (L.) Merr.Soybean
Chrysanthemum morifolium Ramat.Chrysanthemum
Cichorium glandulosum Boiss. et Huet/Cichorium intybus L.Chicory
Polygonatum kingianum Coll.et Hemsl./Polygonatum sibiricum Red./Polygonatum cyrtonema HuaPolygonati rhizoma
Sinapis alba L.White mustard
Perilla frutescens (L.) BrittonPerilla
Pueraria lobata (Willd.) Ohwi/Pueraria thomsonii Benth.Kudzu
Sesamum indicum L.Sesame
Piper nigrum L.Black pepper
Sophora japonica L.Sophora
Taraxacum mongolicum Hand. Mazz./Taraxacum borealisinense Kitam.Dandelion
Torreya grandis Fort.Torreya
Ziziphus jujuba Mill. var. spinosa (Bunge) Hu ex H. F. ChouChinese date
Imperata cylindrical Beauv. var. major (nees) C. E. Hubb.Cogongrass
Phragmites communis Trin.Reed
Mentha haplocalyx Briq.Chinese mint
Coix lacryma-jobi L. var. mayuen (Roman.) StapfJob’s tears
Allium macrostemon Bge./Allium chinense G. DonChinese onion
Rubus chingii HuChinese raspberry
Pogostemon cablin (Blanco) Benth./Agastache rugosus (Fisch. et Mey.) O. Ktze.Patchouli
Angelica sinensis (Oliv.) DielsAngelica root
Kaempferia galanga L.Aromatic ginger
Crocus sativus L.Saffron
Amomum tsao-ko Crevost & LemariéCardamom
Curcuma longa L.Turmeric
Piper longum L.Long pepper
Codonopsis pilosula (Franch.) Nannf./Codonopsis pilosula Nannf. var. modesta (Nannf.) L. T. Shen/Codonopsis tangshen Oliv.Dangshen
Cistanche deserticola Y. C. MaDesert cistanche
Dendrobium officinale Kimura et MigoChinese orchid
Panax quinquefolius L.American ginseng
Astragalus membranaceus (Fisch.) Bge.Mongolian milkvetch (huangqi)
Ganoderma lucidum (Leyss. ex Fr.) Karst./Ganoderma sinense Zhao, Xu et ZhangGanoderma
Cornus officinalis Sieb. et Zucc.Japanese cornel dogwood
Gastrodia elata Bl.Gastrodia
Eucommia ulmoides Oliv.Hardy rubber tree
EMA: European Medicines Agency; EFSA: European Food Safety Authority; NIH: National Institutes of Health; DSLD: Dietary Supplement Label Database; √: Indicating this botanical also appears in the index or database of ingredients; var.: Variety; f.: Form; cv.: Cultivar.
Table 2. Nutrients from these botanical food ingredients with kidney-protective effects.
Table 2. Nutrients from these botanical food ingredients with kidney-protective effects.
Botanical ResourcesCompoundsModelsEffectsRef.
Flavonoids
Allium macrostemon Bge., Crocus sativus L., Plantago asiatica L. SQuercetinDN rats, ochratoxin A-induced AKI mice↓ PI3K/AKT signaling
↑ Nrf2/HO-1, fatty acid oxidation
[37,38,39,74,75]
Glycine max (L.) Merr., Lonicera japonica Thunb., Ginkgo biloba L., Lycium barbarum L., Plantago asiatica L. SKaempferolCLP-induced AKI mice, DN mice, DOX-induced AKI mice↓ ICAM-1, VCAM-1, MCP-1, caspase-3, Bax, MAPK signaling, TGF-β1, and α-SMA
↑ Bcl-2, SOD, and GSH
[40,76,77,78]
Myricaceae, Polygonaceae, Primulaceae, Pinaceae, and AnacardiaceaeMyricetinDN mice, EG-induced AKI mice↓ IL-1β, TNF-α, ROCK1/ERK/P38 signaling, and NF-κB signaling
