Next Article in Journal
Association between Single Nucleotide Polymorphisms of SULT1A1, SULT1C4, ABCC2 and Phase II Flavanone Metabolites Excretion after Orange Juice Intake
Previous Article in Journal
Maternal Vitamin B12 Deficiency Detected by Newborn Screening—Evaluation of Causes and Characteristics
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Effects of Genistein on Common Kidney Diseases

1
Institute of Nursing and Health, College of Nursing and Health, Henan University, Jinming Avenue, Kaifeng 475004, China
2
Department of Art, School of Kaifeng Culture and Tourism, Kaifeng 475004, China
3
School of Business, Henan University, Kaifeng 475004, China
4
School of Basic Medical Sciences, Henan University, Jinming Avenue, Kaifeng 475004, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Nutrients 2022, 14(18), 3768; https://doi.org/10.3390/nu14183768
Submission received: 21 July 2022 / Revised: 6 September 2022 / Accepted: 6 September 2022 / Published: 13 September 2022
(This article belongs to the Section Phytochemicals and Human Health)

Abstract

:
Genistein is a naturally occurring phytoestrogen (soy or soybean products) that is classified as an isoflavone, and its structure is similar to that of endogenous estrogens; therefore, genistein can exert an estrogen-like effect via estrogen receptors. Additionally, genistein is a tyrosine kinase inhibitor, which enables it to block abnormal cell growth and proliferation signals through the inhibition of tyrosine kinase. Genistein is also an angiogenesis inhibitor and an antioxidant. Genistein has effects on kidney cells, some of the kidney’s physiological functions, and a variety of kidney diseases. First, genistein exerts a protective effect on normal cells by reducing the inflammatory response, inhibiting apoptosis, inhibiting oxidative stress, inhibiting remodeling, etc., but after cell injury, the protective effect of genistein decreases or even has the opposite effect. Second, genistein can regulate renin intake to maintain blood pressure balance, regulate calcium uptake to regulate Ca2+ and Pi balances, and reduce vasodilation to promote diuresis. Third, genistein has beneficial effects on a variety of kidney diseases (including acute kidney disease, kidney cancer, and different chronic kidney diseases), such as reducing symptoms, delaying disease progression, and improving prognosis. Therefore, this paper reviews animal and human studies on the protective effects of genistein on the kidney in vivo and in vitro to provide a reference for clinical research in the future.

1. Introduction

Genistein, as a functional component existing in food, has received the attention of researchers for attenuating many diseases. Particularly in recent years, its beneficial effects on kidney diseases have constantly been proved in extensive research.
Genistein (4,5,7-Trihydrox-yisoflavone and 5,7-Dihydroxy-3-(4-hydroxyphenyl) chromen-4-one) is a naturally occurring compound that has an isoflavone compound structure. In 1899, genistein was first isolated from dyer’s broom. Its structure was established in 1926. It was chemically synthesized in 1928 [1]. Genistein is extensively distributed in edible plants and medicinal plants, such as a variety of legumes [2,3], Flemingia vestita, and F macrophylla [4,5,6]. It is also found in cell cultures of Maackia amurensis [7]. Additionally, it is a primary secondary metabolite in Trifolium species and Glycine max L [8]. Generally speaking, there are several purification methods for genistein, including maceration extraction, Soxhlet extraction, ultrasound-assisted extraction (UAE), microwave-assisted extraction (MAE), supercritical fluid extraction (SFE), and accelerated solvent extraction (ASE). The content of genistein in various foods is shown in Table 1.
It is well known that genistein is absorbed from plants in the form of glycoside, which is hydrolyzed to genistein though the intestinal microbiota or by phlorizin hydrolase (a small intestine brush-border lactase) [9,10]. Genistein is then absorbed by intestinal microflora in the form of aglycone; its oral bioavailability is approximately 10% [11]. The major signaling pathways involved in genistein metabolism are sulfation and glucuronidation. In addition, metabolites such as O-demethyldaidzein, dihydrogenistein, dihydrodaidzein, dihydroequol, and 6-hydroxy-O-demethylflavin generated by the liver and intestines have also been found in blood and urine [12]. Additionally, the kidneys, lungs, and heart can also effectively metabolize genistein [13]. After metabolism, genistein and its metabolites are completely excreted through the urethra and intestines within 24 h.
Genistein has many biological functions. First, genistein can exert an estrogen-like effect. This is due to the structural similarities between genistein and estrogen, making genistein an estrogen mimic in the body [14]. It mainly acts on estrogen receptors (ERs). However, unlike estrogen, genistein has more potential to act on ER β than ER α. Genistein is also used in treating postmenopausal symptoms, such as reductions in bone mass, vaginitis, and hot flashes [15], based on this effect. Second, genistein is a tyrosine kinase inhibitor. Therefore, genistein can be used to inhibit the development of many types of cancers, including breast cancer [16,17,18], brain cancer [19], colon cancer [17], prostate cancer [16,20], and cervical cancer [21], by blocking cell growth and proliferation signals mediated by tyrosine kinase. In addition, genistein is an angiogenesis inhibitor, as well as an antioxidant, etc. This means that genistein has other therapeutic effects besides those on postmenopausal symptoms and cancers, including on cardiovascular disease [22], obesity [23], and diabetes mellitus [24], as well as antidepressant and anxiolytic effects [25]. Recently, an increasing number of studies have shown that genistein has a variety of protective effects on kidney cells and kidney diseases [26].
Table 1. The content of genistein in various foods.
Table 1. The content of genistein in various foods.
FoodsContent (μg/100 g Hydrated Portion)Reference
Soybean26,800–102,500[27]
Kidney bean18.0–518.0[27]
Chickpea69.0–214.0[27]
Pea0–49.7[27]
Lentil7.0–19.0[27]
Kudzu leaf2520[27]
Kudzu root12600[27]
Black gram1900[28]
Alfalfa5.0[28]
Peanut8.0[29]
Caraway seed64.0[28]
Sunflower seed13.9[30]
Barley7.7[28]
Broccoli8.0[28]
Cauliflower9.0[29]
The kidney is one of the most important organs: it is involved in the homeostasis of the whole body, such as regulating the acid–base balance, electrolyte concentrations, osmotic pressure, and blood pressure, eliminating toxins, etc. Additionally, the kidneys can also produce and secrete a variety of endocrine hormones, including renin, erythropoietin, and calcitriol. There are different kinds of kidney diseases treated in the clinic. When kidney diseases occur, they can cause serious harm to the body’s functioning and even lead to death. In the clinical setting, there are many methods for treating kidney diseases, but the vast majority of kidney diseases are irreversible and difficult to cure, even with high-cost treatment. Therefore, it is extremely necessary to develop new strategies to prevent the occurrence and development of kidney diseases. Therefore, it is urgent to summarize these data (human and animal studies) to inspire future research and possible clinical applications of genistein to attenuate kidney diseases.

2. The Role of Genistein on Pathologies of Kidney Cells

2.1. The Effects of Genistein on Mesangial Cells

Glomerular mesangial cells (MCs) lie between and around the glomerular capillaries, creating a support structure for the tuft of capillaries, and are involved in the production of inflammatory mediators, such as cytokines, macromolecules, and immune complexes. Additionally, MCs have a contractile function, and under the influence of vasoconstrictors and vasodilators, they affect filtration by changing the surface area of the filtration slits through contraction and relaxation [31]. In the presence of pathological conditions (e.g., growth factors, inflammation, glomerular capillary hypertension, and glucose toxicity), the mesangial cells are activated, resulting in a glomerular pathology [32].
Previous studies have shown that genistein can inhibit the abnormal activation of mesangial cells in vitro, potentially preventing the occurrence of kidney injuries [26]. Inhibition of inflammation or proliferation by impeding the production and effect of proliferation stimulators (e.g., EGF and PDGF) and inflammatory factors (e.g., TGF-β, IL-1β, and PGE2) is considered to be involved in the effect of genistein, as summarized in a previous review [26]. However, the related mechanisms were not further discussed, except for the ability of genistein to inhibit tyrosine kinase. In recent years, a large number of studies have further expanded the understanding of the effects of genistein on MCs, and the relevant mechanisms have also been deeply explored (Figure 1). First, genistein is able to inhibit inflammation in MCs via the suppression of interleukin-1 beta (IL-1β)/MCP-1, NF-κB (nuclear factor-kappaB)/matrix metalloproteinase-9, and Gro chemokine transcription, and increasing group II phospholipase A2 transcription (PLA2) [33,34,35,36] prevented the activation of the glutamine:fructose-6-phosphate amidotransferase (GFAT) promoter and disrupted AGE-RAGE binding [37,38]. Second, genistein can inhibit the abnormal proliferation of MCs through multiple pathways, including by inhibiting the synthesis of DNA [39,40] and calcium (Ca2+) accumulation, which impedes proliferation-stimulating receptors (e.g., 5-HT2A receptors and erythropoietin) [41,42] or the formation of complexes with other components (e.g., PLC-γ1 and PDGF-β receptor membrane complex; growth factor receptor-binding protein 2, son of sevenless, and PDGF-β receptor complex) [43,44,45], preventing the internalization of the angiotensin II receptor and its downstream responses (e.g., PAI-1 mRNA; PLC-gamma 1/IP3 and Ca2+) [46,47,48,49]; attenuating vascular permeability factor (VPF)/cGMP and reducing MAPK signals through the inhibition of ERK2/Elk/(AP-1 and Fos) [50,51], PTK/PKC-Ras--MAP kinase activity, and autophosphorylation of pp60c-src [52,53,54,55]; and preventing the secretion and expression of TGF-β1 [56,57]. Third, genistein can restrain the pathological apoptosis of MCs by reducing the activation of c-Jun phosphorylation (SAPK) [58,59] by angiotensin II/(ATP and UTP) and nitric oxide. The production of nitric oxide can also be suppressed by genistein [60,61]. Moreover, genistein can inhibit remodeling, which results from inhibited collagen gel contraction, reconstruction, and degradation by impeding ERK and collagenase mRNA [62,63]. For collagen synthesis, low-dose genistein mimics the effect of estrogen through estrogen receptors to inhibit the synthesis of collagen in mesangial cells rather than by inhibiting tyrosine kinase, which is effective for inhibiting remodeling [64]. High-dose genistein promotes collagen synthesis mainly by inhibiting tyrosine kinase and collagen degradation, which is detrimental to inhibiting remodeling [65]. Therefore, the dose relationship should be considered when genistein is used to remodel MCs. Therefore, genistein can block the development of kidney diseases by improving the function of mesangial cells.

2.2. The Effects of Genistein on Endothelial Cells

As is well known, the urinary function of the kidney is accomplished by the nephron and the collecting duct. Nephrons are tiny or microscopic structural and functional units of the kidneys. They consist of a kidney corpuscle and a kidney tubule. The kidney corpuscle consists of a cluster of capillaries called a glomerulus and a cup-shaped structure called Bowman’s capsule. The kidney tubules extend from the capsule. The capsule is connected to the tubule and is composed of epithelial cells with a lumen. The function of the kidney corpuscle is to filter the original urine, while the kidney tubule is responsible for processing and removing the filtered fluid. The collecting duct system consists of a series of tubules and ducts that connect the nephron to the calyx or directly to the kidney pelvis. Collecting tubes participate in the balance of electrolytes and liquids through reabsorption and excretion. Studies have shown that genistein plays a role in nephrons and collecting ducts, including glomeruli, proximal tubule cells, distal tubule cells, and collecting duct cells (Figure 2).
In glomerular endothelial cells, genistein can inhibit inflammation caused by GEC MCP-1 mRNA expression induced by LysoPC [66]. In the proximal tubule, genistein plays a more diverse role. Genistein can not only protect dopamine precursors, reduce insulin uptake, and increase insulin accumulation in proximal tubular cells but also protect dopamine receptors, prevent their serine phosphorylation, and inhibit their mediated NKA activity [67,68,69,70], thereby maintaining kidney function and reducing the occurrence of disease. Additionally, genistein can inhibit the Na+-glucose cotransporter (e.g., IL-6) and induce the Na+-glucose cotransporter (e.g., Ang II and EGF); the former may be due to the inhibition of abnormal proliferation induced by inflammation [71,72,73]. Other mechanisms include suppressing the Na+/H+ exchanger (NHE, including NHE1/3), which can balance proliferation, differentiation, and cell forms [74,75,76]; restrain inflammation (↓MCP-1) and downstream signals (↓HIF-1) [77,78]; block DNA synthesis (e.g., thrombin and HGF) and DNA damage (hypoxia or chemical) [77,79]; prevent PAI-1 (induced by Ang II and 15d-PGJ2) production to reduce the emergence of fibrosis [80,81]; inhibit HSP70 after heat shock and prevent the disorder of normal protein formation caused by its excess [82]; inhibit protein hyperphosphorylation (β-catenin and plakoglobin phosphorylation) to maintain tight junctions [83]; restrain PAH, L-alanine transport (↑EGF), H(+)-ATPase (Ang II), and Pi, acting as a tyrosinase inhibitor, indicating that they all need tyrosinase activation (but with regard to PI uptake, genistein blocks the inhibition of EGF on its uptake, because EGF inhibits Pi uptake by activating EGF receptors) [84,85,86,87,88]; block the decrease in Mg2+; and prevent the production of nitric oxide [89]. In distal tubules, genistein mainly regulates ions and maintains the acid–base balance, e.g., by decreasing Na+ transport (e.g., hyposmolality, insulin, and aldosterone [90,91,92]) and HCO3- (NGF and hyposmolality [93,94]). In collecting ducts, genistein can inhibit the excessive activity of H-K-ATPase induced by isoproterenol and maintain the balance of H+-K+ exchange [92], impeding apoptosis in hypertonic conditions (NaCl and urea) [95], suppressing the expression of COX-2 to maintain the stability of blood volume [96], and increasing the activity of SMIT (sodium/myo-inositol cotransporter) so as to increase the activity of cells [97]. Additionally, genistein can inhibit IGF-I-induced (insulin-like growth factor-I) nitric oxide in endothelial cells of interlobular vessels [98]. However, the effects of genistein on kidney tubules and collecting ducts are not all favorable. For example, in proximal tubule cells, genistein blocks the stimulating effect of Ang II on the expression of the Pax-2 gene, which plays an important role in kidney repair (lack of expression leads to apoptosis in cells [99]). In the distal tubule, genistein reduces the synthesis of sulfoglycolipid, which can protect cells from a hypertonic environment [100]. This indicates that it is necessary to further study the effects of genistein on kidney tubules and collecting ducts.

2.3. The Effects of Genistein on Podocytes

Podocytes are cells that wrap around glomerular capillaries and are located in the kidney’s Bowman’s capsule, forming the epithelium of Bowman’s capsule, which is the third layer of blood filtration [101]. Studies have shown that the decrease in or depletion of podocytes and their morphological changes play an important role in the development of progressive nephropathies, such as glomerulosclerosis and diabetic nephropathy [102,103,104]. A number of studies have shown that genistein has multiple favorable and unfavorable effects on podocytes (Table 2). The beneficial effects include the inhibition of inflammation (EVs and IL-1β) and the promotion of autophagy. The mechanism is the reduced activation of the inflammasome via the inhibition of TGβ1/FAK in normal podocytes and the induction of autophagy through the inactivation of mTOR in HG (high-glucose)-treated podocytes. The unfavorable effect is the promotion of the loss of podocytes under the effect of fluid shear force and the apoptosis of podocytes in the presence of integrinα3β1, the reorganization of the actin cytoskeleton, and circular ruffles. The mechanism of the latter effect inhibits FAK/integrinα3β1–ECM interaction. The double-sided effect of genistein on podocytes indicates that genistein should be used with caution; we can increase the use of genistein in terms of its known beneficial effects and avoid unfavorable factors so as not to cause greater damage to the kidneys.

3. The Effects of Genistein on Kidney Physiology (Figure 3)

3.1. The Effects of Genistein on Renin

Renin, which is secreted by granule cells of the paracellular apparatus in the kidney, participates in the activation of the renin–angiotensin–aldosterone system and regulates the mean arterial blood pressure. Studies have shown that genistein may be able to maintain the balance of blood pressure by affecting the secretion of renin. In a previous study [110], researchers found that genistein treatment suppressed the IL-1β-mediated attenuation of renin gene transcription in vitro. In another in vitro study on the effect of epidermal growth factor (EGF) on the secretion of renin, the researchers observed that genistein treatment eliminated the inhibitory effect of EGF on renin secretion [111]. Therefore, genistein may be used to maintain normal blood pressure by restoring renin release, which is suppressed in some conditions, such as inflammation (the former study), and endogenous vasoconstrictors (e.g., EGF). However, more studies are needed to explore the mechanism.

