Volume 2024, Issue 1 3475485
Research Article
Open Access

Sanziguben Polysaccharides Attenuate Renal Epithelial-Mesenchymal Transition in Diabetic Nephropathy through Nrf2-Mediated Regulation of TGF-β1/Smad7 Signaling Pathway

Jianing Zhang

Jianing Zhang

School of Pharmaceutical Sciences , Guangzhou University of Chinese Medicine , Guangzhou , 510006 , Guangdong, China , gzucm.edu.cn

Department of Clinical Medicine , Yuncheng Vocational Nursing College , Yuncheng , 044000 , Shanxi, China

Search for more papers by this author
Fan Wang

Fan Wang

School of Pharmaceutical Sciences , Guangzhou University of Chinese Medicine , Guangzhou , 510006 , Guangdong, China , gzucm.edu.cn

Search for more papers by this author
Chang Liu

Chang Liu

School of Pharmaceutical Sciences , Guangzhou University of Chinese Medicine , Guangzhou , 510006 , Guangdong, China , gzucm.edu.cn

Search for more papers by this author
Xiangyi Lu

Xiangyi Lu

School of Pharmaceutical Sciences , Guangzhou University of Chinese Medicine , Guangzhou , 510006 , Guangdong, China , gzucm.edu.cn

Search for more papers by this author
Weiping Xu

Weiping Xu

Guangdong Provincial People’s Hospital , Guangzhou , 519041 , Guangdong, China , gdghospital.org.cn

Search for more papers by this author
Yang Yu

Yang Yu

School of Pharmaceutical Sciences , Guangzhou University of Chinese Medicine , Guangzhou , 510006 , Guangdong, China , gzucm.edu.cn

Search for more papers by this author
Shasha Bai

Corresponding Author

Shasha Bai

School of Pharmaceutical Sciences , Guangzhou University of Chinese Medicine , Guangzhou , 510006 , Guangdong, China , gzucm.edu.cn

Search for more papers by this author
Zhilian Chen

Corresponding Author

Zhilian Chen

Guangzhou University of Traditional Chinese Medicine First Affiliated Hospital , Guangzhou , 510405 , Guangdong, China

Search for more papers by this author
First published: 05 August 2024
Citations: 1
Academic Editor: Muhammad Iqhrammullah

Abstract

Context. Sanziguben polysaccharides (SZP) have renal protection properties and can reduce renal fibrosis in diabetic nephropathy (DM). However, the mechanism of SZP’s renal protection effect is not yet clear. Objectives. Our study intended to clarify the mechanism of SZP’s renal protection effect in DM. Materials and Methods. In this study, streptozotocin-induced C57BL/6J diabetic nephropathy mice and high glucose combined with TGF-β1-induced EMT in HK-2 cells were used to investigate the effect of Sanziguben polysaccharides. ShRNA-constructed Nrf2 knockdown HK-2 cells were used to explore the role of Nrf2 in Sanziguben polysaccharides inhibiting epithelial-mesenchymal transition. Results. In vivo, the results showed that Sanziguben polysaccharides improved renal epithelial-mesenchymal transition and oxidative stress, and SZP was shown to activate the renal Nrf2, increase Smad7, and inhibit the expression of TGF-β1 (1.05- to 0.71-fold, 1.66- to 0.40-fold and 0.96- to 1.31-fold, respectively). In vitro, SZP ameliorated HK-2 cell epithelial-mesenchymal transition induced by HG combined with TGF-β1, increased the expression of Nrf2 and Smad7, and suppressed the expression of TGF-β1 (1.50- to 1.12-fold, 1.49- to 1.07-fold, and 0.94- to 1.38-fold, respectively). In addition, the above effects of Sanziguben polysaccharides on Nrf2 knockdown HK-2 cells were weakened. Conclusions. The findings suggest that Sanziguben polysaccharides may improve renal epithelial-mesenchymal transition in diabetic nephropathy through Nrf2-mediated regulation of TGF-β1/Smad7 signaling pathway.

1. Introduction

Diabetic nephropathy (DN), a global health problem, is one of the most common microvascular complications of diabetes. The main clinical symptom of diabetic nephropathy is proteinuria. Histopathologically characterized by the decreased glomerular filtration rate, glomerular damage, extracellular matrix accumulation, and tubular atrophy, in addition to more serious glomerular sclerosis and tubular interstitial fibrosis (TIF) [1], it was thought that DN was mainly a glomerular disease with vascular damage, but recent studies have found that the glomerular membrane and tubulointerstitial area have an accumulation of extracellular matrix (ECM), and it is hypothesized that TIF is a key step in end-stage DN [24].

The physiological process of epithelial-mesenchymal transition (EMT) may lead to tubulointerstitial fibrosis [5, 6]. Oldfield et al. observed a significant increase in α-SMA expression in tubular regions of the kidneys of T1DM patients with DN [7]. Rastaldi et al. found that tubular epithelial cells were altered in kidneys of DN patients, which indicated that cytoskeletal components have changed and further demonstrated that EMT was involved in the development of DN [8]. Several evidences have shown that the expression levels of EMT biomarker were not only associated with renal fibrosis in DN but also closely related to the progression of DN and the function of the kidney [9, 10]. EMT was first confirmed during embryonic development (type I EMT) and later found to exist during tissue repair and fibrosis (type II EMT), as well as during tumor invasion and migration (type III EMT). The three types of EMT have many common characteristics, such as destruction of cell-cell tight junctions, separation of epithelial cells and basement membranes, loss of epithelial cell morphology, acquisition of mesenchymal cell markers, and enhanced cell migration capabilities. Type II EMT is traditionally regarded as an important way to produce fibroblasts, which results in tissue fibrosis [11, 12]. The pathogenesis of EMT and drugs that can affect EMT has been a focus of recent research.

The EMT process is regulated by a variety of intracellular signaling pathways and cytokines. Studies have found that the accumulation of ROS can induce the secretion of inflammatory factors and profibrosis factors, thereby promoting the occurrence of renal fibrosis [13]. Keap1-Nrf2-ARE is the most important antioxidant signaling pathway in the body. After Nrf2 is activated, it induces the expression of downstream antioxidant genes such as NQO1 and HO-1 and antiapoptotic genes such as Bcl-2. It also inhibits the generation of ROS, promotes the recovery of the organism’s redox reaction, and maintains cell homeostasis [14]. TGF-β is one of the widely confirmed profibrosis factors. The TGF-β/Smads pathway is considered to be the most important signaling pathway that regulates EMT and tissue fibrosis [15]. Studies have shown that under pathological conditions associated with diabetic nephropathy, the TGF-β/Smads signaling pathway is activated, accompanied by the suppression of Smad7 expression, and the activation and translocation of Smad2/3 [16]. Specifically, in mice with overexpression of TGF-β1 or TGF-β1 receptor (TβRI), there have been reports of spontaneous fibrosis, especially in the kidney, leading to constitutive activation of the TGF-β signaling pathway. Oxidative stress induced by the diabetic environment and inflammatory mediators all promote the secretion of TGF-β1 in the organism [17]. Therefore, inhibiting the activation of the TGF-β pathway in pathological environments is one of the key strategies for the treatment of renal fibrosis.