↑ Nrf2, CAT, and SOD
[41,42,79]
Ginkgo biloba L., Hippophae rhamnoides L., Citrus reticulata Blanco, Citrus aurantium L., and Citrus medica L.IsorhamnetinLPS-induced AKI mice↓ IL-1β, IL-6, and TNF-α and M1 macrophage
↑ M2 macrophage
[43,44]
Epimedium brevicornu Maxim. SIcariinAdenine/UUO-induced CKD rats, DOX-induced AKI mice↓ TGF-β, α-SMA, and E-cadherin
↑ Nrf2/HO-1, SOD, CAT
[47,48,80,81,82]
Plantago asiatica L. SApigeninDOX/EG/oxonate-induced AKI mice, UAN mice↓ Oxidative and nitrosative stress, IL-6, TNF-α, NLRP3, caspase-1, IL-1β, TGF-β, Wnt/β-catenin signaling[49,50,83,84,85,86,87]
Citrus reticulata Blanco, Citrus aurantium L., Citrus medica L.NobiletinIRI-induced AKI mice↑ PI3K/AKT signaling[55]
Crataegus pinnatifida Bge.VitexinUUO-induced CKD mice, oxalate-induced AKI mice↓ NLRP3, caspase-1, and IL-1β
↑ Nrf2/HO-1, SOD, GSH
[56,57]
Citrus reticulata Blanco, Citrus aurantium L., Citrus medica L.Hesperidin, hesperetinLPS/Zn-induced AKI mice, DDP-induced HK-2 cells↓ p53, caspase-3
↑ Nrf2/HO-1, SOD, GSH, and CAT
Regulating gut microbiota
[58,59,60,61]
Plantago asiatica L. SLuteolinCd/HgCl2/K2Cr2O7-induced AKI mice, LN mice↓ HIF-1α, α-SMA, collagen I, and fibronectin
↑ Nrf2/HO-1, GSH, SOD, CAT, and AMPK/mTOR autophagy
[88,89,90,91,92]
Pueraria lobata (Willd.) OhwiPuerarinDN mice, UUO-induced CKD mice↑ Nrf2, cAMP/PKA/CREB
↓ TLR4/MyD8, and M1 macrophage
[72,93,94,95]
Polysaccharides
Astragalus membranaceus (Fisch.) Bge./DDP-induced AKI mice↓ ROS generation and mitochondrial vacuolation[19]
/LPS-induced AKI mice↓ Caspase-3/9 and Bax
↑ Bcl-2
[96]
/LPS-induced AKI miceRegulating gut microbiota
↑ SCFAs
[97]
Bletilla striataSMw: 260 kDaAng II-induced HMCs↓ ROS generation and NOX4[18]
/TGF-β-induced HMCs↓ TGF-β, and α-SMA[98]
Ganoderma lucidum (Leyss. ex Fr.) Karst.Mw: 72.9 kDa;
Ara:Gal:Rha:Glc = 0.08:0.21:0.24:0.47
DN mice↓ Collagen-1, fibronectin, α-SMA, TGF- β, and MAPK/NF-κB signaling [99]
/IRI-induced AKI mice↓ p53, caspase-3, Bax, cytochrome c, and ER stress
↑ Bcl-2
[100]
Lycium barbarum L.Ara:Gal:Glc:GalA:Man:Rha = 12.25: 8.66: 7.66: 2.86: 1.70: 1.00DN mice↓ TNF-α, IL-1β, IL-6, and NF-κB signaling[16]
/Lead-induced AKI mice↓ Bax and caspase-3
↑ Bcl-2
[101]
Laminaria japonica Aresch./Ecklonia kurome Okam.Mw: 7 kDaIRI-induced AKI mice↓ p53, Bax, MMP, and cytochrome c
↑ Bcl-2
[102]
Mw: 1960 kDaDOX-induced AKI mice↓ TNF-α, IL-1β, MCP-1, and podocyte injury[103]
Mw: 8.84 kDa;
Fuc:Gal:Man:Glc:Rha:Xyl = 1:0.057:0.041:0.008:0.029:0.019
DN mice↓ collagen-1, fibronectin, and α-SMA[104]
Mw: 7 kDaDN mice↓ α-SMA, fibronectin, and TGF-β/Smad
↑ E-cadherin
[105]
/DOX-induced CKD mice↓ α-SMA, fibronectin, and TGF-β/Smad[106]
Mw: 7.774 kDaAGE-induced HRMCs↓ Fibronectin [107]
Mw: 8.84 kDaTGF-β1- or FGF-2-induced HK-2 cells↓ α-SMA, MMP9, and EMT[108]
Polygonatum kingianum Coll.et Hemsl./Polygonatum sibiricum Red./Polygonatum cyrtonema HuaMw: 141 kDa;