3.2. The Effects of Genistein on Regulating Calcium and Phosphate

Ca2+ and phosphate (Pi) are two key factors in maintaining the balance of bone formation and bone loss as well as other physiological functions (muscle contraction, blood coagulation, and neurotransmitter release [112]). When the balance of calcium is disrupted, bone pathologies or other dysfunctions in the body will occur. Two studies explored the protective effect and potential mechanism of genistein on the dynamic balance of calcium in male animals. A study showed that genistein treatment (30 mg/kg b.m/day) decreased Ca2+ content in urine and increased 25 (OH) vitamin D content in serum. Additionally, increased expression of the Klotho gene and protein also contributes to calcium reabsorption, possibly activating the TRPV5 Ca2+ channel in the kidneys of orchidectomized rats (an andropause model) [113]. Additionally, the downregulated expression of FGFR and PTH1R reduces Pi reabsorption via the inhibition of the activity of the NaPi 2a cotransporter. However, in another study, genistein treatment (10−6 M) eliminated the effect of testosterone-enhancing kidney Ca2+ reabsorption in the distal luminal membrane of rabbit kidneys [114]. Therefore, the dual effects of genistein in regulating Ca2+ and Pi balances depend on the endocrine hormone (e.g., testosterone). Thus, genistein may be used carefully in regulating Ca2+ under different conditions.

3.3. The Diuretic Effect of Genistein

Recent studies have shown that genistein has a diuretic effect by decreasing vasodilatation instead of decreasing the glomerular filtration rate. A study [115] showed that there is a diuretic effect of genistein (15 mg/kg) in rats, which is similar to that of furosemide. However, the effective dose was 3–5 times lower than that of furosemide (a diuretic). However, genistein had no significant effect on the glomerular filtration rate, despite decreasing kidney vascular resistance. While the diuretic mechanism of genistein has not been further explained, another study showed that the diuretic effect of genistein is due to the reduction in the membrane Na-K-Cl cotransporter concentration [116]. Accordingly, genistein may be used as a new diuretic in future treatment, and more in-depth clinical research is needed in the future.

3.4. The Effects of Genistein on Nephron Barrier

It is well known that the filtration function of the kidney depends on the normal kidney permeability barrier. However, when the permeability is pathologically damaged, pathological filtration dysfunctions occur, e.g., increased or decreased filtration. Studies have shown that genistein protects the permeability barrier of the kidney. In one study [117], genistein treatment (100 μM, 1 h) significantly inhibited the increase in Pa (albumin permeability) in acute glomerular inflammatory injury induced by SNAP (s-nitroso-N-acetyl-penicillamine) in rats in a dose-dependent manner. This effect may be achieved by inhibiting tyrosine kinases. However, another study found the opposite results that genistein (50 or 100 μM: 6 h) was not conducive to the repair (the relocalization of ZO-1 and occludin to the tight junction) of kidney tubular tight junction damage caused by oxidative stress and decreased ATP [118]. The differences in the effect of genistein may be due to the fact that genistein can protect the kidney from the acute kidney injury barrier when it is not seriously damaged, but it is not conducive to weakening the silent junction damage that has already occurred.
Figure 3. The effects of genistein on kidney physiology. IL-1β: interleukin-1 beta; BP: blood pressure; FGFR: fibroblast growth factor receptor; PTH1R: parathyroid hormone 1 receptor; TRPV5: transient receptor potential vanilloid 5; Pa: albumin permeability, ↓: inhibit; ↑: upregulate.
Figure 3. The effects of genistein on kidney physiology. IL-1β: interleukin-1 beta; BP: blood pressure; FGFR: fibroblast growth factor receptor; PTH1R: parathyroid hormone 1 receptor; TRPV5: transient receptor potential vanilloid 5; Pa: albumin permeability, ↓: inhibit; ↑: upregulate.
Nutrients 14 03768 g003

4. The Effects of Genistein on Common Kidney Diseases

4.1. The Effects of Genistein on Acute Kidney Injuries

4.1.1. LPS

Lipopolysaccharide (LPS), which is a component of the outer membrane of Gram-negative bacteria, can stimulate the innate immune response. Additionally, LPS, as a special antigen [119], is able to induce endotoxemia [120], sepsis [121], and even DIC [122]. Therefore, LPS is usually used to induce different animal models, such as neuroinflammation [123], endometritis [124], memory impairment [125], and acute kidney injuries [126]. LPS is co-sensed by the CD14 protein and TLR4-MD2 complex at the plasma membrane [127]. Ligated TLR4 then activates the transcription of immune factors (e.g., cytokines and chemokines) mediated by NF-κB and IRF3 through MYD88 and TRIF. This subsequently upregulates the expression of various inflammatory mediators, such as tumor necrosis factor α (TNF-α), interleukin-6 (IL-6), and IL-1β.
Several studies have shown that genistein treatments can significantly improve acute kidney injuries caused by LPS in vitro and in vivo. For example, an animal experiment showed that genistein pretreatment (10 mg/kg) dramatically suppressed fractalkine expression through LPS-mediated TNF-α in the arterial endothelial cells of rat kidneys [128]. In addition, in another animal experiment [129], genistein (10, 30, and 90 mg/kg injected i.p. 0.5 h before the LPS injection and 2 h and 8 h after the LPS injection) significantly improved the morphological structure, fiber protein deposition, function indicators (inhibition of blood urea nitrogen, BUN), and inflammation reaction (↓NF-κB, IL-6, and IL-1) in kidneys.
Additionally, in a clinical study [130] of hemodialysis patients with end-stage kidney disease, genistein treatment (25 μM for 24 h) inhibited LPS-induced inflammation activation (downregulated TNF-α levels) in whole blood and monocytes in vitro. Therefore, genistein may be used to attenuate acute kidney injury induced by LPS. Further clinical experiments need to be carried out.

4.1.2. The Effects of Genistein on Kidney Ischemia/Reperfusion Injury

Ischemia refers to the partial or complete occlusion of blood perfusion in tissues or organs. It can lead to a lack of oxygen and other nutrients as well as the accumulation of metabolic wastes. If perfusion is not restored in time, ischemia can quickly lead to tissue necrosis [131]. Early reperfusion is the preferred intervention to prevent pathological processes. However, reperfusion after ischemia often causes inflammation and apoptosis through oxidative stress [132]. Kidney I/R injury usually occurs due to shock [133], sepsis [134], low perfusion [135], and kidney transplantation [136]. Kidney I/R injury will cause direct damage to tubules and blood vessels. Then, glomeruli acute kidney injury will occur as a result of oxidative stress, inflammation, and mitochondrial dysfunction, leading to a sharp decrease in kidney function and even an increase in mortality [137]. As the kidneys maintain the water balance of the human body, its I/R process can also cause severe damage to other organs [138,139].
Several studies have shown that under normal circumstances, genistein can inhibit the contraction of kidney blood vessels, including glomerular arterioles, afferent arteries, or vascular beds [140,141,142]. In addition, genistein can inhibit norepinephrine-induced vasoconstriction, but this effect is not completely dependent on extracellular Ca2+ [143]. It may be partly related to the inhibition of the large-conductance K+ channel, according to another study [144]. Therefore, genistein may attenuate kidney ischemic injury by inhibiting the contraction of kidney blood vessels. For instance, in one study [145], genistein treatment (3 μ for 45 min) significantly inhibited kidney vasoconstriction induced by G1 (a G protein-coupled estrogen receptor1 agonist) in isolated perfused (perfusion immediately after separation) rats. The effect of genistein is exerted through the blocking of the estrogenic effect. However, in another study [146], genistein (10 mg/kg, 30 min before hemorrhagic shock) reduced kidney injury during ischemic shock in rats by inhibiting inflammation. However, the effect of genistein on vasoconstriction was not studied.
Encouragingly, some studies have shown that genistein pretreatments at a shorter time before I/R can attenuate kidney I/R injury in animal models. One study showed that genistein treatment (5, 10, or 15 mg/kg, 30 min before ischemia) significantly reduced kidney cell death through the stimulation of kidney cell proliferation mediated by the up-regulation of SIRT1 expression (exerting cytoprotective effects) in kidney I/R mice (45 min/24 h) [147]. In another study, genistein (15 mg/kg body weight, i.p.), administered 30 min before ischemia and 1 h after ischemia, reduced kidney injuries in kidney I/R (45 min/24 h) rats by reducing the inflammation response (decreasing TLR-4 and TNF-α expression) and oxidative stress (intensifying antioxidant ability) [148]. However, in a previous study [149] exploring the effects of EPO on kidney ischemic injury in ischemic shock mice, genistein treatment (10 mg/kg, 2 h before ischemia) alone increased kidney ischemic injury in model mice, and treatment could also reverse the beneficial effects of EPO in attenuating the kidney injury of model mice. The adverse effect in the latter study may be due to the time point of genistein administration; that is, genistein administration before ischemia may be the reason behind this. The exact mechanism still requires further investigation in the future.

4.2. The Effects of Genistein on Kidney Cancer Cells

Renal cancer affects nearly 300,000 people worldwide each year, causing more than 100,000 deaths annually [150]. However, owing to the lack of obvious symptoms in the early stage, only 30% of patients are diagnosed according to clinical symptoms in the advanced stage, including local symptoms (acute or chronic flank pain, gross hematuria, and palpable abdominal mass and varicocele) and paraneoplastic disorders (e.g., hematuria, high blood pressure, anemia, cachexia, weight loss, hypercalcemia, etc.) [151]. Increasing 5-year survival rates after diagnosis required the development of new drugs for systemic treatment. In recent years, research on the effect of genistein on renal cell carcinoma has mainly been focused on human kidney cancer cells. A few studies have conducted animal experiments. The main effects of genistein on kidney cell carcinoma are the decrease in the proliferation and migration of cancer cells, the inhibition of the neovascularization of solid tumors, the retardation of the growth of RCC cells, the induction of cell apoptosis, and the induction of tumor growth. These effects are achieved through the following mechanisms (Table 3): inhibiting cancer cell proliferation by ① increasing the expression of CDKN2a and CDKN2a methylation, ② suppressing EED (embryonic ectoderm development) levels in PRC2, ③ inhibiting HOTAIR (HOX transcript antisense RNA)/PRC2 (polycomb repressive complex 2) interaction, ④ suppressing HOTAIR/PRC2 recruitment to the ZO-1 promoter and enhanced ZO-1 transcription, and ⑤ inhibiting SNAIL transcription by reducing HOTAIR/SMARCB1 (subfamily B member 1) interaction and decreasing EGF; impeding angiogenesis by decreasing the expression of the angiogenic factors vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (FGF); suppressing miR-1260b expression (in A-498 cells, inhibiting Wnt signaling), which is highly expressed in kidney cancers; reducing the basal activity of sulfotransferase; and inducing cell apoptosis.

4.3. The Effects of Genistein on Diabetic Nephropathy

Diabetic nephropathy is one of the most common and serious complications of diabetes, which is related to an increase in morbidity and mortality in patients with diabetes [162]. It is considered to be one of the leading causes of chronic kidney disease (CKD) and end-stage renal disease (ESRD) worldwide. Therefore, diabetic nephropathy has become a global health problem [163,164]. The etiology of diabetic kidney injury is complex and has multiple factors, including hyperglycemia, hypertension, dyslipidemia, the production of inflammatory cytokines, and oxidative stress [165]. Some studies have suggested that genistein may be a good protective agent against diabetic nephropathy in type 1 and 2 diabetes models (Table 4). The mechanism of genistein to improve diabetic nephropathy mainly includes the following aspects: inhibiting AGE (advanced glycation end product) formation by trapping MGO to form adducts and upregulating the expression of glyoxalase I and II and aldose reductase (AR); improving levels of fasting blood glucose (FBG) levels; reducing kidney inflammation (decreasing levels of interleukin-6, TNF-α, and C-reactive protein; attenuating kidney oxidative stress (nuclear-related factor E2, heme oxygenase-1/HO-1, glutathione peroxidase, and superoxide dismutase isoforms); inhibiting fibrosis-related markers (protein kinase C, protein kinase C-beta II, and transforming growth factor-beta I); and reducing apoptosis and normalizing vasoconstriction induced by agonists (norepinephrine, endothelin-1, and Ang II).
Additionally, a recent in vitro study [166] showed that genistein can block the process of decoy receptor 2 interacting with peroxiredoxin 1, which may be related to the amelioration of kidney fibrosis in diabetic nephropathy progression. Another study [167] found that genistein had an inhibitory effect on AR purified from sheep kidneys, which plays a vital role in the development of diabetic nephropathy. Genistein also had synergistic antioxidant effects, alongside other substances (e.g., resveratrol), on high-glucose-incubated Madin-Darby canine kidney (MDCK) epithelial cells [168]. The mechanism was the reduction in ROS via the inhibition of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase expression and increased γ-glutamylcysteine synthetase expression. The genistein–chromium (III) complex can also improve pathological injuries in the kidneys of diabetic mice, which provides a new way to apply genistein treatment [169]. Therefore, the combined use of genistein may have better prospects.
In addition, excessive sugar intake can induce multiple metabolic syndromes, such as insulin resistance, hyperglycemia, hypertriglyceridemia, hypertension, and also kidney damage associated with oxidative stress. Genistein treatment (1 mg/kg/day, i.p., for 60 days) significantly improved insulin sensitivity, glomerular function (increased clearance rates), and kidney parenchymal injury, as confirmed histologically, in a rat model of kidney injury induced by a high-glucose diet [170]. Additionally, genistein treatment can also impede the occurrence of nephropathy through a high-fructose diet. However, genistein treatment (2, 6, and 20 mg/kg for 90–180 days) had little effect on the diabetic nephropathy of females, according to a previous study [171]. Although a genistein-containing diet could protect female mice from developing type 1 diabetes, only kidney weights were increased, and the histopathology of kidneys was not ameliorated. This may be because the female rats used in this study with a certain amount of estrogen in their bodies altered the effect of genistein.
Table 4. The effects of genistein attenuating diabetic nephropathy.
Table 4. The effects of genistein attenuating diabetic nephropathy.
AnimalDiabetes ModelsTreatments (Genistein)Effects and MechanismsRef.
C57BL/6J miceHigh-fat diet0.25% genistein in diet for 18 weeksInhibiting AGE formation by trapping MGO to form adducts and upregulating the expression of glyoxalase I and II and aldose reductase in kidney to detoxify MGO[172]
Albino ratsAlloxan-induceddiabetes20 mg/kg/day for 30 dNormalizing kidney function (biomarkers: creatinine and BUN) by downregulating inflammatory responses (IL-6, TNF-α, and C-reactive protein in serum)[173]
MiceStreptozotocin-induced diabetes10 mg/kg, i.p. three times a week for 10 weeksReducing kidney inflammation, oxidative stress, and apoptosis[174]
ICR miceAlloxan-induced diabetes0.25 and 1 mg/g in diet for 2 weeksImproving levels of FBG and attenuated kidney oxidative stress; decreasing inflammatory and fibrosis-related markers[175]
Wistar ratsSTZ-induced diabetes4 mg/kg b.w/day, i.p. for 7 dProtecting against kidney dysfunction, lowering blood glucose levels, and protecting against kidney dysfunction[176]
KKAy mouseType 2 diabetes12 mg /kg, oral gavage, once a day for 3 monthsInhibiting inflammatory responses, repressing HGA-induced activator protein 1 activation and oxidase stress generation, and reducing NADPH oxidase (NOX) gene expression[177]
Wistar ratsSTZ-induced diabetes1.5 mg/kg/alt diem for 4 weeksNormalizing vasoconstriction induced by agonist (norepinephrine, endothelin-1, and Ang II)[178]

4.4. The Effects of Genistein on Hypertensive Kidney Disease

At present, hypertension affects approximately 30% of the general population and causes damage to multiple organs [179]. It is worth noting that hypertension is a risk factor for the progression of kidney damage (hypertensive kidney disease) [180]. Hypertensive kidney disease is considered to be one of the consequences of long-term and poorly controlled hypertension. In turn, kidney disease can cause or aggravate hypertension. Hypertensive kidney disease is a leading cause of end-stage kidney failure, second only to diabetic kidney disease [179]. Many studies focus on how to reduce hypertension. Therefore, several studies have shown that genistein treatment is able to improve hypertension and kidney injuries in hypertensive animal models (Table 5), which is mainly through the reduction in blood pressure and kidney vascular tension. The mechanisms are as follows: lowering BP (restored ACE, PKC-βII and eNOS/NO, and cGMP); blunting the dose-related increase in arterial pressure; and reducing kidney vascular resistance relative to vehicle in isolated perfused kidneys. Therefore, genistein can protect the kidney by reducing kidney hypertension and maintaining the ultrastructural integrity of the kidney. It may provide a therapeutic option for the treatment of kidney hypertension in the future.