The Sanziguben Recipe is a classic traditional Chinese medicine treatment for diabetic nephropathy, a mixture composed of Schisandra chinensis (Turcz.), Baill (Schisandraceae), Rosa laevigata Michx (Rosaceae), Phyllanthus emblica L. (Phyllanthaceae), and Gynostemma pentaphyllum (Thunb.) Makino (Cucurbitaceae), four herbs which are rich in polysaccharides. Previous studies have reported that selenium-containing polysaccharides isolated from the fruit of R. laevigata have antioxidant and neuroprotective effects [18]. The water-soluble polysaccharides isolated from P. emblica showed significant antioxidant capacity in terms of superoxide anion and hydroxyl radical scavenging and lipid peroxidation inhibition [19]. G. pentaphyllum polysaccharides have been reported to improve oxidative stress and anti-inflammatory effects in diabetic mice [20]. Moreover, S. chinensis polysaccharides can improve CSA-induced liver injury by activating the Nrf2 signaling pathway [21]. Previous studies in our laboratory showed that Sanziguben granule can activate the Nrf2 signaling pathway to improve renal EMT in diabetes nephropathy rats [22]. Due to the large amount of polysaccharides in the compound granules, we speculate that polysaccharides are the main pharmacological components. Besides, the polysaccharides mainly composed of glucans derived from Sanziguben Recipe, which were a type of heteropolysaccharides with pyran configurations, had been confirmed to ameliorate inflammatory states. However, the effect of Sanziguben polysaccharides (SZP) on EMT has not been studied.

Therefore, we hypothesized that SZP can improve DN oxidative stress homeostasis and prevent the progression of diabetic nephropathy EMT. We used a standard streptozotocin (STZ)-induced diabetic nephropathy mouse model and high glucose (HG) combined with a TGF-β1-induced human renal tubular epithelial (HK-2) cell EMT model to explore the protective mechanism of SZP on diabetic nephropathy EMT.

2. Materials and Methods

2.1. Sanziguben Polysaccharides

The medicinal materials of Sanziguben Recipe were purchased from Zhongshan Zhongzhi Traditional Chinese Medicine Co., Ltd. and identified. The voucher specimens (JYZ-20-ZJN, YGZ-20-ZJN, WWZ-20-ZJN, and JGL-20-ZJN, respectively) were deposited at the authors’ laboratory in the Guangzhou University of Chinese Medicine. The pulverized medicinal materials were degreased by refluxing with petroleum ether, and the dried medicinal powders were extracted twice in distilled water at 100°C under reflux for 2.5 hours each time, and the filtrates were combined and concentrated. We added ethanol to the concentrated solution to 80% of the concentration, stood still, and separated to obtain precipitation. Free proteins from polysaccharide solutions were removed by the Sevage method [23]. Finally, the solutions were concentrated and lyophilized to obtain SZP. Chemical composition analysis showed that the total sugar, uronic acid, sulfate, and protein contents of the obtained SZP were 53.06 ± 1.53%, 29.14 ± 0.73%, 5.60 ± 0.33%, and 7.95 ± 0.28%. The FT-IR (Nicolet™ iS™ 5N; Thermo Fisher Scientific) spectrum of SZP [24] and analysis of monosaccharide composition based on HPAEC-PAD (ICS5000; Thermo Fisher Scientific, Waltham, MA, USA) are shown in Supplementary Figure 1.

2.2. Reagents

Streptozotocin (STZ) was purchased from Sigma-Aldrich (Saint Louis, MO, USA); metformin was purchased from Sino-American Shanghai Squibb Pharmaceuticals Ltd (Shanghai, China); Super-Bradford Protein Assay Kit and BCA Protein Assay Kit were purchased from Beijing ComWin Biotech Co., Ltd. (Beijing, China); creatinine (Cre), total cholesterol (T-CHO), urea nitrogen, triglyceride (TG), malondialdehyde (MDA), catalase (CAT), and Superoxide Dismutase (SOD) assay kit were purchased from Nanjing Jiancheng Bioengineering Institute (Nanjing, China); Mouse TGF-β1 ELISA Kit was purchased from MultiSciences (Lianke) Biotech Co., Ltd. (Hangzhou, China); E-cadherin (ab76319), ZO-1 (ab96587), NQO1 (ab80588), and HO-1 (ab13248) were purchased from Abcam (Burlingame, CA, USA); vimentin (10366-1-AP), α-SMA (14395-1-AP), and Nrf2 (16396-1-AP) were purchased from Proteintech (Wuhan, China); Smad7 (sc-365846) was purchased from Santa Cruz Biotechnology, Inc. (Delaware Avenue, Santa Cruz, California); Horseradish peroxidase (HRP)-conjugated secondary antibodies were purchased from Beijing ComWin Biotech Co., Ltd. (Beijing, China); Recombinant human TGF-β1 (HEK293 derived) was purchased from Peprotech (Cranbury, NJ, USA).

2.3. Animals and Experimental Design

Male SPF C57BL/6J mice (purchased from Liaoning Changsheng Biotechnology Co., Ltd.) were 7 weeks old and weighed 22 ± 2 g. All animal care and experimental studies were approved following the guidelines of the Animal Ethics Committee of Guangzhou University of Chinese Medicine (license number: SCXK 2019-0202). Ethical review no. ZYD-2021-013, review date: January 29, 2021. After one week of adaptive feeding, the mice were fasted for 4–6 hours and treated with STZ (dissolved in pH 4.2 citric acid buffer) 50 mg/kg, continuously injected for 5 days. After one week, the fasting blood glucose of mice greater than 16.7 mmol/L was determined to be a model of diabetes. We continued to feed mice for 4 weeks, during which the urine protein levels were tested. When the STZ injection group and the normal group showed significant differences in urine protein, we started a gavage procedure for the following groups: control group; model group; MET group (300 mg/kg/day metformin); SZP low-dose group (SZP-L, 250 mg/kg/day SZP); SZP high-dose group (SZP-H, 500 mg/kg/day SZP). The control group and model group were given equal proportions of drinking water. The administration was continued for 8 weeks, during which weight was monitored, the amount of food and water consumed was measured, and fasting blood glucose was measured every two weeks. The animal experimental protocol is shown in Figure 1(a). Urine was collected 24 h after the administration. After sacrificing the mice, blood was taken from the orbital vein, and the serum was obtained by centrifugation at 3,500 rpm for 10 minutes; the liver and kidney were weighed and stored in tissue fixative or −80 refrigerator.