Gal:GalA:Ara:Glc = 57.67:26.82:4.59:4.54
Uranium-induced HK-2 cells↓ ROS
↑ GSK-3β/Fyn/Nrf2
[17]
Panax ginseng C. A. Mey. SGlc:Gal:Ara:GalA:Rha:Man = 76.7:6.5:5.1:9.2:1.4:1.1DDP-induced AKI mice↓ p53, caspase-3, caspase-6, and ER stress by PERK/eIF2α/ATF4 signaling[109]
Paeonia × suffruticosaSMw: 164 kDa;
D-Glc:L-Ara = 3.31:2.25;
DN rats↓ TGF-β, ICAM-1, and VCAM-1[110]
Mw: 164 kDa;
Ara = 3.31:2.25
DN ratsRegulating gut microbiota
↑ SCFAs
[111]
Plantago asiaticaS/Adenine-induced rats, UAN↓ IL-6, TNF-α, NLRP3, and caspase-1[112]
Dendrobium officinale Kimura et Migo/HG-induced HK-2 cells, db/db mice↓ TGF-β, and α-SMA
↑ SIRT1
[113]
Salvia miltiorrhiza Bunge S/Florfenicol-induced AKI broilers↓ p53, caspase-3
↑ Nrf2/HO-1
[114]
Phyllostachys nigraSMw: 34 kDaDN miceRegulating gut microbiota
Lactobacillales
[20]
Terpenoids
Rehmannia glutinosaS, Plantago asiaticaS, Scrophularia ningpoensisS, and Crocus sativus L.CatalpolDN mice, DOX/Ang II/Fru/DDP-induced AKI mice, adenine-induced CKD mice↓ TRPC6, NF-κB, TGF-β1/Smad, TLR4/MyD88 signaling, RAGE/RhoA/ROCK signaling
↑ AMPK signaling
[115,116,117,118,119,120,121]
Cornus officinalis Sieb. et Zucc.LoganinDN mice, IRI/DOX/CLP-induced AKI mice↓ NLRP3, AGE/RAGE signaling
↑ Nrf2/HO-1
[122,123,124,125]
Gardenia jasminoides Ellis, Eucommia ulmoides Oliv., Rehmannia glutinosa S, and Scrophularia ningpoensis SGeniposideDN mice, CLP-induced AKI mice, H2O2-induced HK-2 cells↓ ICAM-1, TNF-α, IL-1, IL-6, NF-κB, and NETs
↑ GSK3β, AMPK-PI3K/AKT, Bcl-2
Regulating gut microbiota
[126,127,128,129,130,131,132]
Cornus officinalis Sieb. et Zucc.MorronisideH2O2-induced podocytes↓ NOX4[133]
Canarium album Raeusch.OleuropeinDN mice, acrylamide-induced AKI mice↓ TNF-α, IFN-γ, IL-2, IL-6, and IL-17α
↑ SOD, GSH-Px, and CAT
[134,135]
Poria cocos (Schw.) Wolf.Poricoic acid (A, B, C, D, E, F, G, H, AM, AE, BM, DM, and derivatives)UUO-induced CKD mice↓ MMP-13, Wnt/β-catenin, TGF-β/Smad3/MAPK
↑ AMPK, Nrf2
[136,137,138,139,140,141,142]
Ligustrum lucidum Ait. SOleanic acidUUO-induced CKD mice, DN mice↓ NF-κB/TNF-α, TGF-β
↑ Nrf2/SIRT1/HO-1
Regulating gut microbiota
[143,144,145,146,147]
Alisma orientatle (Sam.) Juzep. SAlisol (A, B, and derivatives)IRI/DDP-induced AKI mice, UUO-induced CKD mice↓ ICAM-1, MCP-1, COX-2, iNOS, IL-6, TNF-α, FXR activation, TGF-β/Smad3
↑ SOD and GSH and HO-1
[148,149,150]
Panax ginseng C. A. Mey.GinsenosidesDDP-induced AKI mice, renal carcinoma, DN mice, UUO-induced CKD mice↓ ER stress, lipid peroxidation, PPARγ, and NOX4-MAPK pathways and TGF-β[151,152,153,154,155,156,157,158]
Ganoderma lucidum (Leyss. ex Fr.) Karst.Ganoderic acid, ganodermanontriol, lucidenic acidADPKD mice, IRI-induced AKI mice, UUO-induced CKD mice↓ Ras/MAPK, TGF-β/Smad, IL-6, COX-2, and iNOS
↓ TLR4/MyD88/NF-κB, and caspase-3
[159,160,161]
Glycyrrhiza uralensis Fisch.Glycyrrhizinic acid, glycyrrhizic acidDN mice, TAC/PC-induced AKI mice↓ ROS, IL-1β, IL-6, TNF-α, CCR2