4.5. The Effects of Genistein on Kidney Injury by Medications and Irradiation (Table 6)

Medications are a relatively common cause of kidney injury [184,185,186]. At present, the epidemiology of drug-induced nephrotoxicity is mainly based on the AKI (acute kidney injury)-related literature. Drug-induced nephrotoxicity in adults was approximately 14–26%, while in the pediatric population, 16% of AKI hospitalizations were caused by drugs [185,186]. Common nephrotoxic drugs include cisplatin, cephaloridine, and gentamicin. Cephaloridine can produce dose-related nephrotoxicity when administered in high doses [187]. Its nephrotoxicity can be distinguished by the degree of acute proximal tubular necrosis in laboratory animals and humans [188]. A study showed that genistein (25 μg/mL, 2 h) inhibited increases in LDH leakage (an index of cellular injury) and lipid peroxidation in LLC-PK1 cells exposed to cephaloridine [189]. Gentamicin, an aminoglycoside antibiotic, is highly effective in the treatment of severe Gram-negative infections. However, it has nephrotoxic effects on the epithelial cells of the proximal tubules. Genistein treatment (10 mg/kg/day i.p.) produced reno-protective effects (decreased serum levels of Kim-1, cystatin C, LDH, and GGT) in gentamicin-induced acute kidney injury [190]. Cisplatin is one of the most effective and active cancer chemotherapy drugs for the treatment of various malignancies [191], inducing nephrotoxicity mainly through the reaction of platinum and thiol protein groups [192]. Genistein treatment (10 mg/kg/day orally for 3 days) inhibited oxidative stress (reduced reactive oxygen species production) and the inflammation response by decreasing the expression of intercellular adhesion molecule-1 (ICAM) and monocyte chemoattractant protein-1 (MCP-1) proteins and the translocation of the p65 subunit of NF-κB [193]. Genistein also decreased cisplatin-induced apoptosis (regulating p53 induction in the kidney). Therefore, genistein may protect the kidney from drug-induced nephrotoxicity.
Radiation therapy (RT) is one of the most common and important cancer treatment techniques [194]. As a radiosensitive organ, the kidney is inevitably exposed to radiation in the abdominal cancer treatment room. High-dose radiation can cause kidney damage (even radiation nephropathy), including increased vascular permeability, perfusion disorders, inflammation, and fibrosis [195,196]. In a previous study, genistein treatment 24 h before RT decreased the incidence of kidney tubular atrophy and the level of malondialdehyde (MDA) in mouse kidneys. Therefore, genistein supplementation prior to irradiation can be used to protect mice against radiation-induced nephrotoxicity.
Table 6. The effects of genistein attenuating injury caused by medications and irradiation.
Table 6. The effects of genistein attenuating injury caused by medications and irradiation.
AnimalModelTreatments (Genistein)Effects and MechanismsRef.
LLC-PK1Cephaloridine-induced kidney injury25 µg/mL preincubated for 2 hInhibiting increases in LDH leakage and lipid peroxidation in LLC-PK1 cells exposed to cephaloridine[189]
Sprague-Dawley ratsp-Nonylphenol-induced polycystic kidneys0.005 μM/10 μL for 35 dModulating the development of PKD induced by dietary NP in rats[197]
MiceCisplatin-induced kidney injury and cisplatin-treated normal human kidney HK-2 cells10 mg/kg orally once a day for 3 dDecreasing oxidative stress (reactive oxygen species), inflammation (ICAM, MCP-1, and NF-κB), and apoptosis (regulating p53 induction)[193]
Wistar albino ratsGentamicin-induced acute kidney injury rats10 mg/kg/day, i.p, one week before gentamicin treatment, for 17 dDecreasing serum levels of Kim-1, cystatin C, LDH, and GGT[190]
Swiss albino miceA single dose of 6 Gy γ-radiation (Co60)200 mg/kg, subcutaneous injection, for 24 weeksDecreasing the incidence of kidney tubular atrophy and the level of MDA[198]

4.6. The Effects of Genistein on Kidney Fibrosis

Kidney fibrosis, an inevitable consequence of chronic kidney disease [199], is caused by multiple diseases, such as diabetes, obstructive urinary tract, glomerulonephritis, glomerulosclerosis, kidney hypertrophy, and mutation [200]. In the process of kidney fibrosis, many events in kidney cells occur, including inflammation, oxidative stress, fibroblast activation and the expression of fibrosis-related cytokines, vascular remodeling and hypertension, kidney tubular apoptosis, and autophagy [201]. Ultimately, this leads to the loss of kidney function and the replacement of kidney parenchyma by scar tissue [202].
In order to reduce kidney fibrosis, it is important to find novel and reliable therapeutic methods. Several experiments have shown that genistein can improve kidney fibrosis in vivo and in vitro. In vivo, genistein treatment can significantly alleviate fibrosis in several animal models of kidney interstitial fibrosis, streptozotocin-induced type 1 diabetes, and fibrosis induced by a standard pellet diet with high fructose and UUO. The mechanism includes decreasing the proliferation of connective tissue collagen (Table 7); inhibiting oxidative stress by activating the Nrf2-HO-1/NQO1 pathway; inhibiting kidney fibrosis by suppressing TGF-β1/Smad3; increasing kidney ALKBH5 expression; reducing RNA m6A levels; restoring Klotho; and decreasing α-SMA expression. In vitro, genistein blocks kidney transdifferentiation and epithelial-to-mesenchymal transition (inhibits α-SMA and CTGF expression and restores E-cadherin expression) in human kidney tubular epithelial cells. Therefore, it is suggested that genistein has potential anti-kidney fibrosis effects, and further clinical research is needed.

4.7. The Effects of Genistein on Nephrotic Syndrome

Nephrotic syndrome is characterized by a series of symptoms, such as edema, proteinuria, hypoalbuminemia, and hyperlipidemia, and is common in children and adolescents [208]. However, when nephrotic syndrome is serious, it can cause harm to other organs of the body and even endanger life. Therefore, it is necessary to propose a new method for the treatment of nephrotic syndrome. A previous study [209] showed that genistein significantly increased the scores of pathological examinations by increasing the total antioxidant capacity and catalase activity and decreasing the contents of protein carbonyl and MDA in the kidneys of nephrotic model rats. In addition, the proliferation of the WEHI-164 kidney fibrosarcoma cell line in vitro was also inhibited by genistein in the study. Therefore, the result suggests that genistein may be used to reduce the symptoms of nephrotic syndrome in the future.

4.8. The Effects of Genistein on Menopausal Kidney Injury

Menopause is the period when a woman’s menstruation stops permanently, resulting in infertility. It usually occurs between the ages of 48 and 52. For those who have their uterus removed but their ovaries retained, hormone (e.g., estrogen) levels drop sharply, followed by menopause symptoms, usually including obesity; a metabolic imbalance in some tissues, such as the liver, bones, nervous system, and skeletal muscle [210]; and damage to the kidney tissue (e.g., glomerular and tubulointerstitial damage, even leading to diabetic nephropathy) [211]. Since genistein can exert estrogen-like effects, a previous study [212] found that genistein treatment (0.1% in the diet for 4 weeks) reduced insulin resistance and fat accumulation, promoted lipid metabolism, and improved abnormal protein expression induced by oxidative stress in ovariectomized rats fed a high-fat diet. Therefore, genistein can be considered to attenuate the kidney injury of climacteric women.

4.9. The Effects of Genistein on Aging-Induced Kidney Injury

Aging is a natural process that represents the cumulative changes in a person’s state over time, including a decline in physical functions, as well as disorders in psychological and social states, which eventually lead to the emergence of disease and death [213]. Previous studies have shown that genistein can improve aging-related diseases (e.g., bone loss [214] and Alzheimer’s disease [215]). According to a previous study [216], genistein treatment (2 and 4 mg/kg/day for 10 days) can also improve aging-induced kidney injury in male rats by decreasing age-related NF-κB activity (activated by angiotensin II during senescence) and the expression of downstream pro-inflammatory genes, which indicated that genistein had an obvious anti-inflammatory effect by inhibiting the activation of NF-κB induced by angiotensin II during aging. However, there is no relevant research on the effect of genistein on female aging (except for menopausal kidney injury), so it may be further discussed in the future.

5. The Mechanism of Genistein Actions in Kidney

Kidney diseases are very harmful to the human body, and there is no specific treatment, so it is necessary to find better functional components to delay the occurrence and development of kidney disease. As a phytoestrogen and isoflavone found in soybeans, genistein treatment can attenuate many kidney diseases (e.g., acute kidney injuries, kidney cancer, diabetic nephropathy, hypertensive kidney disease, kidney fibrosis, nephrotic syndrome, menopausal kidney damage, and aging-induced kidney injury).
The beneficial effects of genistein are mainly exerted through the following mechanisms: ① It reduces the inflammatory response by downregulating a variety of inflammatory factors (including IL-1, IL-6, NF-κB, TNF-α, TLR-4, and C-reactive protein). ② It inhibits oxidative stress (protein carbonyl, MDA, and Nrf2-HO-1/NQO1) that causes the oxidation of DNA, proteins, and lipids, thus affecting their structure and function. ③ Genistein can decrease arterial pressure through the inhibition of ACE, PKC-βII, NO/NOS, and vasoconstriction. ④ AGEs are formed through the non-enzymatic glycosylation of amino groups on proteins by reducing sugars or dicarbonyl [217], which is related to the pathogenesis of complications (e.g., kidney) of type 2 diabetes mellitus [218], and can be inhibited by genistein via the upregulation of glyoxalase I/II, AR, and FBG. ⑤ It reduces fibrosis by increasing ALKBH5 and Klotho and decreasing α-SMA and CTGF. ⑥ It inhibits apoptosis and promotes cell apoptosis through the inhibition of SIRT1 expression in common kidney diseases. ⑦ It inhibits proliferation and promotes apoptosis by depressing EED, HOTAIR/PRC2, SNAIL, and EGF and increasing CDKN2a in cancer. ⑧ It inhibits angiogenesis through the inhibition of VEGF and FGF in kidney cancer (Figure 4).

6. Conclusions

Genistein has many effects. For example, it can inhibit fibrosis; mesangial dilation; oxidative stress; and inflammatory cytokines, inhibiting mesangial dilation, inflammatory cytokines, oxidative stress, fibrosis, apoptosis, etc. In addition, genistein also has beneficial effects (prevents structural changes and antioxidation, improves activity, and reduces pathological damage) on a variety of cells in the kidney (e.g., mesangial cells, endothelial cells, and podocytes).
Genistein also plays an important role in the treatment of kidney disease. The effects include the modulation of renin release, calcium and phosphorus balance, excessive LPS, and kidney damage caused by a variety of acute and chronic diseases. In animal model experiments for many diseases, genistein reduced kidney damage caused by ischemia, reduced drug- and radiation-induced kidney injury, reduced kidney fibrosis, improved nephrotic syndrome, reduced postmenopausal kidney damage, reduced old-age-induced kidney damage, and reduced kidney barrier dysfunction by improving the glucose balance, reducing inflammation, increasing antioxidant activity and kidney function, reducing vascular resistance, and improving kidney blood pressure. Meanwhile, in cancer cells, genistein can prevent the abnormal proliferation of cancer cells, induce cancer cell apoptosis, and inhibit tumor growth. Genistein has a wide range of health benefits, which make it the first choice for the treatment of kidney disease. However, the effect of genistein on kidney cancer requires more studies in humans and animals.
In addition, there are a large number of cellular and animal experiments in this review, which may be used to predict whether genistein can prevent human kidney disease.
(1) However, at present, there is a lack of research exploring the relationship between the content and concentration of genistein in blood and kidney diseases. (2) Although many foods contain genistein (as mentioned earlier), the exact amount of genistein consumed in the same area and the concentration of genistein in the blood of different people may be different. (3) We can carry out more experiments using human pathological cell models to provide a more valuable reference for the application of genistein. Additionally, the following aspects need to be further studied and clarified: (i) the dose and bioavailability of genistein in different kidney diseases, (ii) genistein metabolism and biological effects, and (iii) the signaling mechanisms involved. (4) At present, only a limited number of studies have examined the role of genistein in human kidneys. Because there are no current ongoing clinical trials in humans, and the side effects and dosage of genistein in human kidneys, the most effective stages of kidney diseases, and the magnitude of the benefits are unknown. Therefore, more human trials are needed to address the above problems in the future.

Author Contributions

Conceptualization, Q.P. and Z.X.; writing—original draft preparation, Q.P. and Y.L. (Yuanyuan Li); writing—review and editing, Q.P., Y.L. (Yuanyuan Li), J.S., H.H., Y.L. (Yipei Liang), Y.Z., Y.D., C.C. and Z.X. Graphic abstracts: Z.X. All authors have read and agreed to the published version of the manuscript.

Funding

This research was sponsored by Graduate Education Reform and Quality Improvement Project of Henan Province (grant number: YJS2021AL074), Graduate Education Innovation and Quality Improvement Project of Henan University (grant number: SYL19060141), Planning and Decision Consultation Project of Henan Province (grant number: 2018JC38), Henan Province Higher Education Teaching Reform Research and Practice Project (HDXJJG2020-25), and Henan Province Science and Technology Development Plan Soft Science Research (N0.222400410497).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

Qianwen Peng and Yuanyuan Li (co-first authors) contributed equally to this work, and Zhenxing Xie is the co-corresponding author.

Conflicts of Interest

The authors declare that they have no conflict of interest.