Details are in the caption following the image
Effects of SZP on general indicators and renal function in DN mice. (a) STZ-induced DN model establishment and administration time design. (b) The weekly weight change curve of mice during the treatment period. (c) The curve of blood glucose changes in mice every two weeks. (d) The kidney index of mice in each group was administered 8 weeks later. (e) 24-h urine protein in each group of mice after 8 weeks of administration. (f) Serum creatinine (Scr), (g) urea nitrogen (BUN), (h) total cholesterol (T-CHO), and (i) triglyceride (TG) content of each group of mice. The data are presented as means ± SD (n = 6). ###p < 0.001 vs. control,  ∗∗∗p < 0.001,  ∗∗p < 0.01, and  p < 0.05 vs. model group.
Details are in the caption following the image
Effects of SZP on general indicators and renal function in DN mice. (a) STZ-induced DN model establishment and administration time design. (b) The weekly weight change curve of mice during the treatment period. (c) The curve of blood glucose changes in mice every two weeks. (d) The kidney index of mice in each group was administered 8 weeks later. (e) 24-h urine protein in each group of mice after 8 weeks of administration. (f) Serum creatinine (Scr), (g) urea nitrogen (BUN), (h) total cholesterol (T-CHO), and (i) triglyceride (TG) content of each group of mice. The data are presented as means ± SD (n = 6). ###p < 0.001 vs. control,  ∗∗∗p < 0.001,  ∗∗p < 0.01, and  p < 0.05 vs. model group.
Details are in the caption following the image
Effects of SZP on general indicators and renal function in DN mice. (a) STZ-induced DN model establishment and administration time design. (b) The weekly weight change curve of mice during the treatment period. (c) The curve of blood glucose changes in mice every two weeks. (d) The kidney index of mice in each group was administered 8 weeks later. (e) 24-h urine protein in each group of mice after 8 weeks of administration. (f) Serum creatinine (Scr), (g) urea nitrogen (BUN), (h) total cholesterol (T-CHO), and (i) triglyceride (TG) content of each group of mice. The data are presented as means ± SD (n = 6). ###p < 0.001 vs. control,  ∗∗∗p < 0.001,  ∗∗p < 0.01, and  p < 0.05 vs. model group.
Details are in the caption following the image
Effects of SZP on general indicators and renal function in DN mice. (a) STZ-induced DN model establishment and administration time design. (b) The weekly weight change curve of mice during the treatment period. (c) The curve of blood glucose changes in mice every two weeks. (d) The kidney index of mice in each group was administered 8 weeks later. (e) 24-h urine protein in each group of mice after 8 weeks of administration. (f) Serum creatinine (Scr), (g) urea nitrogen (BUN), (h) total cholesterol (T-CHO), and (i) triglyceride (TG) content of each group of mice. The data are presented as means ± SD (n = 6). ###p < 0.001 vs. control,  ∗∗∗p < 0.001,  ∗∗p < 0.01, and  p < 0.05 vs. model group.
Details are in the caption following the image
Effects of SZP on general indicators and renal function in DN mice. (a) STZ-induced DN model establishment and administration time design. (b) The weekly weight change curve of mice during the treatment period. (c) The curve of blood glucose changes in mice every two weeks. (d) The kidney index of mice in each group was administered 8 weeks later. (e) 24-h urine protein in each group of mice after 8 weeks of administration. (f) Serum creatinine (Scr), (g) urea nitrogen (BUN), (h) total cholesterol (T-CHO), and (i) triglyceride (TG) content of each group of mice. The data are presented as means ± SD (n = 6). ###p < 0.001 vs. control,  ∗∗∗p < 0.001,  ∗∗p < 0.01, and  p < 0.05 vs. model group.
Details are in the caption following the image
Effects of SZP on general indicators and renal function in DN mice. (a) STZ-induced DN model establishment and administration time design. (b) The weekly weight change curve of mice during the treatment period. (c) The curve of blood glucose changes in mice every two weeks. (d) The kidney index of mice in each group was administered 8 weeks later. (e) 24-h urine protein in each group of mice after 8 weeks of administration. (f) Serum creatinine (Scr), (g) urea nitrogen (BUN), (h) total cholesterol (T-CHO), and (i) triglyceride (TG) content of each group of mice. The data are presented as means ± SD (n = 6). ###p < 0.001 vs. control,  ∗∗∗p < 0.001,  ∗∗p < 0.01, and  p < 0.05 vs. model group.
Details are in the caption following the image
Effects of SZP on general indicators and renal function in DN mice. (a) STZ-induced DN model establishment and administration time design. (b) The weekly weight change curve of mice during the treatment period. (c) The curve of blood glucose changes in mice every two weeks. (d) The kidney index of mice in each group was administered 8 weeks later. (e) 24-h urine protein in each group of mice after 8 weeks of administration. (f) Serum creatinine (Scr), (g) urea nitrogen (BUN), (h) total cholesterol (T-CHO), and (i) triglyceride (TG) content of each group of mice. The data are presented as means ± SD (n = 6). ###p < 0.001 vs. control,  ∗∗∗p < 0.001,  ∗∗p < 0.01, and  p < 0.05 vs. model group.
Details are in the caption following the image
Effects of SZP on general indicators and renal function in DN mice. (a) STZ-induced DN model establishment and administration time design. (b) The weekly weight change curve of mice during the treatment period. (c) The curve of blood glucose changes in mice every two weeks. (d) The kidney index of mice in each group was administered 8 weeks later. (e) 24-h urine protein in each group of mice after 8 weeks of administration. (f) Serum creatinine (Scr), (g) urea nitrogen (BUN), (h) total cholesterol (T-CHO), and (i) triglyceride (TG) content of each group of mice. The data are presented as means ± SD (n = 6). ###p < 0.001 vs. control,  ∗∗∗p < 0.001,  ∗∗p < 0.01, and  p < 0.05 vs. model group.
Details are in the caption following the image
Effects of SZP on general indicators and renal function in DN mice. (a) STZ-induced DN model establishment and administration time design. (b) The weekly weight change curve of mice during the treatment period. (c) The curve of blood glucose changes in mice every two weeks. (d) The kidney index of mice in each group was administered 8 weeks later. (e) 24-h urine protein in each group of mice after 8 weeks of administration. (f) Serum creatinine (Scr), (g) urea nitrogen (BUN), (h) total cholesterol (T-CHO), and (i) triglyceride (TG) content of each group of mice. The data are presented as means ± SD (n = 6). ###p < 0.001 vs. control,  ∗∗∗p < 0.001,  ∗∗p < 0.01, and  p < 0.05 vs. model group.

2.4. Cell Culture

Human proximal renal tubular epithelial cells (HK-2 cell) and ShRNA constructed Nrf2 stable knockdown HK-2 cell line (HK-2-Nrf2-KD cell) (purchased from Xi’an CytoBiotech Biological Technology Co., Ltd.) were cultured in DMEM/F12 (Gibco) containing 10% FBS fetal bovine serum and 1% antibiotic-antimycotic solution. The EMT model of HK-2 cells was induced by HG (30 mM glucose) combined with TGF-β1. The SZP treatment group increased the required dose of SZP according to the model group. After 5 days of continuous cultivation under the above conditions, the total protein of each group was extracted for measurement.

2.5. Kidney Pathological Staining and Immunohistochemistry

For pathological staining of kidney tissue, tissue was fixed in 4% paraformaldehyde, dehydrated, and embedded in paraffin, sliced, and stained with H&E, PAS, MASSON, and Sirius red according to the instructions.

For immunohistochemistry, paraffin sections were deparaffinized to water, and the EDTA (PH9.0) antigen retrieval solution was used for antigen retrieval; sections were incubated with 3% hydrogen peroxide in the dark for 25 minutes and washed with PBS. Then, 3% BSA was used for blocking at room temperature for 30 minutes and washed and incubated overnight at 4° with the primary antibody TGF-β, E-cadherin, or α-SMA. Sections were washed with PBS, and then the secondary antibody (HRP-conjugated) was added and incubated at room temperature for 50 min. Sections were washed and then slightly dried. DAB was added as a color developing solution, which was monitored under the microscope for optimal time. The section was then washed with tap water to stop the color development, the cell nucleus was counterstained, and the section was dehydrated and mounted.

