1. Introduction
Chronic kidney disease (CKD), a slowly progressive and irreversible kidney function failure, affected approximately 10% of global population[
1]. Ascending clinical evidence suggests that excessive sodium intake accelerate the progression of CKD and relevant hypertension[
2,
3]. Besides, high level of sodium had been proved to induce kidney cytokine expression as well as tissue regeneration and fibrosis by activating Wnt / β-catenin and TGF-β signaling[
4,
5,
6]. It follows that excessive sodium intake is one of the important risk factors for CKD progression. Besides advocating salt reduction, it is critical to explore remedy to reconcile the adverse impact of excessive sodium consumption on CKD.
Intestine has a rich and structurally diverse intestinal microbiome that engaged in multiple interactions affecting host health by influencing the intrinsic immunity and metabolism. Moreover, the composition and function of the gut microbiota is dynamic and easily affected by diet properties. By importing dietary element into the bond between the host and its microbiota, nutrition maintains homeostasis or leads to disease susceptibility[
7]. As one of the most essential dietary components, excessive salt intake has been extensively studied and proven to cause a wide range of diseases through its effects on gut microbiome[
8]. Previous studied have shown that consumption of high salt resulted in alteration of gut microbiota structure that may be pro-inflammatory to exacerbates colitis and hypertension[
9,
10]. High salt induced gut barrier disruption triggers renal function injury[
11]. These indicated that excessive salt intake could cause susceptibility of a wide range of diseases through gut microbiome. Moreover, depletion of gut microbiome or administering probiotics can ease the progression of many diseases[
11,
12]. Together, this provides us with ideas to reverse progression of high salt related CKD through regulation of gut microbiome.
Puerariae Lobatae Radix (PLR) is rich in nutrients including flavonoids, starch, saponins, and amino acids, and has always been a medicine and food homologous herb to relieve gastrointestinal and cardiovascular diseases[
13]. Multiple studies have indicated that PLR and its active ingredient might alleviate diabetes related kidney damage and nonalcoholic fatty liver disease through modulating gut microbiota[
14,
15]. In our previous study, we also confirmed PLR promoted gut microbiota homeostasis, increased the relative abundance of beneficial bacteria and protected the structural and functional integrity of the gut barrier, blood-brain barrier, and placental barrier, thus contributed to vascular anomalies related diseases including ischemic stroke and pre-eclampsia[
13,
16]. CKD is a disease closely related to host metabolic disorder and vascular dysfunction, bioflavonoids and other bioactive compound have been verified to relieve CKD associated biochemical abnormalities and kidney inflammation[
17], however, few studies have focused on whether PLR exerts a protective effect on it.
In this study, we constructed a high salt induced CKD murine model and investigated the renoprotective effects of PLR. We found that PLR alleviated high salt diet induced CKD by repairing intestinal epithelial barrier and down-regulating fibrotic signaling. In addition, this protective effect was exerted, at least partly by promoting homeostasis of gut microbiota.
2. Materials and Methods
2.1. Source and preparation of PLR decoction
Medicinal slices form of PLR were purchased from Guangzhou Weida Company (Guangzhou, China). Standard substance of
puerarin (purity ≥ 99.0%),
daidzin (purity ≥ 98.0%) and
daidzein (purity ≥ 98.0%) were purchased from Chengdu Must Bio-Technology Company (Chengdu, Sichuan, China). PLR decoction preparation was conducted as previously described[
13,
16]. In brief, take a certain quality of PLR and extract in boiling distilled water at a ratio of 1:10 (
w/v) for 1 hour, collect the filtrate and boiling the residue with distilled water (1:6,
w/v) for another 1 hour. Concentrate the above filtrates and quantify the content of
puerarin,
daidzin, and
daidzein by using ultra-performance liquid chromatography (specific method is listed in
Supplementary Table S1). Organic phase methanol was obtained from Tianjin Da Mao chemical reagents factory (Tianjin, China). The respectively concentrations of these three compounds were 10.9, 17.5, and 0.3 mg/mL.