Abbreviations

AP-1Activating protein-1
Ang IIAngiotensin II
ATPAdenosine-triphosphate
ARAldose reductase
ACEAngiotensin-converting enzyme
AKIAcute kidney injury
ACEAngiotensin-converting enzyme
α-SMAα-Smooth muscle actin
BPBlood pressure
BUNBlood urea nitrogen
Ca2+Calcium
cGMPCyclic guanosine monophosphate
COX-2Cyclooxygenase-2
CDKN2aCyclin-dependent kinase inhibitor 2a
CKDChronic kidney disease
CTGFConnective tissue growth factor
DIRD1 receptor
D2RD1 receptor
DICDisseminated intravascular coagulation
EREstrogen receptor
EGFEpidermal growth factor
ERK2Extracellular signal-regulated kinase 2
EvsExtracellular vesicles
ECMExtracellular matrix
EPOErythropoietin
EEDEmbryonic ectoderm development
ESRDEnd-stage renal disease
eNOSEndothelial NO synthase
Fosc-Fos gene
FAKFocal adhesion kinase
FGFFibroblast growth factor
FBGFasting blood glucose
GFATGlutamine:fructose-6-phosphate amidotransferase
GGTGamma-glutamyl transferase
HIF-1Hypoxia-inducible factor 1
HGFHepatocyte growth factor
HSP70Heat shock protein 70
HGHigh glucose
HOTAIRHOX transcript antisense RNA
HO-1Heme oxygenase-1
IL-1βInterleukin-1 beta
IP3Inositol triphosphate
IGF-IInsulin-like growth factor-I
IRF3Interferon regulatory factor 3
IL-6Interleukin-6
I/RIschemia/reperfusion
ICAMIntercellular adhesion molecule-1
L-dopal-Dihydroxyphenylalanine
LysoPCLysophosphatidylcholine
LPSLipopolysaccharide
LDHLactate dehydrogenase
MCGlomerular mesangial cells
MCP-1Monocyte chemoattractant protein-1
MAPKMitogen-activated protein kinases
MAPMitogen-activated protein
mTORMammalian target of rapamycin
MyD88Myeloid differentiation primary response 88
MVDMicrovessel density
MGOMethylglyoxal
MDCKMadin-Darby canine kidney
MDAMalondialdehyde
NF-κBNuclear factor NF-kappaB
NONitric oxide
NOSNitric oxide synthesis
NKANa (+), K (+)-ATPase
NHENa+/H+ exchanger
NGFNerve growth factor
NADPHNicotinamide adenine dinucleotide phosphate
NPp-Nonylphenol
Nrf2Nuclear factor erythroid 2-related factor 2
NQO1NAD(P)H:Quinone Oxidoreductase 1
PDGFPlatelet-derived growth factor
PGE2Prostaglandin E2
PLA2Phospholipase A2 transcription
PLC-γ1Phospholipase C-γ1
PAI-1Plasminogen activator inhibitor-1
PTKPhototherapeutic keratectomy
PKCProtein kinase C
PAHPhenylalanine hydroxylase
Pax-2Paired homeobox-2 gene
PiPhosphate
PTH1RParathyroid hormone 1 receptor
PaAlbumin permeability
PRC2Polycomb repressive complex 2
PKC-βIIProtein kinase C-βII
PTHParathyroid hormone
RAGEReceptor for advanced glycation end products
RasRenin–angiotensin system
RCCRenal cell carcinoma
ROSReactive oxygen species
RTRadiation therapy
SAPKc-Jun phosphorylation
SMITSodium/myo-inositol cotransporter
SNAPs-Nitroso-N-acetyl-penicillamine
SMARCB1Subfamily B member 1
SHR-SPsStroke-prone spontaneously hypertensive rats
TGF-βTransforming growth factor-β
TRPV5Transient receptor potential vanilloid 5
TLR4-MD2Toll-like receptor 4- myeloid differentiation 2
TRIFTIR domain-containing adapter-inducing interferon-beta
TNF-αTumor necrosis factor α
TLR-4Toll-like-receptor-4
UTPUridine triphosphate
UUOUnilateral ureteral obstruction
VPFVascular permeability factor
VEGFVascular endothelial growth factor
5-HT2A5-Hydroxytryptamine 2A
15d-PGJ215-Deoxy-delta12,14-prostaglandin J2