2.6. Detection of Biochemical Indexes

All serum or tissue biochemical indicators were tested according to the instructions of the corresponding kit.

2.7. Cell CCK-8 Assay

HK-2 cells were seeded in 96-well plates at 1 × 104 cells/well, cultured for 24 h to adhere, and then treated for 48 h in the following groups: 200 μL/well of the cell-free blank medium group, HK-2 cells were treated with SZP groups 200 μL/well (SZP administration concentrations were 0, 25 μg/ml, 50 μg/ml, 100 μg/ml, 200 μg/ml, 300 μg/ml, 400 μg/ml, and 500 μg/ml), and repeated 5 replicate wells. After 48 hours, the medium in the wells was discarded and a new DMEM/F12 complete medium and 10 μL of CCK-8 reagent were added to each well and incubated in the incubator for 1.5 hours in the dark. Finally, the OD values were detected at 450 nm of the microplate reader for statistics.

2.8. Western Blot

The kidney tissue was homogenized with RIPA lysis solution and centrifuged to obtain the total renal protein supernatant. Cells were washed twice with PBS, and then the lysis buffer was added and collected in a centrifuge tube. The tubes were centrifuged to obtain the HK-2 cell total protein supernatant. BCA kit was used to detect protein content. After adding the loading buffer, the protein was denatured at 100°C for 5 minutes, and the protein was separated by SDS-Page gel electrophoresis. After transfer to a membrane, the membrane was blocked with 5% skimmed milk powder. The membrane was incubated with a primary antibody overnight at 4°C, and then the HRP-conjugated secondary antibody was used for incubation at room temperature for 1 hour. After adding the chemiluminescence substrate, we used an automatic chemiluminescence instrument for exposure.

2.9. Statistical Analysis

All data are expressed as mean ± SD. P<0.05 was considered statistically significant. There was statistical significance using one-way ANOVA followed by LSD or Dunn’s test for multiple comparisons. GraphPad prism 8 software was used to graph data. Image J software was used for immunohistochemistry and western blot quantification.

3. Results

3.1. SZP Improves the Disease Status of STZ-Induced DN Mice

During the administration period, the body weight, blood sugar, diet, and drinking were measured. In our experiment, the weight of the diabetic mice was significantly lower than that of the normal group. During the administration period, the weight of the mice in the normal group increased with time and the weight of the model mice decreased due to the influence of diabetes. After 8 weeks administration, body weight of mice-administered metformin and SZP were improved compared with the model group (Figure 1(b)). The fasting blood glucose of normal C57BL/6J mice was in the range of 5–10, compared to 20–33 in the STZ-injected mice. After administration, the blood sugar of diabetic nephropathy mice treated with metformin and SZP-H decreased significantly (Figure 1(c)). By measuring the water intake and diet of the mice weekly, it was found that the SZP treatment can improve the symptoms of polyphagia and polyphagia in diabetic mice (Supplementary Figure 2). The kidney index is shown in Figure 1(d), mice in the model group developed kidney enlargement, and SZP treatment reduced the kidney index.

3.2. Effects of SZP on Renal Function in STZ-Induced DN Mice

In our experiment, 24 h urine protein, serum creatinine, and urea nitrogen of model group mice were significantly higher than those of normal mice and the urinary protein content was 10 times higher than the normal group, suggesting that there is kidney dysfunction in mice with diabetic nephropathy. SZP significantly reduced 24 h urine protein measures, serum creatinine, and urea nitrogen levels, which indicate that SZP can improve diabetes-induced kidney damage (Figure 1(e) and 1(f)). By observing the T-CHO and TG, we found that DN mice had abnormal blood lipid metabolism. The T-CHO and TG of the model group were significantly increased, and these two indicators were also reduced after SZP treatment (Figures 1(h) and 1(i)). These indicate that SZP has protective effects on kidney function and blood lipid metabolism in diabetic nephropathy mice.

3.3. SZP Reduces Kidney Damage and Fibrosis in DN Mice

We observed the pathological changes in mouse kidney tissue, as shown in Figure 2. HE staining of the model group showed that there was glomerular mesangial hyperplasia, glomerular sclerosis, vacuolar degeneration of renal tubular epithelial cells, and compensatory renal tubule dilation (marked by arrows). SZP treatment attenuated glomerular damage and renal tubular disease. In PAS staining, the thickening of the basement membrane of the renal tubules in the model group was observed (marked by arrows), but the basement membrane of the tubules returned to normal in SZP-L and SZP-H groups. MASSON staining showed a large number of blue-stained collagen fibers in the tubular interstitium of the model group (pointed by arrows), and the area of collagen fibers decreased after metformin and SZP treatments. Similarly, Sirius red staining showed that there were a large number of red-stained collagen fibers in the tubular interstitium (pointed by arrows). Furthermore, under the polarized light microscope, the yellow strong refractive type I collagen fibers in the model group increased significantly, while the green weak refractive type III collagen fibers did not change significantly. Both SZP-L and SZP-H significantly reduced type I collagen fibers but had no significant effect on type III collagen fibers.

Details are in the caption following the image
Effects of SZP on kidney tissue in DN mice. Representative images of pathological staining of kidney tissues. Kidney HE staining (200×), the arrows in the _x0010_model group indicate glomerular sclerosis, vacuolar degeneration of renal tubular epithelial cells, and renal tubule dilatation; PAS staining (200×), the arrows in the model group indicate renal tubular basement membrane increase thick; MASSON staining (200×), the arrows indicate the collagen fibers stained blue in the renal tubule interstitium, which represents tubular interstitial fibrosis; Sirius red staining (200×) includes normal light and polarized light views, and arrows indicate collagen fibril-positive sites.

3.4. SZP Can Inhibit the Kidney EMT in Mice with DN

E-cadherin and ZO-1 are the proteins associated with tight junctions of epithelial cells, while α-SMA and vimentin are markers of mesenchymal cells. During the EMT process, there is the loss of tight junction proteins and elevated mesenchymal markers. Immunohistochemical analysis showed that E-cadherin in the renal tubules of model mice was decreased and α-SMA increased (Figures 3(a), 3(b), 3(c)). Similarly, western blot detection of total renal tissue protein found that the expressions of E-cadherin and ZO-1 proteins in the model group were decreased, but α-SMA and vimentin were significantly increased. This phenomenon was reversed after the administration of metformin and SZP (Figures 3(d), 3(e), 3(f), 3(g), 3(h)). This indicates that EMT occurred in the kidney of diabetic nephropathy mice, and SZP can prevent the occurrence of EMT to a certain extent. This may be why SZP reduces DN renal tissue fibrosis.