↑ AMPK/SIRT1/PGC-1α
Regulates autophagy
[162,163,164,165,166,167]
Alkaloids
Ligusticum sinense Chuanxiong STetramethylpyrazine PC/IRI-induced AKI rats, DN rats↓ CCL2/CCR2, ROS, NLRP3, TNF-α
↑ AKT, Bcl-2
[168,169,170,171,172]
Leonurus japonicus Houtt. SLeonurineDDP/LPS/vancomycin-induced AKI mice, UUO-induced CKD mice↑ Nrf2
↓ TLR4/MyD88/NF-κB, TNF-α, IL-1β, TGF-β/Smad3
[173,174,175,176,177]
Coptis chinensisBerberineIRI/DOX/DDP/MTX/gentamicin-induced AKI, DN mice, and UUO-induced CKD mice, UAN mice↓ IL-6, IL-10, TGF-β/Smad3, mitochondrial stress, and ER stress
↑ Nrf2, MDA, SOD, CAT, GSH, and Bcl-2
Regulating gut microbiota
[178,179,180,181,182,183,184,185,186,187,188,189,190]
Trigonella foenum-graecum L. STrigonellineON mice, DN mice↓ EMT, ROS, α-SMA
↑ AMPK pathway
[191,192,193,194,195,196]
Piper longum L. SPiperlonguminineUUO-induced CKD mice↓ TRPC6[197]
Sophora japonica L.Matrine, oxymatrineIRI/DDP/gentamicin-induced AKI mice, UUO-induced CKD mice↓ IL-6, IL-10, TGF-β/Smad3[198,199,200]
Nelumbo nucifera Gaertn.Liensinine, IsoliensinineIRI-induced AKI rats, LPS-induced AKI mice↓ TGF-β1/Smad3[201,202]
Nelumbo nucifera Gaertn.Neferine, nuciferineUAN rats, LPS/IRI-induced AKI mice, DN mice↓ TLR4/MyD88/NF-κB, IL-1β
↑ Bcl-2
[203,204,205,206,207,208,209,210]
Others
Curcuma longa L.CurcuminLN mice, patulin-induced AKI mice, CKD patients, UAN rats↑ Nrf2/FOXO-3a, GSH
↓ PI3K/AKT/NF-κB, TGF-β, ROS
Regulating gut microbiota
[211,212,213,214,215]
Rheum palmatum L.Emodin, aloe emodin, rheinUUO-induced CKD mice/rats, DN mice↓ IL-1β, TGF-β, PERK-eIF2α
Regulating gut microbiota
[216,217,218,219,220]
Eugenia caryophyllata Thunb.EugenolIRI/patulin-induced AKI mice↓ TGF-β
↑ Nrf2
[221,222]
Sesamum indicum L.SesaminCKD mice, DDP/LPS-induced AKI mice↑ GSH, CAT, and SOD
Regulating gut microbiota
[223,224,225,226]
Ligusticum sinense Chuanxiong SFerulic acidUAN mice↓ TLR4/NF-κB
Regulating gut microbiota
[227]
Lonicera japonica Thunb.Chlorogenic acidUAN mice↓ IL-1β, TNF-α, IL-6
Regulating gut microbiota
[228,229,230]
S: These botanicals are medical herbs that could be added to dietary supplements in the index of Table S1; ↑:upregulating or promoting; ↓: downregulating or inhibiting; PI3K: Phosphoinositide 3-kinase; AKT: Protein kinase B; Nrf2: Nuclear factor erythroid 2-related factor 2; HO-1: Heme oxygenase 1; ICAM: Intercellular adhesion molecule; VCAM: Vascular cell adhesion molecule; NLRP3: NOD-, LRR-, and pyrin domain-containing protein 3; Bax: BCL2-associated X; Bcl-2: B-cell lymphoma 2; MAPK: Mitogen-activated protein kinase; AMPK: Adenosine monophosphate-activated protein kinase; NF-κB: Nuclear factor kappa-B; mTOR: Mammalian target of rapamycin; cAMP: Cyclic adenosine monophosphate; PKA: Protein kinase A; CREB: cAMP-response element binding protein; TLR: Toll-like receptor; RAGE: Receptor for AGEs; AGEs: Advanced glycation end products; RhoA: Ras homolog family member A; Ras: Rat sarcoma; ROCK: Rho-associated coiled-coil-containing protein kinase; TGF: Transforming growth factor; iNOS: Inducible nitric oxide synthase; COX: Cyclooxygenase; CCR: Chemokine (C-C motif) receptor; CCL/MCP: Chemokine (C-C motif) ligand; PGC: Peroxisome proliferator-activated receptor-gamma coactivator; FXR: Farnesoid X receptor; α-SMA: α-Smooth muscle actin; ROS: Reactive oxygen species; GSK-3β: Glycogen synthase kinase 3 beta; SOD: Superoxide dismutase; GSH: Glutathione; CAT: Catalase; HIF: Hypoxia-inducible factor; NOX: NADPH oxidase; IL: Interleukin; TNF: Tumor necrosis factor; FOXO: Forkhead box O; ERK: Extracellular regulated protein kinase; MMP: Matrix metalloproteinase; ER: Endoplasmic reticulum; PERK: Protein kinase R-like endoplasmic reticulum kinase; eIF2α: Phosphorylation of eukaryotic initiation factor-2α; ATF4: Activating transcription factor 4; SIRT1: Silent information regulator 1; TRPC: Transient receptor potential canonical; SCFAs: Short-chain fatty acids; CKD: Chronic kidney disease; IRI: Ischemia/reperfusion injury; ADPKD: Autosomal dominant polycystic kidney disease; UUO: Unilateral ureteral obstruction; UAN: Uric acid nephropathy; CLP: Cecum ligation puncture; DOX: Doxorubicin; DDP: Cisplatin; LPS: Lipopolysaccharide; EG: Ethylene glycol; LN: Lupus nephritis; Ang II: Angiotensin II; HMCs: Human mesangial cells; HK-2 cells: Human kidney-2 cells; Fru: Fructose; TAC: Tacrolimus; PC: Post-contrast; MTX: Methotrexate; ON: Oxalate nephropathy.
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

Zhao, Y.; Song, J.-Y.; Feng, R.; Hu, J.-C.; Xu, H.; Ye, M.-L.; Jiang, J.-D.; Chen, L.-M.; Wang, Y. Renal Health Through Medicine–Food Homology: A Comprehensive Review of Botanical Micronutrients and Their Mechanisms. Nutrients 2024, 16, 3530. https://doi.org/10.3390/nu16203530

AMA Style

Zhao Y, Song J-Y, Feng R, Hu J-C, Xu H, Ye M-L, Jiang J-D, Chen L-M, Wang Y. Renal Health Through Medicine–Food Homology: A Comprehensive Review of Botanical Micronutrients and Their Mechanisms. Nutrients. 2024; 16(20):3530. https://doi.org/10.3390/nu16203530

Chicago/Turabian Style

Zhao, Yi, Jian-Ye Song, Ru Feng, Jia-Chun Hu, Hui Xu, Meng-Liang Ye, Jian-Dong Jiang, Li-Meng Chen, and Yan Wang. 2024. "Renal Health Through Medicine–Food Homology: A Comprehensive Review of Botanical Micronutrients and Their Mechanisms" Nutrients 16, no. 20: 3530. https://doi.org/10.3390/nu16203530

APA Style

Zhao, Y., Song, J. -Y., Feng, R., Hu, J. -C., Xu, H., Ye, M. -L., Jiang, J. -D., Chen, L. -M., & Wang, Y. (2024). Renal Health Through Medicine–Food Homology: A Comprehensive Review of Botanical Micronutrients and Their Mechanisms. Nutrients, 16(20), 3530. https://doi.org/10.3390/nu16203530

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