References

  1. Walter, E.D. Genistin (an Isoflavone Glucoside) and its Aglucone, Genistein, from Soybeans. J. Am. Chem.Soc. 1941, 63, 3273–3276. [Google Scholar] [CrossRef]
  2. Coward, L.; Barnes, N.C.; Setchell, K.D.R.; Barnes, S. Genistein, daidzein, and their.beta.-glycoside conjugates: Antitumor isoflavones in soybean foods from American and Asian diets. J. Agr. Food Chem. 1993, 41, 1961–1967. [Google Scholar] [CrossRef]
  3. Kaufman, P.B.; Duke, J.A.; Brielmann, H.; Boik, J.; Hoyt, J.E. A comparative survey of leguminous plants as sources of the isoflavones, genistein and daidzein: Implications for human nutrition and health. J. Altern. Complement. Med. 1997, 3, 7–12. [Google Scholar] [CrossRef] [PubMed]
  4. Rao, H. Isoflavones from Flemingia vestita. Fitoterapia 1991, 62, 458. [Google Scholar]
  5. Rao, K.N.; Srimannarayana, G. Fleminone, a flavanone from the stems of Flemingia macrophylla. Phytochemistry. 1983, 22, 2287–2290. [Google Scholar] [CrossRef]
  6. Wang, B.-S.; Juang, L.-J.; Yang, J.-J.; Chen, L.-Y.; Tai, H.-M.; Huang, M.-H. Antioxidant and Antityrosinase Activity of Flemingia macrophylla and Glycine tomentella Roots. Evid. Based Complement. Altern. Med. 2012, 20, 431081. [Google Scholar]
  7. Fedoreyev, S.A.; Pokushalova, T.V.; Veselova, M.V.; Glebko, L.I.; Kulesh, N.I.; Muzarok, T.I.; Seletskaya, L.D.; Bulgakov, V.P.; Zhuravlev, Y.N. Isoflavonoid production by callus cultures of Maackia amurensis. Fitoterapia 2000, 71, 365–372. [Google Scholar] [CrossRef]
  8. Popiolkiewicz, J.; Polkowski, K.; Skierski, J.S.; Mazurek, A.P. In vitro toxicity evaluation in the development of new anticancer drugs-genistein glycosides. Cancer Lett. 2005, 22, 67–75. [Google Scholar] [CrossRef]
  9. Walsh, K.R.; Haak, S.J.; Bohn, T.; Tian, Q.; Schwartz, S.J.; Failla, M.L. Isoflavonoid glucosides are deconjugated and absorbed in the small intestine of human subjects with ileostomies. Am. J. Clin. Nutr. 2007, 85, 1050–1056. [Google Scholar] [CrossRef]
  10. Bokkenheuser, V.D.; Shackleton, C.H.; Winter, J. Hydrolysis of dietary flavonoid glycosides by strains of intestinal Bacteroides from humans. Biochem. J. 1987, 248, 953–956. [Google Scholar] [CrossRef]
  11. Hu, M. Commentary: Bioavailability of flavonoids and polyphenols: Call to arms. Mol. Pharm. 2007, 4, 803–806. [Google Scholar] [CrossRef]
  12. Yang, Z.; Zhu, W.; Gao, S.; Xu, H.; Wu, B.; Kulkarni, K.; Singh, R.; Tang, L.; Hu, M. Simultaneous determination of genistein and its four phase II metabolites in blood by a sensitive and robust UPLC-MS/MS method: Application to an oral bioavailability study of genistein in mice. J. Pharm. Biomed. Anal. 2010, 53, 81–89. [Google Scholar] [CrossRef] [Green Version]
  13. Kurkela, M.; García-Horsman, J.A.; Luukkanen, L.; Mörsky, S.; Taskinen, J.; Baumann, M.; Kostiainen, R.; Hirvonen, J.; Finel, M. Expression and characterization of recombinant human UDP-glucuronosyltransferases (UGTs). UGT1A9 is more resistant to detergent inhibition than other UGTs and was purified as an active dimeric enzyme. J. Biol. Chem. 2003, 278, 3536–3544. [Google Scholar] [CrossRef]
  14. Rozman, K.K.; Bhatia, J.; Calafat, A.M.; Chambers, C.; Culty, M.; Etzel, R.A.; Flaws, J.A.; Hansen, D.K.; Hoyer, P.B.; Jeffery, E.H.; et al. NTP-CERHR expert panel report on the reproductive and developmental toxicity of genistein. Birth Defects Res. B Dev. Reprod Toxicol. 2006, 77, 485–638. [Google Scholar] [CrossRef]
  15. Andersen, O.M.; Markham, K.R. Flavonoids: Chemistry, Biochemistry and Applications; CRC Press: Boca Raton, FL, USA, 2006. [Google Scholar]
  16. Morito, K.; Hirose, T.; Kinjo, J.; Hirakawa, T.; Okawa, M.; Nohara, T.; Ogawa, S.; Inoue, S.; Muramatsu, M.; Masamune, Y. Interaction of Phytoestrogens with Estrogen Receptors & alpha; and & beta. Biol. Pharam. Bull. 2001, 24, 351–356. [Google Scholar]
  17. Sakamoto, T.; Horiguchi, H.; Oguma, E.; Kayama, F. Effects of diverse dietary phytoestrogens on cell growth, cell cycle and apoptosis in estrogen-receptor-positive breast cancer cells. J. Nutr. Biochem. 2010, 21, 856–864. [Google Scholar] [CrossRef]
  18. De Lemos, M.L. Effects of Soy Phytoestrogens Genistein and Daidzein on Breast Cancer Growth. Ann. Pharmacother. 2001, 35, 1118–1121. [Google Scholar] [CrossRef]
  19. Das, A.; Banik, N.L.; Ray, S.K. Flavonoids activated caspases for apoptosis in human glioblastoma T98G and U87MG cells but not in human normal astrocytes. Cancer. 2010, 116, 164–176. [Google Scholar] [CrossRef]
  20. Hwang, Y.W.; Kim, S.Y.; Jee, S.H.; Kim, Y.N.; Nam, C.M. Soy Food Consumption and Risk of Prostate Cancer: A Meta-Analysis of Observational Studies. Nutr. Cancer. 2009, 61, 598–606. [Google Scholar] [CrossRef]
  21. Kim, S.-H.; Kim, S.-H.; Kim, Y.-B.; Jeon, Y.-T.; Lee, S.-C.; Song, Y.-S. Genistein Inhibits Cell Growth by Modulating Various Mitogen-Activated Protein Kinases and AKT in Cervical Cancer Cells. Ann. N.Y. Acad. Sci. 2009, 117, 495–500. [Google Scholar] [CrossRef]
  22. Atteritano, M.; Marini, H.; Minutoli, L.; Polito, F.; Bitto, A.; Altavilla, D.; Mazzaferro, S.; D’anna, R.; Cannata, M.L.; Gaudio, A.; et al. Effects of the phytoestrogen genistein on some predictors of cardiovascular risk in osteopenic, postmenopausal women: A two-year randomized, double-blind, placebo-controlled study. J. Clin. Endocrinol. Metab. 2007, 92, 3068–3075. [Google Scholar] [CrossRef]
  23. Szkudelska, K.; Nogowski, L. Genistein—A dietary compound inducing hormonal and metabolic changes. J. Steroid. Biochem. Mol. Biol. 2007, 105, 37–45. [Google Scholar] [CrossRef]
  24. Choi, M.S.; Jung, U.J.; Yeo, J.; Kim, M.J.; Lee, M.K. Genistein and daidzein prevent diabetes onset by elevating insulin level and altering hepatic gluconeogenic and lipogenic enzyme activities in non-obese diabetic (NOD) mice. Diabetes Metab. Res. Rev. 2008, 24, 74–81. [Google Scholar] [CrossRef]
  25. Rodríguez-Landa, J.F.; Cueto-Escobedo, J.; Puga-Olguín, A.; Rivadeneyra-Domínguez, E.; Bernal-Morales, B.; Herrera-Huerta, E.V.; Santos-Torres, A. The Phytoestrogen Genistein Produces Similar Effects as 17β-Estradiol on Anxiety-Like Behavior in Rats at 12 Weeks after Ovariectomy. Biomed. Res. Int. 2017, 2017, 9073816. [Google Scholar] [CrossRef]
  26. Velasquez, M.T.; Bhathena, S.J. Dietary phytoestrogens: A possible role in renal disease protection. Am. J. Kidney Dis. 2001, 37, 1056–1068. [Google Scholar] [CrossRef]
  27. Mazur, W.M.; Duke, J.A.; Wähälä, K.; Rasku, S.; Adlercreutz, H. Isoflavonoids and Lignans in Legumes: Nutritional and Health Aspects in Humans. J. Nutr. Biochem. 1998, 9, 193–200. [Google Scholar] [CrossRef]
  28. Adlercreutz, H.; Mazur, W. Phyto-oestrogens and Western diseases. Ann. Med. 1997, 29, 95–120. [Google Scholar] [CrossRef]
  29. Mazur, W.; Adlercreutz, H. Naturally occurring oestrogens in food. Pure Appl. Chem. 1998, 70, 1759–1776. [Google Scholar] [CrossRef]
  30. Mazur, W.; Fotsis, T.; Wähälä, K.; Ojala, S.; Salakka, A.; Adlercreutz, H. Isotope dilution gas chromatographic-mass spectrometric method for the determination of isoflavonoids, coumestrol, and lignans in food samples. Anal. Biochem. 1996, 233, 169–180. [Google Scholar] [CrossRef]
  31. Schlondorff, D. The glomerular mesangial cell: An expanding role for a specialized pericyte. Faseb. J. 1987, 1, 272–281. [Google Scholar] [CrossRef]
  32. Kitching, A.R.; Hutton, H.L. The Players: Cells Involved in Glomerular Disease. Clin. J. Am. Soc. Nephrol. 2016, 11, 1664–1674. [Google Scholar] [CrossRef] [PubMed]
  33. Yokoo, T.; Kitamura, M. Dual regulation of IL-1 beta-mediated matrix metalloproteinase-9 expression in mesangial cells by NF-kappa B and AP-1. Am. J. Physiol. 1996, 270 Pt 2, F123–F130. [Google Scholar] [CrossRef] [PubMed]
  34. Mühl, H.; Geiger, T.; Pignat, W.; Märki, F.; Van Den Bosch, H.; Vosbeck, K.; Pfeilschifter, J. PDGF suppresses the activation of group II phospholipase A2 gene expression by interleukin 1 and forskolin in mesangial cells. FEBS Lett. 1991, 291, 249–252. [Google Scholar] [CrossRef] [Green Version]
  35. Rovin, B.H.; Tan, L.C. Role of protein kinase pathways in IL-1-induced chemoattractant expression by human mesangial cells. Kidney Int. 1994, 46, 1059–1068. [Google Scholar] [CrossRef]
  36. Wu, X.; Dolecki, G.J.; Lefkowith, J.B. GRO chemokines: A transduction, integration, and amplification mechanism in acute renal inflammation. Am. J. Physiol. 1995, 269 Pt 2, F248–F256. [Google Scholar] [CrossRef]
  37. James, L.R.; Ingram, A.; Ly, H.; Thai, K.; Cai, L.; Scholey, J.W. Angiotensin II activates the GFAT promoter in mesangial cells. Am. J. Physiol. Renal. Physiol. 2001, 281, F151–F162. [Google Scholar] [CrossRef]
  38. Jung, D.H.; Kim, Y.S.; Kim, J.S. Screening system of blocking agents of the receptor for advanced glycation endproducts in cells using fluorescence. Biol. Pharm. Bull. 2012, 35, 1826–1830. [Google Scholar] [CrossRef]
  39. Grandaliano, G.; Choudhury, G.G.; Biswas, P.; Abboud, H.E. Mitogenic signaling of thrombin in mesangial cells: Role of tyrosine phosphorylation. Am. J. Physiol. 1994, 267 Pt 2, F528–F536. [Google Scholar] [CrossRef]
  40. Wenzel, U.O.; Fouqueray, B.; Biswas, P.; Grandaliano, G.; Choudhury, G.G.; Abboud, H.E. Activation of mesangial cells by the phosphatase inhibitor vanadate. Potential implications for diabetic nephropathy. J. Clin. Investig. 1995, 95, 1244–1252. [Google Scholar] [CrossRef]
  41. Goppelt-Struebe, M.; Stroebel, M. Mechanisms of serotonin-induced Ca2+ responses in mesangial cells. Naunyn Schmiedebergs Arch. Pharmacol. 1997, 356, 240–247. [Google Scholar] [CrossRef]
  42. Marrero, M.B.; Venema, R.C.; Ma, H.; Ling, B.N.; Eaton, D.C. Erythropoietin receptor-operated Ca2+ channels: Activation by phospholipase C-gamma 1. Kidney Int. 1998, 53, 1259–1268. [Google Scholar] [CrossRef]
  43. Ma, H.; Matsunaga, H.; Li, B.; Marrero, M.B.; Ling, B.N. Regulation of PDGF-beta receptor-operated Ca2+ channels by phospholipase C-gamma 1 in glomerular mesangial cells. Am. J. Physiol. 1996, 271 Pt 2, F994–F1003. [Google Scholar] [CrossRef]
  44. Ma, H.; Matsunaga, H.; Li, B.; Schieffer, B.; Marrero, M.B.; Ling, B.N. Ca2+ channel activation by platelet-derived growth factor-induced tyrosine phosphorylation and Ras guanine triphosphate-binding proteins in rat glomerular mesangial cells. J. Clin. Investig. 1996, 97, 2332–2341. [Google Scholar] [CrossRef] [Green Version]
  45. Narisawa-Saito, M.; Kimura, S.; Fujiwara, N.; Oite, T.; Shimoji, K.; Shimizu, F. Thy-1-mediated phosphatidylinositol turnover in cultured rat glomerular mesangial cell. J. Cell. Physiol. 1996, 168, 705–710. [Google Scholar] [CrossRef]
  46. Motojima, M.; Kakuchi, J.; Yoshioka, T. Association of TGF-beta signaling in angiotensin II-induced PAI-1 mRNA upregulation in mesangial cells: Role of PKC. Biochim. Biophys. Acta 1999, 1449, 217–226. [Google Scholar] [CrossRef]
  47. Marrero, M.B.; Schieffer, B.; Ma, H.; Bernstein, K.E.; Ling, B.N. ANG II-induced tyrosine phosphorylation stimulates phospholipase C-gamma 1 and Cl-channels in mesangial cells. Am. J. Physiol. 1996, 270 Pt 1, C1834–C1842. [Google Scholar] [CrossRef]
  48. Becker, B.N.; Kondo, S.; Chen, J.K.; Harris, R.C. Tyrosine kinase inhibition affects type 1 angiotensin II receptor internalization. J. Recept. Signal Transduct. Res. 1999, 19, 975–993. [Google Scholar] [CrossRef]
  49. Yuan, H.T.; Yang, S.P.; Woolf, A.S. Hypoxia up-regulates angiopoietin-2, a Tie-2 ligand, in mouse mesangial cells. Kidney Int. 2000, 58, 1912–1919. [Google Scholar] [CrossRef]
  50. Wu, Z.L.; Wang, Y.C.; Zhou, Q.; Ge, Y.Q.; Lan, Y. Oxidized LDL induces transcription factor activator protein-1 in rat mesangial cells. Cell. Biochem. Funct. 2003, 21, 249–256. [Google Scholar] [CrossRef]
  51. El-Dahr, S.S.; Dipp, S.; Baricos, W.H. Bradykinin stimulates the ERK-->Elk-1-->Fos/AP-1 pathway in mesangial cells. Am. J. Physiol. 1998, 275, F343–F352. [Google Scholar] [CrossRef]
  52. Gruden, G.; Thomas, S.; Burt, D.; Lane, S.; Chusney, G.; Sacks, S.; Viberti, G. Mechanical stretch induces vascular permeability factor in human mesangial cells: Mechanisms of signal transduction. Proc. Natl. Acad. Sci. USA 1997, 94, 12112–12116. [Google Scholar] [CrossRef]
  53. Alric, C.; Pecher, C.; Tack, I.; Schanstra, J.P.; Bascands, J.L.; Girolami, J.P. Inhibition of cGMP accumulation in mesangial cells by bradykinin and tyrosine kinase inhibitors. Int. J. Mol. Med. 1999, 4, 557–564. [Google Scholar] [CrossRef]
  54. Bassa, B.V.; Roh, D.D.; Vaziri, N.D.; Kirschenbaum, M.A.; Kamanna, V.S. Lysophosphatidylcholine activates mesangial cell PKC and MAP kinase by PLCgamma-1 and tyrosine kinase-Ras pathways. Am. J. Physiol. 1999, 277, F328–F337. [Google Scholar]
  55. Simonson, M.S.; Herman, W.H. Protein kinase C and protein tyrosine kinase activity contribute to mitogenic signaling by endothelin-1. Cross-talk between G protein-coupled receptors and pp60c-src. J. Biol. Chem. 1993, 268, 9347–9357. [Google Scholar] [CrossRef]
  56. Hirakata, M.; Kaname, S.; Chung, U.G.; Joki, N.; Hori, Y.; Noda, M.; Takuwa, Y.; Okazaki, T.; Fujita, T.; Katoh, T.; et al. Tyrosine kinase dependent expression of TGF-beta induced by stretch in mesangial cells. Kidney Int. 1997, 51, 1028–1036. [Google Scholar] [CrossRef]
  57. Yuan, W.J.; Jia, F.Y.; Meng, J.Z. Effects of genistein on secretion of extracellular matrix components and transforming growth factor beta in high-glucose-cultured rat mesangial cells. J. Artif. Organs. 2009, 12, 242–246. [Google Scholar] [CrossRef]
  58. Tetsuka, T.; Morrison, A.R. Tyrosine kinase activation is necessary for inducible nitric oxide synthase expression by interleukin-1 beta. Am. J. Physiol. 1995, 269 Pt 1, C55–C59. [Google Scholar] [CrossRef]
  59. Pfeilschifter, J.; Huwiler, A. Nitric oxide stimulates stress-activated protein kinases in glomerular endothelial and mesangial cells. FEBS Lett. 1996, 396, 67–70. [Google Scholar] [CrossRef]
  60. Huwiler, A.; Van Rossum, G.; Wartmann, M.; Pfeilschifter, J. Angiotensin II stimulation of the stress-activated protein kinases in renal mesangial cells is mediated by the angiotensin AT1 receptor subtype. Eur. J. Pharmacol. 1998, 343, 297–302. [Google Scholar] [CrossRef]
  61. Huwiler, A.; Van Rossum, G.; Wartmann, M.; Pfeilschifter, J. Stimulation by extracellular ATP and UTP of the stress-activated protein kinase cascade in rat renal mesangial cells. Br. J. Pharmacol. 1997, 120, 807–812. [Google Scholar] [CrossRef] [PubMed]
  62. Kagami, S.; Urushihara, M.; Kondo, S.; Löster, K.; Reutter, W.; Tamaki, T.; Yoshizumi, M.; Kuroda, Y. Requirement for tyrosine kinase-ERK1/2 signaling in alpha 1 beta 1 integrin-mediated collagen matrix remodeling by rat mesangial cells. Exp. Cell Res. 2001, 268, 274–283. [Google Scholar] [CrossRef] [PubMed]
  63. Daphna-Iken, D.; Morrison, A.R. Interleukin-1 beta induces interstitial collagenase gene expression and protein secretion in renal mesangial cells. Am. J. Physiol. 1995, 269 Pt 2, F831–F837. [Google Scholar] [CrossRef] [PubMed]
  64. Neugarten, J.; Acharya, A.; Lei, J.; Silbiger, S. Selective estrogen receptor modulators suppress mesangial cell collagen synthesis. Am. J. Physiol. Renal Physiol. 2000, 279, F309–F318. [Google Scholar] [CrossRef] [PubMed]
  65. Neugarten, J.; Medve, I.; Lei, J.; Silbiger, S.R. Estradiol suppresses mesangial cell type I collagen synthesis via activation of the MAP kinase cascade. Am. J. Physiol. 1999, 277, F875–F881. [Google Scholar] [CrossRef] [PubMed]