Details are in the caption following the image
Effect of SZP on EMT of DN mice kidney. (a) Representative images of immunohistochemical detection of E-cadherin and α-SMA expression in kidney tissue (400×). (b, c) E-cadherin, α-SMA immunohistochemical protein expression level statistics. (d) Western blot of EMT-related proteins in kidney tissue: ZO-1, E-cadherin, α-SMA, and vimentin. (e–h) Quantifications of (d). The data are presented as means ± SD (n = 3). ###p < 0.001, ##p < 0.01, and #p < 0.05 vs. control;  ∗∗∗p < 0.001,  ∗∗p < 0.01, and  p < 0.05 vs. model group.
Details are in the caption following the image
Effect of SZP on EMT of DN mice kidney. (a) Representative images of immunohistochemical detection of E-cadherin and α-SMA expression in kidney tissue (400×). (b, c) E-cadherin, α-SMA immunohistochemical protein expression level statistics. (d) Western blot of EMT-related proteins in kidney tissue: ZO-1, E-cadherin, α-SMA, and vimentin. (e–h) Quantifications of (d). The data are presented as means ± SD (n = 3). ###p < 0.001, ##p < 0.01, and #p < 0.05 vs. control;  ∗∗∗p < 0.001,  ∗∗p < 0.01, and  p < 0.05 vs. model group.
Details are in the caption following the image
Effect of SZP on EMT of DN mice kidney. (a) Representative images of immunohistochemical detection of E-cadherin and α-SMA expression in kidney tissue (400×). (b, c) E-cadherin, α-SMA immunohistochemical protein expression level statistics. (d) Western blot of EMT-related proteins in kidney tissue: ZO-1, E-cadherin, α-SMA, and vimentin. (e–h) Quantifications of (d). The data are presented as means ± SD (n = 3). ###p < 0.001, ##p < 0.01, and #p < 0.05 vs. control;  ∗∗∗p < 0.001,  ∗∗p < 0.01, and  p < 0.05 vs. model group.
Details are in the caption following the image
Effect of SZP on EMT of DN mice kidney. (a) Representative images of immunohistochemical detection of E-cadherin and α-SMA expression in kidney tissue (400×). (b, c) E-cadherin, α-SMA immunohistochemical protein expression level statistics. (d) Western blot of EMT-related proteins in kidney tissue: ZO-1, E-cadherin, α-SMA, and vimentin. (e–h) Quantifications of (d). The data are presented as means ± SD (n = 3). ###p < 0.001, ##p < 0.01, and #p < 0.05 vs. control;  ∗∗∗p < 0.001,  ∗∗p < 0.01, and  p < 0.05 vs. model group.
Details are in the caption following the image
Effect of SZP on EMT of DN mice kidney. (a) Representative images of immunohistochemical detection of E-cadherin and α-SMA expression in kidney tissue (400×). (b, c) E-cadherin, α-SMA immunohistochemical protein expression level statistics. (d) Western blot of EMT-related proteins in kidney tissue: ZO-1, E-cadherin, α-SMA, and vimentin. (e–h) Quantifications of (d). The data are presented as means ± SD (n = 3). ###p < 0.001, ##p < 0.01, and #p < 0.05 vs. control;  ∗∗∗p < 0.001,  ∗∗p < 0.01, and  p < 0.05 vs. model group.
Details are in the caption following the image
Effect of SZP on EMT of DN mice kidney. (a) Representative images of immunohistochemical detection of E-cadherin and α-SMA expression in kidney tissue (400×). (b, c) E-cadherin, α-SMA immunohistochemical protein expression level statistics. (d) Western blot of EMT-related proteins in kidney tissue: ZO-1, E-cadherin, α-SMA, and vimentin. (e–h) Quantifications of (d). The data are presented as means ± SD (n = 3). ###p < 0.001, ##p < 0.01, and #p < 0.05 vs. control;  ∗∗∗p < 0.001,  ∗∗p < 0.01, and  p < 0.05 vs. model group.
Details are in the caption following the image
Effect of SZP on EMT of DN mice kidney. (a) Representative images of immunohistochemical detection of E-cadherin and α-SMA expression in kidney tissue (400×). (b, c) E-cadherin, α-SMA immunohistochemical protein expression level statistics. (d) Western blot of EMT-related proteins in kidney tissue: ZO-1, E-cadherin, α-SMA, and vimentin. (e–h) Quantifications of (d). The data are presented as means ± SD (n = 3). ###p < 0.001, ##p < 0.01, and #p < 0.05 vs. control;  ∗∗∗p < 0.001,  ∗∗p < 0.01, and  p < 0.05 vs. model group.
Details are in the caption following the image
Effect of SZP on EMT of DN mice kidney. (a) Representative images of immunohistochemical detection of E-cadherin and α-SMA expression in kidney tissue (400×). (b, c) E-cadherin, α-SMA immunohistochemical protein expression level statistics. (d) Western blot of EMT-related proteins in kidney tissue: ZO-1, E-cadherin, α-SMA, and vimentin. (e–h) Quantifications of (d). The data are presented as means ± SD (n = 3). ###p < 0.001, ##p < 0.01, and #p < 0.05 vs. control;  ∗∗∗p < 0.001,  ∗∗p < 0.01, and  p < 0.05 vs. model group.

3.5. SZP Improves Oxidative Stress Levels in DN Mice by Activating the Nrf2 Signaling Pathway

In Figures 4(a), 4(b), 4(c), the expression of serum MDA in the model group was shown to increase, but it decreased significantly after SZP administration. The levels of SOD and CAT in the kidney of DN mice were significantly lower than those in the normal group, while expression in the SZP groups was significantly increased after treatment. MDA is a lipid peroxide produced during oxidation reaction, and the increase in its content indicates that the body is attacked by free radicals. Since SOD and CAT exert antioxidant effects in vivo, the content of both suggests the body’s ability to scavenge oxygen free radicals. The above results indicate that SZP administration improves the level of oxidative stress in DN. By detecting Nrf2 and its downstream markers HO-1 and NQO1 in the total protein of kidney tissue, we found that SZP-H significantly increased the level of Nrf2, HO-1, and NQO1 (Figures 4(d), 4(e), 4(f), 4(g)). Therefore, these results indicate that SZP could improve the oxidative stress level of DN mice by activating the Nrf2 signaling pathway.

Details are in the caption following the image
Effects of SZP on oxidative stress and Nrf2 signal in DN mice. (a) Serum MDA (n = 6). (b) Kidney tissue SOD content (n = 6). (c) Kidney tissue CAT content (n = 6); (d) western blot of Nrf2, HO-1, and NQO1. (e-g) Quantifications of (d) (n = 3). The data are presented as means ± SD. ###p < 0.001, ##p < 0.01, and #p < 0.05 vs. control;  ∗∗∗p < 0.001,  ∗∗p < 0.01, and  p < 0.05 vs. model group.
Details are in the caption following the image
Effects of SZP on oxidative stress and Nrf2 signal in DN mice. (a) Serum MDA (n = 6). (b) Kidney tissue SOD content (n = 6). (c) Kidney tissue CAT content (n = 6); (d) western blot of Nrf2, HO-1, and NQO1. (e-g) Quantifications of (d) (n = 3). The data are presented as means ± SD. ###p < 0.001, ##p < 0.01, and #p < 0.05 vs. control;  ∗∗∗p < 0.001,  ∗∗p < 0.01, and  p < 0.05 vs. model group.
Details are in the caption following the image
Effects of SZP on oxidative stress and Nrf2 signal in DN mice. (a) Serum MDA (n = 6). (b) Kidney tissue SOD content (n = 6). (c) Kidney tissue CAT content (n = 6); (d) western blot of Nrf2, HO-1, and NQO1. (e-g) Quantifications of (d) (n = 3). The data are presented as means ± SD. ###p < 0.001, ##p < 0.01, and #p < 0.05 vs. control;  ∗∗∗p < 0.001,  ∗∗p < 0.01, and  p < 0.05 vs. model group.
Details are in the caption following the image
Effects of SZP on oxidative stress and Nrf2 signal in DN mice. (a) Serum MDA (n = 6). (b) Kidney tissue SOD content (n = 6). (c) Kidney tissue CAT content (n = 6); (d) western blot of Nrf2, HO-1, and NQO1. (e-g) Quantifications of (d) (n = 3). The data are presented as means ± SD. ###p < 0.001, ##p < 0.01, and #p < 0.05 vs. control;  ∗∗∗p < 0.001,  ∗∗p < 0.01, and  p < 0.05 vs. model group.
Details are in the caption following the image
Effects of SZP on oxidative stress and Nrf2 signal in DN mice. (a) Serum MDA (n = 6). (b) Kidney tissue SOD content (n = 6). (c) Kidney tissue CAT content (n = 6); (d) western blot of Nrf2, HO-1, and NQO1. (e-g) Quantifications of (d) (n = 3). The data are presented as means ± SD. ###p < 0.001, ##p < 0.01, and #p < 0.05 vs. control;  ∗∗∗p < 0.001,  ∗∗p < 0.01, and  p < 0.05 vs. model group.
Details are in the caption following the image
Effects of SZP on oxidative stress and Nrf2 signal in DN mice. (a) Serum MDA (n = 6). (b) Kidney tissue SOD content (n = 6). (c) Kidney tissue CAT content (n = 6); (d) western blot of Nrf2, HO-1, and NQO1. (e-g) Quantifications of (d) (n = 3). The data are presented as means ± SD. ###p < 0.001, ##p < 0.01, and #p < 0.05 vs. control;  ∗∗∗p < 0.001,  ∗∗p < 0.01, and  p < 0.05 vs. model group.
Details are in the caption following the image
Effects of SZP on oxidative stress and Nrf2 signal in DN mice. (a) Serum MDA (n = 6). (b) Kidney tissue SOD content (n = 6). (c) Kidney tissue CAT content (n = 6); (d) western blot of Nrf2, HO-1, and NQO1. (e-g) Quantifications of (d) (n = 3). The data are presented as means ± SD. ###p < 0.001, ##p < 0.01, and #p < 0.05 vs. control;  ∗∗∗p < 0.001,  ∗∗p < 0.01, and  p < 0.05 vs. model group.

3.6. SZP Inhibits the Expression of EMT-Related TGF-β1/Smad7 Signaling Pathway in Diabetic Nephropathy Mice

Through the detection of serum TGF-β1, we found that the serum TGF-β1 content of mice in the model group increased, which was significantly decreased after the administration of SZP (Figure 5(a)). Using immunohistochemistry (Figure 5(b)) and western blot analysis (Figures 5(c), 5(d), 5(e)) measured changes in TGF-β1 in the kidney tissues of mice. TGF-β1 was significantly increased in the model group, while TGF-β1 in the MET, SZP-L, and SZP-H administration groups was significantly lower than that of the model group. Furthermore, Smad7 plays an antagonistic role in the TGF-β1 signaling pathway and showed the opposite trend, and SZP increased the expression of smad7. This indicates that the effect of SZP’s inhibition of EMT is closely related to its inhibition of the renal TGF-β1 signaling pathway in DN mice.

Details are in the caption following the image
Effects of SZP on the expression level of the TGF-β1/Smad7 signaling pathway in DN mice. (a) ELISA detected the expression level of serum TGF-β1 in mice (n = 6). (b) Representative immunohistochemical images of mice kidney tissue TGF-β1 and TGF-β1 immunohistochemical quantification (n = 3). (c) Western blot detected kidney TGF-β1 and Smad7 expression. (d, e) Quantifications of C (n = 3). The data are presented as means ± SD. ###p < 0.001, ##p < 0.01, and #p < 0.05 vs. control;  ∗∗∗p < 0.001, ∗∗p<0.01, and  p < 0.05 vs. model group.
Details are in the caption following the image
Effects of SZP on the expression level of the TGF-β1/Smad7 signaling pathway in DN mice. (a) ELISA detected the expression level of serum TGF-β1 in mice (n = 6). (b) Representative immunohistochemical images of mice kidney tissue TGF-β1 and TGF-β1 immunohistochemical quantification (n = 3). (c) Western blot detected kidney TGF-β1 and Smad7 expression. (d, e) Quantifications of C (n = 3). The data are presented as means ± SD. ###p < 0.001, ##p < 0.01, and #p < 0.05 vs. control;  ∗∗∗p < 0.001, ∗∗p<0.01, and  p < 0.05 vs. model group.
Details are in the caption following the image
Effects of SZP on the expression level of the TGF-β1/Smad7 signaling pathway in DN mice. (a) ELISA detected the expression level of serum TGF-β1 in mice (n = 6). (b) Representative immunohistochemical images of mice kidney tissue TGF-β1 and TGF-β1 immunohistochemical quantification (n = 3). (c) Western blot detected kidney TGF-β1 and Smad7 expression. (d, e) Quantifications of C (n = 3). The data are presented as means ± SD. ###p < 0.001, ##p < 0.01, and #p < 0.05 vs. control;  ∗∗∗p < 0.001, ∗∗p<0.01, and  p < 0.05 vs. model group.
Details are in the caption following the image
Effects of SZP on the expression level of the TGF-β1/Smad7 signaling pathway in DN mice. (a) ELISA detected the expression level of serum TGF-β1 in mice (n = 6). (b) Representative immunohistochemical images of mice kidney tissue TGF-β1 and TGF-β1 immunohistochemical quantification (n = 3). (c) Western blot detected kidney TGF-β1 and Smad7 expression. (d, e) Quantifications of C (n = 3). The data are presented as means ± SD. ###p < 0.001, ##p < 0.01, and #p < 0.05 vs. control;  ∗∗∗p < 0.001, ∗∗p<0.01, and  p < 0.05 vs. model group.
Details are in the caption following the image
Effects of SZP on the expression level of the TGF-β1/Smad7 signaling pathway in DN mice. (a) ELISA detected the expression level of serum TGF-β1 in mice (n = 6). (b) Representative immunohistochemical images of mice kidney tissue TGF-β1 and TGF-β1 immunohistochemical quantification (n = 3). (c) Western blot detected kidney TGF-β1 and Smad7 expression. (d, e) Quantifications of C (n = 3). The data are presented as means ± SD. ###p < 0.001, ##p < 0.01, and #p < 0.05 vs. control;  ∗∗∗p < 0.001, ∗∗p<0.01, and  p < 0.05 vs. model group.