  66. Mizobata, R. Effects of lysophosphatidylcholine on expression of monocyte chemoattractant protein-1 in glomerular endothelial cells. Nihon. Jinzo. Gakkai. Shi. 2003, 45, 76–83. [Google Scholar] [PubMed]
  67. Carranza, A.; Musolino, P.L.; Villar, M.; Nowicki, S. Signaling cascade of insulin-induced stimulation of L-dopa uptake in renal proximal tubule cells. Am. J. Physiol. Cell Physiol. 2008, 295, C1602–C1609. [Google Scholar] [CrossRef] [PubMed]
  68. Soares-Da-Silva, P.; Serrão, M.P. Molecular modulation of inward and outward apical transporters of L-dopa in LLC-PK(1) cells. Am. J. Physiol. Renal Physiol. 2000, 279, F736–F746. [Google Scholar] [CrossRef]
  69. Banday, A.A.; Fazili, F.R.; Lokhandwala, M.F. Insulin causes renal dopamine D1 receptor desensitization via GRK2-mediated receptor phosphorylation involving phosphatidylinositol 3-kinase and protein kinase C. Am. J. Physiol. Renal Physiol. 2007, 293, F877–F884. [Google Scholar] [CrossRef]
  70. Narkar, V.; Hussain, T.; Lokhandwala, M. Role of tyrosine kinase and p44/42 MAPK in D(2)-like receptor-mediated stimulation of Na(+), K(+)-ATPase in kidney. Am. J. Physiol. Renal Physiol. 2002, 282, F697–F702. [Google Scholar] [CrossRef]
  71. Lee, Y.J.; Heo, J.S.; Suh, H.N.; Lee, M.Y.; Han, H.J. Interleukin-6 stimulates alpha-MG uptake in renal proximal tubule cells: Involvement of STAT3, PI3K/Akt, MAPKs, and NF-kappaB. Am. J. Physiol. Renal Physiol. 2007, 293, F1036–F1046. [Google Scholar] [CrossRef]
  72. Han, H.J.; Park, S.H.; Lee, Y.J. Signaling cascade of ANG II-induced inhibition of alpha-MG uptake in renal proximal tubule cells. Am. J. Physiol. Renal Physiol. 2004, 286, F634–F642. [Google Scholar] [CrossRef]
  73. Jae Han, H.; Yeong Park, J.; Jung Lee, Y.; Taub, M. Epidermal growth factor inhibits 14C-alpha-methyl-D-glucopyranoside uptake in renal proximal tubule cells: Involvement of PLC/PKC, p44/42 MAPK, and cPLA2. J. Cell Physiol. 2004, 199, 206–216. [Google Scholar] [CrossRef]
  74. Girardi, A.C.; Knauf, F.; Demuth, H.U.; Aronson, P.S. Role of dipeptidyl peptidase IV in regulating activity of Na+/H+ exchanger isoform NHE3 in proximal tubule cells. Am. J. Physiol. Cell Physiol. 2004, 287, C1238–C1245. [Google Scholar] [CrossRef]
  75. Carraro-Lacroix, L.R.; Ramirez, M.A.; Zorn, T.M.; Rebouças, N.A.; Malnic, G. Increased NHE1 expression is associated with serum deprivation-induced differentiation in immortalized rat proximal tubule cells. Am. J. Physiol. Renal Physiol. 2006, 291, F129–F139. [Google Scholar] [CrossRef]
  76. Miyata, Y.; Asano, Y.; Muto, S. Hyperosmotic urea activates basolateral NHE in proximal tubule from P-gp null and wild-type mice. Am. J. Physiol. Renal Physiol. 2002, 283, F771–F783. [Google Scholar] [CrossRef]
  77. Grandaliano, G.; Monno, R.; Ranieri, E.; Gesualdo, L.; Schena, F.P.; Martino, C.; Ursi, M. Regenerative and proinflammatory effects of thrombin on human proximal tubular cells. J. Am. Soc. Nephrol. 2000, 11, 1016–1025. [Google Scholar] [CrossRef]
  78. Sandau, K.B.; Zhou, J.; Kietzmann, T.; Brüne, B. Regulation of the hypoxia-inducible factor 1alpha by the inflammatory mediators nitric oxide and tumor necrosis factor-alpha in contrast to desferroxamine and phenylarsine oxide. J Biol. Chem. 2001, 276, 39805–39811. [Google Scholar] [CrossRef]
  79. Hagar, H.; Ueda, N.; Shah, S.V. Tyrosine phosphorylation in DNA damage and cell death in hypoxic injury to LLC-PK1 cells. Kidney Int. 1997, 51, 1747–1753. [Google Scholar] [CrossRef]
  80. Fintha, A.; Sebe, A.; Masszi, A.; Terebessy, T.; Huszár, T.; Rosivall, L.; Mucsi, I. Angiotensin II activates plasminogen activator inhibitor-I promoter in renal tubular epithelial cells via the AT1 receptor. Acta Physiol. Hung. 2007, 94, 19–30. [Google Scholar] [CrossRef]
  81. Kimura, H.; Li, X.; Torii, K.; Okada, T.; Takahashi, N.; Fujii, H.; Ishihara, S.; Yoshida, H. A natural PPAR-gamma agonist, 15-deoxy-delta 12,14-prostaglandin J2, may act as an enhancer of PAI-1 in human proximal renal tubular cells under hypoxic and inflammatory conditions. Nephrol. Dial. Transplant. 2008, 23, 2496–2503. [Google Scholar] [CrossRef]
  82. Yamamoto, N.; Smith, M.W.; Maki, A.; Berezesky, I.K.; Trump, B.F. Role of cytosolic Ca2+ and protein kinases in the induction of the hsp70 gene. Kidney Int. 1994, 45, 1093–1104. [Google Scholar] [CrossRef]
  83. Schwartz, J.H.; Shih, T.; Menza, S.A.; Lieberthal, W. ATP depletion increases tyrosine phosphorylation of beta-catenin and plakoglobin in renal tubular cells. J. Am. Soc. Nephrol. 1999, 10, 2297–2305. [Google Scholar] [CrossRef]
  84. Gabriëls, G.; Werners, A.; Mauss, S.; Greven, J. Evidence for differential regulation of renal proximal tubular p-aminohippurate and sodium-dependent dicarboxylate transport. J. Pharmacol. Exp. Ther. 1999, 290, 710–715. [Google Scholar]
  85. Nishida, M.; Kawakatsu, H.; Matsumura, T.; Ishiwari, K.; Tamai, M.; Sawada, T. Effect of epidermal growth factor on sodium-dependent L-alanine transport in LLC-PK1 cells. Kidney Blood Press. Res. 2000, 23, 10–13. [Google Scholar] [CrossRef]
  86. Carraro-Lacroix, L.R.; Girardi, A.C.; Malnic, G. Long-term regulation of vacuolar H(+)-ATPase by angiotensin II in proximal tubule cells. Pflugers. Arch. 2009, 458, 969–979. [Google Scholar] [CrossRef]
  87. Caverzasio, J.; Bonjour, J.P. Tyrosine phosphorylation selectively regulates renal cellular phosphate transport. Evidence that it mediates the stimulatory effect of insulin-like growth factor-1. Endocrinology 1992, 130, 373–380. [Google Scholar] [CrossRef]
  88. Han, H.J.; Park, J.Y.; Lee, Y.J.; Park, S.H. Effect of epidermal growth factor on phosphate uptake in renal proximal tubule cells: Involvement of PKC, MAPK, and cPLA2. Kidney Blood Press. Res. 2003, 26, 315–324. [Google Scholar] [CrossRef]
  89. Ikari, A.; Nakajima, K.; Taki, S.; Suketa, Y. Up-regulation of Na+-dependent Mg2+ transport by nitric oxide and cyclic GMP pathway in renal epithelial cells. Eur. J. Pharmacol. 2002, 451, 133–139. [Google Scholar] [CrossRef]
  90. Record, R.D.; Johnson, M.; Lee, S.; Blazer-Yost, B.L. Aldosterone and insulin stimulate amiloride-sensitive sodium transport in A6 cells by additive mechanisms. Am. J. Physiol. 1996, 27 Pt 1, C1079–C1084. [Google Scholar] [CrossRef]
  91. Niisato, N.; Van Driessche, W.; Liu, M.; Marunaka, Y. Involvement of protein tyrosine kinase in osmoregulation of Na(+) transport and membrane capacitance in renal A6 cells. J. Membr. Biol. 2000, 175, 63–77. [Google Scholar] [CrossRef]
  92. Niisato, N.; Marunaka, Y. Activation of the Na+-K+ pump by hyposmolality through tyrosine kinase-dependent Cl- conductance in Xenopus renal epithelial A6 cells. J. Physiol. 1999, 518 Pt 2, 417–432. [Google Scholar] [CrossRef] [PubMed]
  93. Good, D.W. Nerve growth factor regulates HCO3- absorption in thick ascending limb: Modifying effects of vasopressin. Am. J. Physiol. 1998, 274, C931–C939. [Google Scholar] [CrossRef] [PubMed]
  94. Watts, B.A., 3rd; Good, D.W. Hyposmolality stimulates apical membrane Na(+)/H(+) exchange and HCO(3)(-) absorption in renal thick ascending limb. J. Clin. Investig. 1999, 104, 1593–1602. [Google Scholar] [CrossRef] [PubMed]
  95. Zhang, Z.; Tian, W.; Cohen, D.M. Urea protects from the proapoptotic effect of NaCl in renal medullary cells. Am. J. Physiol. Renal Physiol. 2000, 279, F345–F352. [Google Scholar] [CrossRef] [PubMed]
  96. Yang, T.; Schnermann, J.B.; Briggs, J.P. Regulation of cyclooxygenase-2 expression in renal medulla by tonicity in vivo and in vitro. Am. J. Physiol. 1999, 277, F1–F9. [Google Scholar] [CrossRef] [PubMed]
  97. Atta, M.G.; Dahl, S.C.; Kwon, H.M.; Handler, J.S. Tyrosine kinase inhibitors and immunosuppressants perturb the myo-inositol but not the betaine cotransporter in isotonic and hypertonic MDCK cells. Kidney Int. 1999, 55, 956–962. [Google Scholar] [CrossRef] [PubMed]
  98. Tsukahara, H.; Gordienko, D.V.; Tonshoff, B.; Gelato, M.C.; Goligorsky, M.S. Direct demonstration of insulin-like growth factor-I-induced nitric oxide production by endothelial cells. Kidney Int. 1994, 45, 598–604. [Google Scholar] [CrossRef]
  99. Zhang, S.L.; Guo, J.; Moini, B.; Ingelfinger, J.R. Angiotensin II stimulates Pax-2 in rat kidney proximal tubular cells: Impact on proliferation and apoptosis. Kidney Int. 2004, 66, 2181–2192. [Google Scholar] [CrossRef]
  100. Niimura, Y.; Moue, T.; Takahashi, N.; Nagai, K. Medium osmolarity-dependent biosynthesis of renal cellular sulfoglycolipids is mediated by the MAPK signaling pathway. Biochim. Biophys. Acta 2010, 1801, 1155–1162. [Google Scholar] [CrossRef]
  101. Löwik, M.M.; Groenen, P.J.; Levtchenko, E.N.; Monnens, L.A.; Van Den Heuvel, L.P. Molecular genetic analysis of podocyte genes in focal segmental glomerulosclerosis—A review. Eur. J. Pediatr. 2009, 168, 1291–1304. [Google Scholar] [CrossRef]
  102. Kim, Y.H.; Goyal, M.; Kurnit, D.; Wharram, B.; Wiggins, J.; Holzman, L.; Kershaw, D.; Wiggins, R. Podocyte depletion and glomerulosclerosis have a direct relationship in the PAN-treated rat. Kidney Int. 2001, 60, 957–968. [Google Scholar] [CrossRef]
  103. Maezawa, Y.; Takemoto, M.; Yokote, K. Cell biology of diabetic nephropathy: Roles of endothelial cells, tubulointerstitial cells and podocytes. J. Diabetes Investig. 2015, 6, 3–15. [Google Scholar] [CrossRef]
  104. Bariéty, J.; Nochy, D.; Mandet, C.; Jacquot, C.; Glotz, D.; Meyrier, A. Podocytes undergo phenotypic changes and express macrophagic-associated markers in idiopathic collapsing glomerulopathy. Kidney Int. 1998, 53, 918–925. [Google Scholar] [CrossRef]
  105. Hong, J.; Bhat, O.M.; Li, G.; Dempsey, S.K.; Zhang, Q.; Ritter, J.K.; Li, W.; Li, P.L. Lysosomal regulation of extracellular vesicle excretion during d-ribose-induced NLRP3 inflammasome activation in podocytes. Biochim. Biophys. Acta Mol. Cell Res. 2019, 1866, 849–860. [Google Scholar] [CrossRef]
  106. Chen, C.A.; Hwang, J.C.; Guh, J.Y.; Tsai, J.C.; Chen, H.C. TGF-beta1 and integrin synergistically facilitate the differentiation of rat podocytes by increasing alpha-smooth muscle actin expression. Transl. Res. 2006, 148, 134–141. [Google Scholar] [CrossRef]
  107. Wang, Y.; Li, Y.; Zhang, T.; Chi, Y.; Liu, M.; Liu, Y. Genistein and Myd88 Activate Autophagy in High Glucose-Induced Renal Podocytes In Vitro. Med. Sci. Monit. 2018, 24, 4823–4831. [Google Scholar] [CrossRef]
  108. Chen, C.A.; Tsai, J.C.; Su, P.W.; Lai, Y.H.; Chen, H.C. Signaling and regulatory mechanisms of integrinalpha3beta1 on the apoptosis of cultured rat podocytes. J. Lab. Clin. Med. 2006, 147, 274–280. [Google Scholar] [CrossRef]
  109. Friedrich, C.; Endlich, N.; Kriz, W.; Endlich, K. Podocytes are sensitive to fluid shear stress in vitro. Am. J. Physiol. Renal Physiol. 2006, 291, F856–F865. [Google Scholar] [CrossRef]
  110. Liu, X.; Shi, Q.; Sigmund, C.D. Interleukin-1beta attenuates renin gene expression via a mitogen-activated protein kinase kinase-extracellular signal-regulated kinase and signal transducer and activator of transcription 3-dependent mechanism in As4.1 cells. Endocrinology 2006, 147, 6011–6018. [Google Scholar] [CrossRef]
  111. Antonipillai, I. Epidermal growth factor is a potent inhibitor of renin secretion. Hypertension 1993, 21, 654–659. [Google Scholar] [CrossRef]
  112. Endo, M. Calcium ion as a second messenger with special reference to excitation-contraction coupling. J. Pharmacol. Sci. 2006, 100, 519–524. [Google Scholar] [CrossRef] [PubMed]
  113. Živanović, J.; Jarić, I.; Ajdžanović, V.; Miler, M.; Stanković, S.; Milošević, V.; Filipović, B. Genistein regulates calcium and phosphate homeostasis without activation of MEK 1/2 signalling pathway in an animal model of the andropause. Ann. Anat. 2022, 239, 151836. [Google Scholar] [CrossRef] [PubMed]
  114. Couchourel, D.; Leclerc, M.; Filep, J.; Brunette, M.G. Testosterone enhances calcium reabsorption by the kidney. Mol. Cell Endocrinol. 2004, 222, 71–81. [Google Scholar] [CrossRef] [PubMed]
  115. Giménez, I.; Martinez, R.M.; Lou, M.; Mayoral, J.A.; Garay, R.P.; Alda, J.O. Salidiuretic action by genistein in the isolated, perfused rat kidney. Hypertension 1998, 31, 706–711. [Google Scholar] [CrossRef]
  116. Lou, J.M.; Giménez, I.; Martinez, R.M.; Alda, J.O.; Garay, R.P. Natriuretic effect of equol. J. Med. Food 1999, 2, 257–260. [Google Scholar] [CrossRef]
  117. Li, B.; Yao, J.; Morioka, T.; Oite, T. Nitric oxide increases albumin permeability of isolated rat glomeruli via a phosphorylation-dependent mechanism. J. Am. Soc. Nephrol. 2001, 12, 2616–2624. [Google Scholar] [CrossRef]
  118. Meyer, T.N.; Schwesinger, C.; Ye, J.; Denker, B.M.; Nigam, S.K. Reassembly of the tight junction after oxidative stress depends on tyrosine kinase activity. J. Biol. Chem. 2001, 276, 22048–22055. [Google Scholar] [CrossRef]
  119. Steimle, A.; Autenrieth, I.B.; Frick, J.S. Structure and function: Lipid A modifications in commensals and pathogens. Int. J. Med. Microbiol. 2016, 306, 290–301. [Google Scholar] [CrossRef]
  120. Glaros, T.G.; Chang, S.; Gilliam, E.A.; Maitra, U.; Deng, H.; Li, L. Causes and consequences of low grade endotoxemia and inflammatory diseases. Front. Biosci. 2013, 5, 754–765. [Google Scholar] [CrossRef]
  121. Gutierrez-Ramos, J.C.; Bluethmann, H. Molecules and mechanisms operating in septic shock: Lessons from knockout mice. Immunol. Today 1997, 18, 329–334. [Google Scholar] [CrossRef]
  122. Asakura, H. Classifying types of disseminated intravascular coagulation: Clinical and animal models. J. Intensive Care 2014, 2, 20. [Google Scholar] [CrossRef]
  123. Catorce, M.N.; Gevorkian, G. LPS-induced Murine Neuroinflammation Model: Main Features and Suitability for Pre-clinical Assessment of Nutraceuticals. Curr. Neuropharmacol. 2016, 14, 155–164. [Google Scholar] [CrossRef]
  124. Liang, Y.; Shen, T.; Ming, Q.; Han, G.; Zhang, Y.; Liang, J.; Zhu, D. Alpinetin ameliorates inflammatory response in LPS-induced endometritis in mice. Int. Immunopharmacol. 2018, 62, 309–312. [Google Scholar] [CrossRef]
  125. Zakaria, R.; Wan Yaacob, W.M.; Othman, Z.; Long, I.; Ahmad, A.H.; Al-Rahbi, B. Lipopolysaccharide-induced memory impairment in rats: A model of Alzheimer’s disease. Physiol. Res. 2017, 66, 553–565. [Google Scholar] [CrossRef]
  126. Stasi, A.; Intini, A.; Divella, C.; Franzin, R.; Montemurno, E.; Grandaliano, G.; Ronco, C.; Fiaccadori, E.; Pertosa, G.B.; Gesualdo, L.; et al. Emerging role of Lipopolysaccharide binding protein in sepsis-induced acute kidney injury. Nephrol. Dial. Transpl. 2017, 32, 24–31. [Google Scholar] [CrossRef] [Green Version]
  127. Park, B.S.; Song, D.H.; Kim, H.M.; Choi, B.S.; Lee, H.; Lee, J.O. The structural basis of lipopolysaccharide recognition by the TLR4-MD-2 complex. Nature 2009, 458, 1191–1195. [Google Scholar] [CrossRef]
  128. Sung, M.J.; Kim, D.H.; Davaatseren, M.; Hur, H.J.; Kim, W.; Jung, Y.J.; Park, S.K.; Kwon, D.Y. Genistein suppression of TNF-alpha-induced fractalkine expression in endothelial cells. Cell Physiol. Biochem. 2010, 26, 431–440. [Google Scholar] [CrossRef]
  129. Chen, X.; Tan, J.; Yang, M.; Liao, Z.K.; Lu, C.; Huang, Y.; Wu, L.C. Genistein has the function of alleviating and treating disseminated intravascular coagulation caused by lipopolysaccharide. J. Nat. Med. 2018, 72, 846–856. [Google Scholar] [CrossRef]
  130. Asmis, R.; Stevens, J.; Begley, J.G.; Grimes, B.; Van Zant, G.; Fanti, P. The isoflavone genistein inhibits LPS-stimulated TNFalpha, but not IL-6 expression in monocytes from hemodialysis patients and healthy subjects. Clin. Nephrol. 2006, 65, 267–275. [Google Scholar] [CrossRef]