3.7. SZP Improves EMT Development in Human Renal Tubular Epithelial Cells through Nrf2/Smad7

As shown in Figure 6(a), we observed the effect of different doses of SZP on the proliferation of HK-2 cells through the CCK-8 experiment. The results showed that the dose of SZP greater than or equal to 100 µg/ml had a promoting effect on the proliferation of HK-2 cells. In order to exclude the interference of this phenomenon on the experimental results, we chose 50 µg/ml as the treatment dose. In Figure 6(b), we observed that SZP treatment ameliorated HG plus TGF-β1-induced loss of E-cadherin and gain of vimentin in HK-2 cells. Moreover, Nrf2 expression was elevated after SZP treatment. These were consistent with the results found in mice. In addition, we observed the morphology of HK-2 cells. Compared with the normal group, HK-2 cells in the model group showed significant morphological changes, with sparse cell arrangement and fibroblast-like changes, arranged in a long spindle-shaped vortex, which were improved after SZP treatment (Supplementary Figure 3).

Details are in the caption following the image
Effects of SZP on EMT of HK-2 cells stimulated by HG combined with TGF-β1. (a) The effect of different concentrations of SZP on the proliferation of HK-2 cells detected by CCK-8 (n = 5). (b) Western blot detected HK-2 cell E-cadherin, vimentin, and Nrf2 expression. (c) Western blot detection and quantitative analysis of Nrf2 in HK-2 and HK-2-Nrf2-knockdown cells. (d) Western blotting detected the expressions of E-cadherin, vimentin, Smad7, TGFβ1, and Nrf2 in HK-2 and HK-2-Nrf2-KD cells stimulated by HG combined with TGFβ1. (e) Quantifications of (d). The data are presented as means ± SD (n = 3). ###p < 0.001, ##p < 0.01, and #p < 0.05 vs. HK-2 or HK-2-control group;  ∗∗∗p < 0.001,  ∗∗p < 0.01, and  p < 0.05 vs. HK-2-model.  p < 0.05 vs. HK-2-SZP.  p < 0.05 vs. HK-2-SZP.
Details are in the caption following the image
Effects of SZP on EMT of HK-2 cells stimulated by HG combined with TGF-β1. (a) The effect of different concentrations of SZP on the proliferation of HK-2 cells detected by CCK-8 (n = 5). (b) Western blot detected HK-2 cell E-cadherin, vimentin, and Nrf2 expression. (c) Western blot detection and quantitative analysis of Nrf2 in HK-2 and HK-2-Nrf2-knockdown cells. (d) Western blotting detected the expressions of E-cadherin, vimentin, Smad7, TGFβ1, and Nrf2 in HK-2 and HK-2-Nrf2-KD cells stimulated by HG combined with TGFβ1. (e) Quantifications of (d). The data are presented as means ± SD (n = 3). ###p < 0.001, ##p < 0.01, and #p < 0.05 vs. HK-2 or HK-2-control group;  ∗∗∗p < 0.001,  ∗∗p < 0.01, and  p < 0.05 vs. HK-2-model.  p < 0.05 vs. HK-2-SZP.  p < 0.05 vs. HK-2-SZP.
Details are in the caption following the image
Effects of SZP on EMT of HK-2 cells stimulated by HG combined with TGF-β1. (a) The effect of different concentrations of SZP on the proliferation of HK-2 cells detected by CCK-8 (n = 5). (b) Western blot detected HK-2 cell E-cadherin, vimentin, and Nrf2 expression. (c) Western blot detection and quantitative analysis of Nrf2 in HK-2 and HK-2-Nrf2-knockdown cells. (d) Western blotting detected the expressions of E-cadherin, vimentin, Smad7, TGFβ1, and Nrf2 in HK-2 and HK-2-Nrf2-KD cells stimulated by HG combined with TGFβ1. (e) Quantifications of (d). The data are presented as means ± SD (n = 3). ###p < 0.001, ##p < 0.01, and #p < 0.05 vs. HK-2 or HK-2-control group;  ∗∗∗p < 0.001,  ∗∗p < 0.01, and  p < 0.05 vs. HK-2-model.  p < 0.05 vs. HK-2-SZP.  p < 0.05 vs. HK-2-SZP.
Details are in the caption following the image
Effects of SZP on EMT of HK-2 cells stimulated by HG combined with TGF-β1. (a) The effect of different concentrations of SZP on the proliferation of HK-2 cells detected by CCK-8 (n = 5). (b) Western blot detected HK-2 cell E-cadherin, vimentin, and Nrf2 expression. (c) Western blot detection and quantitative analysis of Nrf2 in HK-2 and HK-2-Nrf2-knockdown cells. (d) Western blotting detected the expressions of E-cadherin, vimentin, Smad7, TGFβ1, and Nrf2 in HK-2 and HK-2-Nrf2-KD cells stimulated by HG combined with TGFβ1. (e) Quantifications of (d). The data are presented as means ± SD (n = 3). ###p < 0.001, ##p < 0.01, and #p < 0.05 vs. HK-2 or HK-2-control group;  ∗∗∗p < 0.001,  ∗∗p < 0.01, and  p < 0.05 vs. HK-2-model.  p < 0.05 vs. HK-2-SZP.  p < 0.05 vs. HK-2-SZP.
Details are in the caption following the image
Effects of SZP on EMT of HK-2 cells stimulated by HG combined with TGF-β1. (a) The effect of different concentrations of SZP on the proliferation of HK-2 cells detected by CCK-8 (n = 5). (b) Western blot detected HK-2 cell E-cadherin, vimentin, and Nrf2 expression. (c) Western blot detection and quantitative analysis of Nrf2 in HK-2 and HK-2-Nrf2-knockdown cells. (d) Western blotting detected the expressions of E-cadherin, vimentin, Smad7, TGFβ1, and Nrf2 in HK-2 and HK-2-Nrf2-KD cells stimulated by HG combined with TGFβ1. (e) Quantifications of (d). The data are presented as means ± SD (n = 3). ###p < 0.001, ##p < 0.01, and #p < 0.05 vs. HK-2 or HK-2-control group;  ∗∗∗p < 0.001,  ∗∗p < 0.01, and  p < 0.05 vs. HK-2-model.  p < 0.05 vs. HK-2-SZP.  p < 0.05 vs. HK-2-SZP.

In addition, compared with HK-2 cell, the protein expression of Nrf2 was significantly reduced in the HK-2-Nrf2-KD stable cell line (Figure 6(c)). HG combined with TGF-β1 induced HK-2-Nrf2-KD cells to produce the same EMT changes as HK-2 cells. We observed that Nrf2 was not significantly elevated by SZP in HK-2-Nrf2-KD cells. After SZP administration, the expression levels of E-cadherin and vimentin were reversed in HK-2 cells, while the effect of SZP disappeared in HK-2-Nrf2-KD cells. Moreover, the expression of Smad7 was significantly increased in HK-2 cells after SZP treatment; however, this phenomenon decreased with the knockdown of Nrf2. Similarly, SZP treatment reduced the expression of TGF-β1 in HK-2 cells induced by HG combined with TGF-β1, and the effect of SZP was weakened in the HK-2-Nrf2-KD group (Figures 6(d) and 6(e)). Above results suggest that the effect of SZP on improving EMT is inseparable from the activation of Nrf2. In addition, the regulation of the TGF-β1/Smad7 pathway by SZP is achieved in part by increasing the expression of Nrf2.

4. Discussion

Herbal extracts are currently used for the treatment of diabetic nephropathy. In our previous studies, it was found that Sanziguben granules are rich in polysaccharides [25]. In this study, SZP was obtained through hot water extraction and purified via the Sevage method. Although the polysaccharides were deproteinized in this study, the obtained polysaccharides were still crude, which possessed a lot of uncertainties. Although there are limitations, this study and the data can also be valuable.

In our experiment, SZP lowered blood sugar and improved kidney damage and fibrosis in DN mice (Figure 1). Previously published reports demonstrated the hypoglycemic and renal protective effects of polysaccharides. Pin Gong et al. showed that SGP-1-1, a new type of polysaccharide from Siraitia grosvenorii, had hypoglycemic and antioxidant effects in vitro and could reduce the inflammatory response in DN mice by inhibiting the TLR4-NFκB signaling pathway [26]. The new polysaccharide of Moutan Cortex (MC-Pa) has been shown to have strong antioxidant activity in vitro and has a protective effect on AGEs-induced diabetic nephropathy in vivo [27]. MDG-1, a polysaccharide from O. japonicas, can lower blood sugar in diabetic KKay mice and reduce kidney fibrosis [28].

Based upon Masson and Sirius red staining (Figure 2), we detected EMT proteins in kidney tissue and found that SZP upregulated the expression of E-cadherin and ZO-1 and decreased the levels of α-SMA and vimentin in the kidneys of DN mice (Figure 3). Recent studies have shown that inflammation, oxidative stress, and EMT are closely related in diabetes. The increase in ROS caused by persistent hyperglycemia and AGE formation is believed to be the driving force behind diabetic kidney damage and structural changes [29]. In the body, elevated ROS levels can induce inflammation, including macrophage infiltration, induce recruitment of a large number of inflammatory cells, and enhance the production of inflammatory cytokines (IL-1, IL-6, TNF-α, and MCP-1). Inflammation can further increase the production of ROS. Under the influence of inflammatory cytokines, macrophages, T-cells, and renal tubular epithelial cells will produce profibrotic mediators, such as TGF-β, CTGF, which induce the EMT and promote tissue fibrosis [3032]. Therefore, based on many reports on the antioxidant function of polysaccharides and the intrinsic relationship between oxidative stress and EMT, we investigated the antioxidant activity of SZP. We found that SZP can regulate oxidative stress homeostasis in DN mice through the improvement of SOD, MDA, and CAT. The Nrf2 signaling pathway is the most important antioxidant signaling pathway. Past studies have found that some polysaccharides can activate Nrf2 in the body and exert antioxidant effects [3335]. Studies have also reported that the expressions of inflammatory mediators TNF-α, IL-1β, and MCP1 are inhibited in endothelial cells with high expression of Nrf2 [36]. There are also reports that drugs can activate the expression of Nrf2 in renal tubular cells and inhibit oxalate-induced EMT [37]. By detecting the expression of Nrf2 and its downstream related molecules NQO1, HO-1, we found that SZP increased DN mice Nrf2 expression in kidney tissue (Figure 4). Moreover, we found that the increase of TGF-β1 in DN mice was reduced by SZP, while Smad7, which was inhibited in the pathological environment of DN, was increased by SZP (Figure 5). Studies have shown that Smad7 plays a key role in the antifibrotic response to TGF-β by inhibiting Smad2/3 phosphorylation by competing for TGFβRI binding and inducing its degradation [38]. In addition, Smad7 can also degrade TGFβRI by recruiting the E3 ubiquitin ligase Smurf2 [39]. Evidence suggested that in renal fibrosis, a large number of fibroblasts are derived from tubular epithelial cells via EMT [40]. We used HG combined with TGF-β1 to simulate the environment of diabetic nephropathy to induce EMT in HK-2 cells. SZP treatment ameliorated the symptoms of HK-2 EMT induced by HG plus TGF-β1. SZP increased the expression of Smad7 but decreased the level of TGF-β1 (Figure 6). These evidences suggest that the effect of SZP to inhibit EMT is closely related to the activation of Nrf2 and the inhibition of TGF-β1.

In order to explore the role of Nrf2 in the improvement of diabetic nephropathy by SZP, Nrf2 knockdown HK-2 cells were treated in the same way. The results indicating that SZP inhibits EMT and increases Smad7 may be related to the Nrf2 signaling pathway. In recent pharmacological studies, some researchers have also found a possible interaction between the Nrf2 pathway and the TGF-β/Smads signaling pathway. Studies have reported that bergenin can improve the accumulation of ECM in the kidneys of diabetic mice and HG-treated glomerular mesangial cells by upregulating the expression of Nrf2 and inhibiting oxidative stress, thus providing renal protection [41]. Bixin can induce the activation of the Nrf2 signaling pathway and inhibit the accumulation of EMT and extracellular matrix in renal tubular cells induced by TGF-β [42]. It has been reported that TanIIA can inhibit EMT and TGF-β1/Smad signaling by activating Nrf2 and improve silica-induced pulmonary fibrosis. Knockdown of Nrf2 by siRNA partially blocks TanIIA’s signal on EMT and TGF-β1/Smad [43]. Min-KyunSong et al. showed that BARD can upregulate Nrf2/Smad7 and inhibit TGF-β/Smad signaling to exert anti-inflammatory and antifibrotic effects. And they proposed that SMURF1/SMAD7 is a new molecular target to explain the renoprotective effects of Nrf2 signaling [44]. Although it is unclear how it interacts in vivo, this result suggests that there is a crosstalk between the Nrf2 signaling pathway and the TGF-β/Smads pathway in our experiments. Our follow-up study will further reveal the interaction between them. In summary, the results of the present experiments demonstrated that SZP may regulate Nrf2 to influence the TGF-β1/Smad7 signaling pathway and improve EMT in HK-2 cells (Figure 7).

Details are in the caption following the image
The pictorial representation of the possible biomechanism of action of SZP in the treatment of DN.

5. Conclusion

In conclusion, our research shows that SZP is an important pharmacodynamic substance of Sanziguben Recipe in the treatment of diabetes nephropathy. SZP can reduce oxidative stress by activating Nrf2, improve EMT by upregulating Smad7, and treat fibrosis in diabetes nephropathy.

Conflicts of Interest

The authors declare that they have no conflicts of interest.

Authors’ Contributions

ZLC and SSB designed the study. JNZ, FW, CL, and XYL conducted the experiment. YY and WPX provided administrative and material support. JNZ and FW did sample analysis and data analysis. JNZ wrote the manuscript. SSB and CL revised the paper. All authors have read and approved the final manuscript. Jianing Zhang and Fan Wang are co-first authors and contributed equally to this work.

Acknowledgments

We thank Dr.Yang Yu for assistance with experimental design. This work was supported by the National Natural Science Foundation of China (Grant no. 81774192) and the Guangdong Provincial Administration of Traditional Chinese Medicine (Project no. 20201003).

    Data Availability

    The data that support the findings of this study are available from the corresponding author upon reasonable request.

      The full text of this article hosted at iucr.org is unavailable due to technical difficulties.