  131. Gourgoutis, G.; Das, G. Gastrointestinal manifestations of cocaine addiction. Int. J. Clin. Pharmacol. Ther. 1994, 32, 136–141. [Google Scholar]
  132. Zhang, F.; Lu, Z.; Wang, F. Advances in the pathogenesis and prevention of contrast-induced nephropathy. Life Sci. 2020, 259, 118379. [Google Scholar] [CrossRef]
  133. Ghionzoli, N.; Sciaccaluga, C.; Mandoli, G.E.; Vergaro, G.; Gentile, F.; D’ascenzi, F.; Mondillo, S.; Emdin, M.; Valente, S.; Cameli, M. Cardiogenic shock and acute kidney injury: The rule rather than the exception. Heart Fail Rev. 2021, 26, 487–496. [Google Scholar] [CrossRef]
  134. Sukkummee, W.; Jittisak, P.; Wonganan, P.; Wittayalertpanya, S.; Chariyavilaskul, P.; Leelahavanichkul, A. The prominent impairment of liver/intestinal cytochrome P450 and intestinal drug transporters in sepsis-induced acute kidney injury over acute and chronic renal ischemia, a mouse model comparison. Ren. Fail. 2019, 41, 314–325. [Google Scholar] [CrossRef]
  135. Jeong, E.K.; Jang, H.J.; Kim, S.S.; Lee, S.Y.; Oh, M.Y.; Kim, H.J.; Eom, D.W.; Ham, J.Y.; Han, D.J. Protective Effect of Polydeoxyribonucleotide Against Renal Ischemia-Reperfusion Injury in Mice. Transpl. Proc. 2016, 48, 1251–1257. [Google Scholar] [CrossRef] [PubMed]
  136. Keller, A.K.; Jorgensen, T.M.; Ravlo, K.; Nielsen, T.K.; Olsen, L.H.; Stolle, L.B. Microdialysis for detection of renal ischemia after experimental renal transplantation. J. Urol. 2009, 182, 1854–1859. [Google Scholar] [CrossRef] [PubMed]
  137. Bonventre, J.V.; Yang, L. Cellular pathophysiology of ischemic acute kidney injury. J. Clin. Investig. 2011, 121, 4210–4221. [Google Scholar] [CrossRef]
  138. Schefold, J.C.; Filippatos, G.; Hasenfuss, G.; Anker, S.D.; Von Haehling, S. Heart failure and kidney dysfunction: Epidemiology, mechanisms and management. Nat. Rev. Nephrol. 2016, 12, 610–623. [Google Scholar] [CrossRef] [PubMed]
  139. Gonsalez, S.R.; Cortês, A.L.; Silva, R.C.D.; Lowe, J.; Prieto, M.C.; Silva Lara, L.D. Acute kidney injury overview: From basic findings to new prevention and therapy strategies. Pharmacol. Ther. 2019, 200, 1–12. [Google Scholar] [CrossRef]
  140. Praddaude, F.; Marchetti, J.; Alhenc-Gelas, F.; Ader, J. Dissimilar mechanisms of Ca(2+) response to bradykinin in different types of juxtamedullary glomerular arterioles. Am. J. Physiol. 1999, 277, F697–F705. [Google Scholar]
  141. Salomonsson, M.; Arendshorst, W.J. Effect of tyrosine kinase blockade on norepinephrine-induced cytosolic calcium response in rat afferent arterioles. Am. J. Physiol. Renal Physiol. 2004, 286, F866–F874. [Google Scholar] [CrossRef]
  142. Ji, E.S.; Zhang, L.H.; Wang, Y.H.; Yue, H.; He, R.R. Responses of regional vascular beds to local injection of genistein in rats. Sheng Li Xue Bao 2003, 55, 255–259. [Google Scholar]
  143. Fetscher, C.; Chen, H.; Schäfers, R.F.; Wambach, G.; Heusch, G.; Michel, M.C. Modulation of noradrenaline-induced microvascular constriction by protein kinase inhibitors. Naunyn Schmiedebergs Arch. Pharmacol. 2001, 363, 57–65. [Google Scholar] [CrossRef]
  144. Sun, C.W.; Falck, J.R.; Harder, D.R.; Roman, R.J. Role of tyrosine kinase and PKC in the vasoconstrictor response to 20-HETE in renal arterioles. Hypertension 1999, 33 Pt 2, 414–418. [Google Scholar] [CrossRef]
  145. Kurt, A.H.; Buyukafsar, K. Vasoconstriction induced by G1, a G-protein-coupled oestrogen receptor1 (GPER-1) agonist, in the isolated perfused rat kidney. Eur. J. Pharmacol. 2013, 702, 71–78. [Google Scholar] [CrossRef]
  146. Algin, M.C.; Hacioglu, A.; Yaylak, F.; Gulcan, E.; Aydin, T.; Hacioglu, B.A.; Ilhan, D.; Cevik, A.A.; Ates, E. The role of erythropoietin in hemorrhagic shock-induced liver and renal injury in rats. Adv. Ther. 2008, 25, 1353–1374. [Google Scholar] [CrossRef]
  147. Li, W.F.; Yang, K.; Zhu, P.; Zhao, H.Q.; Song, Y.H.; Liu, K.C.; Huang, W.F. Genistein Ameliorates Ischemia/Reperfusion-Induced Renal Injury in a SIRT1-Dependent Manner. Nutrients 2017, 9, 403. [Google Scholar] [CrossRef]
  148. Gholampour, F.; Mohammadi, Z.; Karimi, Z.; Owji, S.M. Protective effect of genistein in a rat model of ischemic acute kidney injury. Gene 2020, 753, 144789. [Google Scholar] [CrossRef]
  149. Ates, E.; Yalcin, A.U.; Yilmaz, S.; Koken, T.; Tokyol, C. Protective effect of erythropoietin on renal ischemia and reperfusion injury. ANZ J. Surg. 2005, 75, 1100–1105. [Google Scholar] [CrossRef]
  150. Linehan, W.M.; Schmidt, L.S.; Crooks, D.R.; Wei, D.; Srinivasan, R.; Lang, M.; Ricketts, C.J. The Metabolic Basis of Kidney Cancer. Cancer Discov. 2019, 9, 1006–1021. [Google Scholar] [CrossRef]
  151. Capitanio, U.; Montorsi, F. Renal cancer. Lancet 2016, 387, 894–906. [Google Scholar] [CrossRef]
  152. Balbaa, M.; Honke, K.; Makita, A. Regulation of glycolipid sulfotransferase by tyrosine kinases in human renal cancer cells. Biochim. Biophys. Acta 1996, 1299, 141–145. [Google Scholar] [CrossRef]
  153. Wang, Y.; Zhang, Y.T.; Liu, F.; Zhang, S.; Wang, W.; Li, Y.M. Effect of genistein on proliferation of renal cell carcinoma cell line GRC-1 and its influence to p27 expression. Ai Zheng 2003, 22, 1272–1275. [Google Scholar]
  154. Sasamura, H.; Takahashi, A.; Miyao, N.; Yanase, M.; Masumori, N.; Kitamura, H.; Itoh, N.; Tsukamoto, T. Inhibitory effect on expression of angiogenic factors by antiangiogenic agents in renal cell carcinoma. Br. J. Cancer 2002, 86, 768–773. [Google Scholar] [CrossRef]
  155. Majid, S.; Dar, A.A.; Ahmad, A.E.; Hirata, H.; Kawakami, K.; Shahryari, V.; Saini, S.; Tanaka, Y.; Dahiya, A.V.; Khatri, G.; et al. BTG3 tumor suppressor gene promoter demethylation, histone modification and cell cycle arrest by genistein in renal cancer. Carcinogenesis 2009, 30, 662–670. [Google Scholar] [CrossRef]
  156. Sasamura, H.; Takahashi, A.; Yuan, J.; Kitamura, H.; Masumori, N.; Miyao, N.; Itoh, N.; Tsukamoto, T. Antiproliferative and antiangiogenic activities of genistein in human renal cell carcinoma. Urology 2004, 64, 389–393. [Google Scholar] [CrossRef]
  157. Hirata, H.; Ueno, K.; Nakajima, K.; Tabatabai, Z.L.; Hinoda, Y.; Ishii, N.; Dahiya, R. Genistein downregulates onco-miR-1260b and inhibits Wnt-signalling in renal cancer cells. Br. J. Cancer 2013, 108, 2070–2078. [Google Scholar] [CrossRef]
  158. Imai-Sumida, M.; Dasgupta, P.; Kulkarni, P.; Shiina, M.; Hashimoto, Y.; Shahryari, V.; Majid, S.; Tanaka, Y.; Dahiya, R.; Yamamura, S. Genistein Represses HOTAIR/Chromatin Remodeling Pathways to Suppress Kidney Cancer. Cell Physiol. Biochem. 2020, 54, 53–70. [Google Scholar]
  159. Ji, Z.; Huo, C.; Yang, P. Genistein inhibited the proliferation of kidney cancer cells via CDKN2a hypomethylation: Role of abnormal apoptosis. Int. Urol. Nephrol. 2020, 52, 1049–1055. [Google Scholar] [CrossRef]
  160. Zaman, M.S.; Shahryari, V.; Deng, G.; Thamminana, S.; Saini, S.; Majid, S.; Chang, I.; Hirata, H.; Ueno, K.; Yamamura, S.; et al. Up-regulation of microRNA-21 correlates with lower kidney cancer survival. PLoS ONE 2012, 7, e31060. [Google Scholar] [CrossRef]
  161. Oh, H.Y.; Kwon, S.M.; Kim, S.I.; Jae, Y.W.; Hong, S.J. Antiangiogenic effect of ZD1839 against murine renal cell carcinoma (RENCA) in an orthotopic mouse model. Urol. Int. 2005, 75, 159–166. [Google Scholar] [CrossRef]
  162. Valencia, W.M.; Florez, H. How to prevent the microvascular complications of type 2 diabetes beyond glucose control. BMJ 2017, 356, i6505. [Google Scholar] [CrossRef] [PubMed]
  163. Saran, R.; Robinson, B.; Abbott, K.C.; Agodoa, L.Y.C.; Bragg-Gresham, J.; Balkrishnan, R.; Bhave, N.; Dietrich, X.; Ding, Z.; Eggers, P.W.; et al. US Renal Data System 2018 Annual Data Report: Epidemiology of Kidney Disease in the United States. Am. J. Kidney Dis. 2019, 73 (Suppl. S1), A7–A8. [Google Scholar] [CrossRef] [PubMed]
  164. Xu, Y.; Wang, L.; He, J.; Bi, Y.; Li, M.; Wang, T.; Wang, L.; Jiang, Y.; Dai, M.; Lu, J.; et al. Prevalence and control of diabetes in Chinese adults. JAMA 2013, 310, 948–959. [Google Scholar] [CrossRef] [PubMed]
  165. Sasser, J.M.; Sullivan, J.C.; Hobbs, J.L.; Yamamoto, T.; Pollock, D.M.; Carmines, P.K.; Pollock, J.S. Endothelin A receptor blockade reduces diabetic renal injury via an anti-inflammatory mechanism. J. Am. Soc. Nephrol. 2007, 18, 143–154. [Google Scholar] [CrossRef]
  166. Chen, J.; Chen, K.-H.; Xiao, F.; Dai, H.-Z.; Yang, J.; Wang, L.-M.; Wang, X.-Y.; Zhang, J.-G.; He, Y.-N. Decoy receptor 2 mediation of the senescent phenotype of tubular cells by interacting with peroxiredoxin 1 presents a novel mechanism of renal fibrosis in diabetic nephropathy. Kidney Int. 2020, 98, 645–662. [Google Scholar]
  167. Demir, Y.; Durmaz, L.; Taslimi, P.; Gulcin, I. Antidiabetic properties of dietary phenolic compounds: Inhibition effects on alpha-amylase, aldose reductase, and alpha-glycosidase. Biotechnol. Appl. Biochem. 2019, 66, 781–786. [Google Scholar] [CrossRef]
  168. Chu, C.; Lu, F.J.; Yeh, R.H.; Li, Z.L.; Chen, C.H. Synergistic antioxidant activity of resveratrol with genistein in high-glucose treated Madin-Darby canine kidney epithelial cells. Biomed. Rep. 2016, 4, 349–354. [Google Scholar] [CrossRef]
  169. Li, P.; Cao, Y.; Song, G.; Zhao, B.; Ma, Q.; Li, Z.; He, C. Anti-diabetic properties of genistein-chromium (III) complex in db/db diabetic mice and its sub-acute toxicity evaluation in normal mice. J. Trace Elem. Med. Biol. 2020, 62, 126606. [Google Scholar] [CrossRef]
  170. Palanisamy, N.; Viswanathan, P.; Anuradha, C.V. Effect of genistein, a soy isoflavone, on whole body insulin sensitivity and renal damage induced by a high-fructose diet. Ren. Fail. 2008, 30, 645–654. [Google Scholar] [CrossRef]
  171. Guo, T.L.; Germolec, D.R.; Zheng, J.F.; Kooistra, L.; Auttachoat, W.; Smith, M.J.; White, K.L.; Elmore, S.A. Genistein protects female nonobese diabetic mice from developing type 1 diabetes when fed a soy- and alfalfa-free diet. Toxicol. Pathol. 2015, 43, 435–448. [Google Scholar] [CrossRef]
  172. Zhao, Y.; Wang, P.; Sang, S. Dietary Genistein Inhibits Methylglyoxal-Induced Advanced Glycation End Product Formation in Mice Fed a High-Fat Diet. J. Nutr. 2019, 149, 776–787. [Google Scholar] [CrossRef]
  173. Rehman, K.; Ali, M.B.; Akash, M.S.H. Genistein enhances the secretion of glucagon-like peptide-1 (GLP-1) via downregulation of inflammatory responses. Biomed. Pharmacother. 2019, 112, 108670. [Google Scholar] [CrossRef]
  174. Elmarakby, A.A.; Ibrahim, A.S.; Faulkner, J.; Mozaffari, M.S.; Liou, G.I.; Abdelsayed, R. Tyrosine kinase inhibitor, genistein, reduces renal inflammation and injury in streptozotocin-induced diabetic mice. Vascul. Pharmacol. 2011, 55, 149–156. [Google Scholar] [CrossRef]
  175. Kim, M.J.; Lim, Y. Protective effect of short-term genistein supplementation on the early stage in diabetes-induced renal damage. Mediat. Inflamm. 2013, 2013, 510212. [Google Scholar] [CrossRef]
  176. Rauter, A.P.; Martins, A.; Borges, C.; Mota-Filipe, H.; Pinto, R.; Sepodes, B.; Justino, J. Antihyperglycaemic and protective effects of flavonoids on streptozotocin-induced diabetic rats. Phytother. Res. 2010, 24 (Suppl. 2), S133–S138. [Google Scholar] [CrossRef]
  177. Jheng, H.F.; Hayashi, K.; Matsumura, Y.; Kawada, T.; Seno, S.; Matsuda, H.; Inoue, K.; Nomura, W.; Takahashi, H.; Goto, T. Anti-Inflammatory and Antioxidative Properties of Isoflavones Provide Renal Protective Effects Distinct from Those of Dietary Soy Proteins against Diabetic Nephropathy. Mol. Nutr. Food Res. 2020, 64, e2000015. [Google Scholar] [CrossRef]
  178. Benter, I.F.; Yousif, M.H.; Hollins, A.J.; Griffiths, S.M.; Akhtar, S. Diabetes-induced renal vascular dysfunction is normalized by inhibition of epidermal growth factor receptor tyrosine kinase. J. Vasc. Res. 2005, 42, 284–291. [Google Scholar] [CrossRef]
  179. Stompór, T.; Perkowska-Ptasińska, A. Hypertensive kidney disease: A true epidemic or rare disease? Pol. Arch. Intern. Med. 2020, 130, 130–139. [Google Scholar] [CrossRef]
  180. Wenzel, R.R. Renal protection in hypertensive patients: Selection of antihypertensive therapy. Drugs. 2005, 65 (Suppl. 2), 29–39. [Google Scholar] [CrossRef]
  181. Palanisamy, N.; Venkataraman, A.C. Beneficial effect of genistein on lowering blood pressure and kidney toxicity in fructose-fed hypertensive rats. Br. J. Nutr. 2013, 109, 1806–1812. [Google Scholar] [CrossRef]
  182. Li, J.; Xie, Z.Z.; Tang, Y.B. Genistein prevents myocardial hypertrophy in 2-kidney 1-clip renal hypertensive rats by restoring eNOS pathway. Pharmacology 2010, 86, 240–248. [Google Scholar] [CrossRef]
  183. Cho, T.M.; Peng, N.; Clark, J.T.; Novak, L.; Roysommuti, S.; Prasain, J.; Wyss, J.M. Genistein attenuates the hypertensive effects of dietary NaCl in hypertensive male rats. Endocrinology 2007, 148, 5396–5402. [Google Scholar] [CrossRef]
  184. Mehta, R.L.; Pascual, M.T.; Soroko, S.; Savage, B.R.; Himmelfarb, J.; Ikizler, T.A.; Paganini, E.P.; Chertow, G.M. Spectrum of acute renal failure in the intensive care unit: The PICARD experience. Kidney Int. 2004, 66, 1613–1621. [Google Scholar] [CrossRef]
  185. Hoste, E.A.; Bagshaw, S.M.; Bellomo, R.; Cely, C.M.; Colman, R.; Cruz, D.N.; Edipidis, K.; Forni, L.G.; Gomersall, C.D.; Govil, D.; et al. Epidemiology of acute kidney injury in critically ill patients: The multinational AKI-EPI study. Intensive Care Med. 2015, 41, 1411–1423. [Google Scholar] [CrossRef]
  186. Moffett, B.S.; Goldstein, S.L. Acute kidney injury and increasing nephrotoxic-medication exposure in noncritically-ill children. Clin. J. Am. Soc. Nephrol. 2011, 6, 856–863. [Google Scholar] [CrossRef]
  187. Atkinson, R.M.; Currie, J.P.; Davis, B.; Pratt, D.A.; Sharpe, H.M.; Tomich, E.G. Acute toxicity of cephaloridine, an antibiotic derived from cephalosporin C. Toxicol. Appl. Pharmacol. 1966, 8, 398–406. [Google Scholar] [CrossRef]
  188. Foord, R.D. Cephaloridine, cephalothin and the kidney. J. Antimicrob. Chemother. 1975, 1, 119–133. [Google Scholar] [CrossRef]
  189. Kawai, Y.; Kohda, Y.; Kodawara, T.; Gemba, M. Protective effect of a protein kinase inhibitor on cellular injury induced by cephaloridine in the porcine kidney cell line LLC-PK(1). J. Toxicol. Sci. 2005, 30, 157–163. [Google Scholar] [CrossRef]
  190. Abd El-Lateef, S.M.; El-Sayed, E.M.; Mansour, A.M.; Salama, S.A. The protective role of estrogen and its receptors in gentamicin-induced acute kidney injury in rats. Life Sci. 2019, 239, 117082. [Google Scholar] [CrossRef]
  191. Lebwohl, D.; Canetta, R. Clinical development of platinum complexes in cancer therapy: An historical perspective and an update. Eur. J. Cancer 1998, 34, 1522–1534. [Google Scholar] [CrossRef]
  192. Levi, J.; Jacobs, C.; Kalman, S.M.; Mctigue, M.; Weiner, M.W. Mechanism of cis-platinum nephrotoxicity: I. Effects of sulfhydryl groups in rat kidneys. J. Pharmacol. Exp. Ther. 1980, 213, 545–550. [Google Scholar] [PubMed]
  193. Sung, M.J.; Kim, D.H.; Jung, Y.J.; Kang, K.P.; Lee, A.S.; Lee, S.; Kim, W.; Davaatseren, M.; Hwang, J.T.; Kim, H.J.; et al. Genistein protects the kidney from cisplatin-induced injury. Kidney Int. 2008, 74, 1538–1547. [Google Scholar] [CrossRef] [PubMed]
  194. Robbins, M.E.; Bonsib, S.M. Radiation nephropathy: A review. Scanning Microsc. 1995, 2, 535–560. [Google Scholar]
  195. Cohen, E.P.; Robbins, M.E. Radiation nephropathy. Semin. Nephrol. 2003, 23, 486–499. [Google Scholar] [CrossRef]
  196. Vijayalaxmi; Meltz, M.L.; Reiter, R.J.; Herman, T.S.; Kumar, K.S. Melatonin and protection from whole-body irradiation: Survival studies in mice. Mutat. Res. 1999, 425, 21–27. [Google Scholar] [CrossRef]
  197. Cooper, S.; Latendresse, J.R.; Doerge, D.R.; Twaddle, N.C.; Fu, X.; Delclos, K.B. Dietary modulation of p-nonylphenol-induced polycystic kidneys in male Sprague-Dawley rats. Toxicol. Sci. 2006, 91, 631–642. [Google Scholar] [CrossRef]
  198. Canyilmaz, E.; Uslu, G.H.; Bahat, Z.; Kandaz, M.; Mungan, S.; Haciislamoglu, E.; Mentese, A.; Yoney, A. Comparison of the effects of melatonin and genistein on radiation-induced nephrotoxicity: Results of an experimental study. Biomed. Rep. 2016, 4, 45–50. [Google Scholar] [CrossRef]
  199. Meguid El Nahas, A.; Bello, A.K. Chronic kidney disease: The global challenge. Lancet 2005, 365, 331–340. [Google Scholar] [CrossRef]
  200. Nogueira, A.; Pires, M.J.; Oliveira, P.A. Pathophysiological Mechanisms of Renal Fibrosis: A Review of Animal Models and Therapeutic Strategies. In Vivo 2017, 31, 1–22. [Google Scholar] [CrossRef]
  201. Wang, Y.; Xing, Q.Q.; Tu, J.K.; Tang, W.B.; Yuan, X.N.; Xie, Y.Y.; Wang, W.; Peng, Z.Z.; Huang, L.; Xu, H.; et al. Involvement of hydrogen sulfide in the progression of renal fibrosis. Chin. Med. J. 2019, 132, 2872–2880. [Google Scholar] [CrossRef]
  202. 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]
  203. Ning, Y.; Chen, J.; Shi, Y.; Song, N.; Yu, X.; Fang, Y.; Ding, X. Genistein Ameliorates Renal Fibrosis Through Regulation Snail via m6A RNA Demethylase ALKBH5. Front. Pharmacol. 2020, 11, 579265. [Google Scholar] [CrossRef]
  204. Jia, Q.; Yang, R.; Liu, X.F.; Ma, S.F.; Wang, L. Genistein attenuates renal fibrosis in streptozotocin-induced diabetic rats. Mol. Med. Rep. 2019, 19, 423–431. [Google Scholar] [CrossRef]
  205. Li, Y.; Chen, F.; Wei, A.; Bi, F.; Zhu, X.; Yin, S.; Lin, W.; Cao, W. Klotho recovery by genistein via promoter histone acetylation and DNA demethylation mitigates renal fibrosis in mice. J. Mol. Med. 2019, 97, 541–552. [Google Scholar] [CrossRef]
  206. Palanisamy, N.; Kannappan, S.; Anuradha, C.V. Genistein modulates NF-κB-associated renal inflammation, fibrosis and podocyte abnormalities in fructose-fed rats. Eur. J. Pharmacol. 2011, 667, 355–364. [Google Scholar] [CrossRef]
  207. Guo, Y.; Zhang, A.; Ding, Y.; Wang, Y.; Yuan, W. Genistein ameliorates parathyroid hormone-induced epithelial-to-mesenchymal transition and inhibits expression of connective tissue growth factor in human renal proximal tubular cells. Arch. Med. Sci. 2013, 9, 724–730. [Google Scholar] [CrossRef]
  208. Ghodake, S.R.; Suryakar, A.N.; Ankush, R.D.; Katkam, R.V.; Shaikh, K.; Katta, A.V. Role of free radicals and antioxidant status in childhood nephrotic syndrome. Indian J. Nephrol. 2011, 21, 37–40. [Google Scholar] [CrossRef]
  209. Javanbakht, M.H.; Sadria, R.; Djalali, M.; Derakhshanian, H.; Hosseinzadeh, P.; Zarei, M.; Azizi, G.; Sedaghat, R.; Mirshafiey, A. Soy protein and genistein improves renal antioxidant status in experimental nephrotic syndrome. Nefrologia 2014, 34, 483–490. [Google Scholar]
  210. Bryzgalova, G.; Gao, H.; Ahren, B.; Zierath, J.R.; Galuska, D.; Steiler, T.L.; Dahlman-Wright, K.; Nilsson, S.; Gustafsson, J.A.; Efendic, S.; et al. Evidence that oestrogen receptor-alpha plays an important role in the regulation of glucose homeostasis in mice: Insulin sensitivity in the liver. Diabetologia 2006, 49, 588–597. [Google Scholar] [CrossRef]
  211. Mohamed, M.K.; Abdel-Rahman, A.A. Effect of long-term ovariectomy and estrogen replacement on the expression of estrogen receptor gene in female rats. Eur. J. Endocrinol. 2000, 142, 307–314. [Google Scholar] [CrossRef]
  212. Choi, J.S.; Song, J. Effect of genistein on insulin resistance, renal lipid metabolism, and antioxidative activities in ovariectomized rats. Nutrition 2009, 25, 676–685. [Google Scholar] [CrossRef]
  213. Bowen, R.L.; Atwood, C.S. Living and Dying for Sex. Gerontology 2004, 50, 265–290. [Google Scholar] [CrossRef]
  214. Willems, H.M.E.; Van Den Heuvel, E.; Schoemaker, R.J.W.; Klein-Nulend, J.; Bakker, A.D. Diet and Exercise: A Match Made in Bone. Curr. Osteoporos. Rep. 2017, 15, 555–563. [Google Scholar] [CrossRef] [Green Version]
  215. Duan, X.; Li, Y.; Xu, F.; Ding, H. Study on the neuroprotective effects of Genistein on Alzheimer’s disease. Brain Behav. 2021, 11, e02100. [Google Scholar] [CrossRef]
  216. Kim, J.M.; Uehara, Y.; Choi, Y.J.; Ha, Y.M.; Ye, B.H.; Yu, B.P.; Chung, H.Y. Mechanism of attenuation of pro-inflammatory Ang II-induced NF-κB activation by genistein in the kidneys of male rats during aging. Biogerontology 2011, 12, 537–550. [Google Scholar] [CrossRef]
  217. Singh, R.; Barden, A.; Mori, T.; Beilin, L. Advanced glycation end-products: A review. Diabetologia 2001, 44, 129–146. [Google Scholar] [CrossRef] [PubMed]
  218. Ahmed, N. Advanced glycation endproducts--role in pathology of diabetic complications. Diabetes Res. Clin. Pract. 2005, 67, 3–21. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The effects of genistein on mesangial cells. NF-κB: nuclear factor NF-kappaB; PLA2: phospholipase A2 transcription; IL-1β: interleukin-1 beta; MCP-1: monocyte chemoattractant protein-1; GFAT: glutamine:fructose-6-phosphate amidotransferase; AGE: advanced glycation end products; RAGE: receptor for advanced glycation end products; Ca2+: calcium; 5-HT2A: 5-hydroxytryptamine 2A; PLC-γ1: phospholipase C-γ1; PDGF: platelet-derived growth factor; Ang II: angiotensin II; PAI-1: plasminogen activator inhibitor-1; IP3: inositol triphosphate; ERK2: extracellular signal-regulated kinase 2; AP-1: activating protein-1; Fos: c-fos gene; VPF: vascular permeability factor; cGMP: cyclic guanosine monophosphate; PTK: phototherapeutic keratectomy; PKC: protein kinase C; Ras: renin–angiotensin system; MAP: mitogen-activated protein; TGF-β1: transforming growth factor-β1; NO: nitric oxide; SAPK: c-Jun phosphorylation; ATP: adenosine-triphosphate; UTP: uridine triphosphate, ↓: inhibit; ↑: upregulate.
Figure 1. The effects of genistein on mesangial cells. NF-κB: nuclear factor NF-kappaB; PLA2: phospholipase A2 transcription; IL-1β: interleukin-1 beta; MCP-1: monocyte chemoattractant protein-1; GFAT: glutamine:fructose-6-phosphate amidotransferase; AGE: advanced glycation end products; RAGE: receptor for advanced glycation end products; Ca2+: calcium; 5-HT2A: 5-hydroxytryptamine 2A; PLC-γ1: phospholipase C-γ1; PDGF: platelet-derived growth factor; Ang II: angiotensin II; PAI-1: plasminogen activator inhibitor-1; IP3: inositol triphosphate; ERK2: extracellular signal-regulated kinase 2; AP-1: activating protein-1; Fos: c-fos gene; VPF: vascular permeability factor; cGMP: cyclic guanosine monophosphate; PTK: phototherapeutic keratectomy; PKC: protein kinase C; Ras: renin–angiotensin system; MAP: mitogen-activated protein; TGF-β1: transforming growth factor-β1; NO: nitric oxide; SAPK: c-Jun phosphorylation; ATP: adenosine-triphosphate; UTP: uridine triphosphate, ↓: inhibit; ↑: upregulate.
Nutrients 14 03768 g001
Figure 2. The effects of genistein on endothelial cells. MCP-1: monocyte chemoattractant protein-1; L-dopa: l-dihydroxyphenylalanine; DIR: D1 receptor; D2R: D1 receptor; EGF: epidermal growth factor; NHE: Na+/H+ exchanger; HIF-1: hypoxia-inducible factor 1; HGF: hepatocyte growth factor; PAH: phenylalanine hydroxylase; HSP70: heat shock protein 70; Pi: phosphate; Pax-2: paired homeobox-2 gene; COX-2: cyclooxygenase-2; SMIT: sodium/myo-inositol cotransporter, ↓: inhibit; ↑: upregulate.
Figure 2. The effects of genistein on endothelial cells. MCP-1: monocyte chemoattractant protein-1; L-dopa: l-dihydroxyphenylalanine; DIR: D1 receptor; D2R: D1 receptor; EGF: epidermal growth factor; NHE: Na+/H+ exchanger; HIF-1: hypoxia-inducible factor 1; HGF: hepatocyte growth factor; PAH: phenylalanine hydroxylase; HSP70: heat shock protein 70; Pi: phosphate; Pax-2: paired homeobox-2 gene; COX-2: cyclooxygenase-2; SMIT: sodium/myo-inositol cotransporter, ↓: inhibit; ↑: upregulate.
Nutrients 14 03768 g002
Figure 4. The mechanism of genistein actions in kidney. TLR-4: Toll-like-receptor-4, TNF-α: tumor necrosis factor α; IL-1β: interleukin-1 beta; IL-6: interleukin-6; NF-κB: nuclear factor NF-kappaB; MDA: malondialdehyde; Nrf2: nuclear factor erythroid 2-related factor 2; HO-1: heme oxygenase-1 (HO-1); NQO1: NAD(P) H:Quinone Oxidoreductase 1; ACE: angiotensin-converting enzyme; PKC: protein kinase C beta II; NO: nitric oxide; NOS: nitric oxide synthesis; AR: aldose reductase; FBG: fasting blood glucose; AGE: advanced glycation end products; CTGF: connective tissue growth factor; CDKN2a: cyclin-dependent kinase inhibitor 2a; EED: embryonic ectoderm development; HOTAIR: HOX transcript antisense RNA; PRC2: polycomb repressive complex 2; EGF: epidermal growth factor; VEGF: vascular endothelial growth factor; FGF: fibroblast growth factor, ↓: inhibit; ↑: upregulate.
Figure 4. The mechanism of genistein actions in kidney. TLR-4: Toll-like-receptor-4, TNF-α: tumor necrosis factor α; IL-1β: interleukin-1 beta; IL-6: interleukin-6; NF-κB: nuclear factor NF-kappaB; MDA: malondialdehyde; Nrf2: nuclear factor erythroid 2-related factor 2; HO-1: heme oxygenase-1 (HO-1); NQO1: NAD(P) H:Quinone Oxidoreductase 1; ACE: angiotensin-converting enzyme; PKC: protein kinase C beta II; NO: nitric oxide; NOS: nitric oxide synthesis; AR: aldose reductase; FBG: fasting blood glucose; AGE: advanced glycation end products; CTGF: connective tissue growth factor; CDKN2a: cyclin-dependent kinase inhibitor 2a; EED: embryonic ectoderm development; HOTAIR: HOX transcript antisense RNA; PRC2: polycomb repressive complex 2; EGF: epidermal growth factor; VEGF: vascular endothelial growth factor; FGF: fibroblast growth factor, ↓: inhibit; ↑: upregulate.
Nutrients 14 03768 g004
Table 2. The effects of genistein on podocytes in animal models.
Table 2. The effects of genistein on podocytes in animal models.
CellsTreatmentsEffects and MechanismsRef.
Mouse podocyte cells20 μM, 30 min prior to treatment for 24 hDecreasing D-ribose-induced ceramide accumulation, EV release and IL-1β secretion, and NLRP3 inflammasome[105]
Rat podocytes200 µM for 4 dDecreasing the expression of a-SMA protein and the percentage of a-SMA-positive cells stimulated by TGF-β1[106]
Mouse podocyte cell line, H-2Kb-tsA58, with high D-glucose20 µM for 6 hMaintaining the level of autophagy by inactivating mTOR signaling and the level of MyD88 siRNA[107]
Rat primary podocytes200 µM for 4 hCausing apoptosis of podocytes[108]
Mouse podocyte cell line60 μM for 20 hIncreasing cell loss under fluid flow stress[109]
Table 3. The effects of genistein attenuating kidney cancer cells.
Table 3. The effects of genistein attenuating kidney cancer cells.
Kidney Cancer
Cell Lines
Treatments (Genistein)Effects and MechanismsRef.
SMKT-R3 (human)50 g/mL for 15 minInhibiting tyrosine kinases and glycolipid sulfotransferase[152]
GRC-1 (human)20 and 40 mM/L for 72 hInhibiting the proliferation of kidney cell carcinoma cells; causing cell cycle arrest at the G1/M and G2/S phases[153]
kidney carcinoma cells SMKT-R-1,3 (human)4, 40, and 100 μg/mL under hypoxic conditions for 12 hSuppressing the expression of the angiogenic factors vascular endothelial growth factor and basic FGF[154]
A498, ACHN, and HEK-293 (human)10, 25, and 50 μ mol/L for 3 dInhibiting proliferation by decreasing DNA Methyltransferase and methyl-CpG-binding domain 2 activity and increasing HAT activity and induction of cell cycle arrest[155]
SMKT R-1, 2, 3, 4 lines (human)50 and 100 mg/mL for 48 hInhibiting cell proliferation, inducing apoptosis, and suppressing in vivo angiogenesis[156]
A-498; ATCC numbers: HTB44, HTB-47, 786-O, CRL-1932, and Caki-2 (human)25 µM for 4 dInhibiting Wnt signaling by regulating miR-1260b expression[157]
Human clear cell kidney carcinoma cell lines (ccRCC) (human)25 µM for 96 hReducing cell proliferation and migration by suppressing EED levels in PRC2 HOTAIR/PRC2 interaction, HOTAIR /PRC2 recruitment to the ZO-1 promoter, and enhancing ZO-1 transcription; inhibiting SNAIL transcription by reducing HOTAIR/SMARCB1 interaction[158]
HEK293, HK-2, 786-O, CAKI-1, 769-P, and CAKI-2 cell lines (human)25, 50, and 100 µM for 5 dInducing cell apoptosis and inhibiting cell proliferation of kidney cancer cells by increasing the expression of CDKN2a and decreasing CDKN2a methylation[159]
A-498 cells in nude mice (mouse)25 µM for 4 dInhibiting the expression of miR-21 in A-498 cells and in the tumors[160]
Kidney carcinoma cell (mouse)0.2 mL, 80 mg/kg/day, injected once a day for 14 dSuppressing tumor growth and decreasing MVD and VEGF levels[161]
Table 5. The effects of genistein attenuating hypertensive kidney disease.
Table 5. The effects of genistein attenuating hypertensive kidney disease.
AnimalModelTreatments (Genistein)Effects and MechanismsRef.
Wistar ratsFructose-fed hypertensive1 mg/kg/day in diet for 60 dLowering BP by restoring ACE, PKC-βII, and eNOS expression and preserving kidney ultrastructural integrity[181]
Sprague-Dawley rats2-kidney 1-clip kidney hypertensive5.0 mg/kg/day for 8 weeksRestoring nitric oxide, NOS activity, phosphorylated eNOS expression, and cGMP[182]
SHR-SPsDietary NaCl with hypertension0.6 mg/g diet for 9 weeksBlunting a dose-related increase in arterial pressure[183]
Wistar ratsIsolated perfused rat kidney15 mg/kg for 24 hReducing kidney vascular resistance relative to vehicle in isolated perfused kidney[115]
Table 7. The effects of genistein attenuating kidney fibrosis.
Table 7. The effects of genistein attenuating kidney fibrosis.
AnimalModelTreatments (Genistein)Effects and MechanismsRef.
C57BL miceUUO-induced kidney interstitial fibrosis10 mg/kg/body weight i.p. 24 h prior to the UUO for 7 dIncreasing kidney ALKBH5 expression, reducing RNA m6A levels, and ameliorating kidney damage.[203]
Sprague-Dawley ratsStreptozotocin-induced diabetic5 and 25 mg/kg, daily gavage for 8 weeksInhibiting oxidative stress by activating the Nrf2-HO-1/NQO1 pathway and alleviating kidney fibrosis by inhibiting the TGF-β1/Smad3 pathway[204]
C57BL/6 miceKidney fibrosis, UUO-induced10 mg/kg, intraperitoneal injection daily administered 1 day before UUORestoring Klotho via epigenetic histone acetylation and DNA demethylation[205]
Wistar ratsStandard pelletdiet (fructose-fed)1 mg/kg/day for 45 dDecreasing α-SMA expression and mitigating proliferation of connective tissue collagen deposition in perivascular and intraglomerular regions[206]
Human kidney tubular epithelial HK-2 cellsPTH-induced kidney interstitial fibrosis1, 25, 50, and 100 µM for 30 minInhibiting PTH-induced α-SMA expression, restoring E-cadherin expression, decreasing mRNA, protein expression, and activity of CTGF[207]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Peng, Q.; Li, Y.; Shang, J.; Huang, H.; Zhang, Y.; Ding, Y.; Liang, Y.; Xie, Z.; Chen, C. Effects of Genistein on Common Kidney Diseases. Nutrients 2022, 14, 3768. https://doi.org/10.3390/nu14183768

AMA Style

Peng Q, Li Y, Shang J, Huang H, Zhang Y, Ding Y, Liang Y, Xie Z, Chen C. Effects of Genistein on Common Kidney Diseases. Nutrients. 2022; 14(18):3768. https://doi.org/10.3390/nu14183768

Chicago/Turabian Style

Peng, Qianwen, Yuanyuan Li, Jia Shang, Haitao Huang, Yiming Zhang, Yueming Ding, Yipei Liang, Zhenxing Xie, and Chaoran Chen. 2022. "Effects of Genistein on Common Kidney Diseases" Nutrients 14, no. 18: 3768. https://doi.org/10.3390/nu14183768

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