2.2. Animals and experimental design
All animal experimental procedures were conducted in strict accordance with the National Institute of Health guidelines and were approved by the Institutional Animal Ethical Care Committee of Southern Medical University (Guangzhou, China), Reference number: NFYY-2021-0572. Specific pathogen free male C57BL/6 mice (6-8 week, weighing 20-24 g) were purchased from SPF Biotechnology Co., Ltd. (Beijing, China). All mice were housed under standard conditions of temperature and humidity with a 12h: 12h light-dark cycle and free access to food and water. After one week of acclimatization, high salt feeding mice were given drinking water containing 2% (
w/v) NaCl for 8 weeks as described in previous study[
11] (HS,
n = 6), while PLR intervention mice received
Pueraria decoction (containing 72 mg/kg
Puerarin) by oral gavage once a day along with 8 weeks high salt feeding (PLR,
n = 6). The dosage of PLR had been mentioned in previous study[
13]. Control mice receive normal drinking water during the experiment (CON,
n = 6). There were no differences in baseline covariates among groups. And the feces and urine were collected every week for further examination. Mice were sacrificed under anesthesia and blood, kidney, liver, spleen, colon and cecal contents samples were harvested in a sterile manner for further analysis.
For blood pressure measurement, systolic blood pressure (SBP) and mean blood pressure (MBP) were measured every week via non-invasive tail cuff method, using a BP-2010A instrument (Softron Biotechnology, Beijing, China). All measurements were operated between 8am to 12am. At least six continuous stable results of each mouse were obtained and calculated the average value as the final result.
2.3. Fecal microbiota transplantation (FMT) experiment
FMT experiment was performed based on the modified method previously described[
11]. In brief, feces from the donor mice (HS and PLR group mice) were collected in aseptic centrifugal tubes respectively and resuspended in PBS at 125 mg/mL, the mixtures were centrifuged at 1000×g for 1 min and the supernatants were collected and saved in separate 1.5 mL tubes for subsequently microbiota transplantation. Before FMT experiment, the acceptor male C57BL/6 mice (6-8 week) were given antibiotics (ABX) (vancomycin, 100 mg/kg; neomycin sulfate 200 mg/kg; metronidazole 200 mg/kg; and ampicillin 200 mg/kg) intragastrically once a day for 1 w to deplete the gut microbiota. All the mice then received drinking water containing 2% (
w/v) NaCl for 8 weeks as above described. An amount of 150 μL of the fecal microbiota solution was simultaneously orally gavaged to mice once a day in the first 2 weeks, every other day in the following 2 weeks and twice each week in the last 4 weeks. The mice that received feces from HS mice or PLR mice were referred to as HS-FMT group or PLR-FMT group (
n = 8 per group). Control mice receive normal drinking water during the experiment (CON,
n = 5). Blood pressures were measured as above described every week. All the mice had free access to food and water, and the feces and urine were collected before sacrifice. Mice were sacrificed under anesthesia and then blood, kidney, liver, spleen, colon and cecal contents samples were harvested in a sterile manner for further analysis.
2.4. 16S rDNA gene sequencing and microbe analysis
Feces were collected in sterilized 1.5 mL tubes and frozen at -80 ℃ before DNA extraction. Microbial DNA from fecal samples were extracted as previously described[
18,
19]. V3-V4 region of 16S rDNA was amplified by PCR using the below primers (341F, 5′-CCTACGGGNGGCWGCAG-3′ and 806R, 5′-GGACTACHVGGGTATCTAAT-3′). Amplicons were purified using the AxyPrep DNA Gel Extraction Kit (Axygen Biosciences, Union City, CA, U.S.). Purified amplicons were pooled in equimolar and paired end sequenced (PE250) on an Illumina platform according to the standard protocols. Alpha diversity were calculated in QIIME [
20](version 1.9.1). PCoA and NMDS of weighted unifrac distances were generated in R project Vegan package (version 2.5.3). Biomarker features in each group were screened by LEfSe software[
21] (version 1.0). The KEGG pathway analysis of the OTUs was inferred using Tax4Fun[
22] (version 1.0) and were generated using Omicsmart, a dynamic real-time interactive online platform for data analysis (
http://www.omicsmart.com). Pearson correlation coefficient between environmental factors and species was calculated in R project psych package (version 1.8.4) then generated using the Wekemo Bioincloud (
https://www.bioincloud.tech). The calculated
p value was gone through FDR correction, taking FDR ≤ 0.05 as a threshold.
2.5. RNA-seq analysis
Kidney samples were collected in sterilized 1.5 mL tubes and frozen at -80℃ before DNA extraction. Total RNA was extracted using the EastepTM Super Total RNA Extraction Kit (Promega Corporation, Madison, WI, USA) according to the manufacturer’s instructions. After total RNA was extracted, eukaryotic mRNA was enriched by Oligo(dT) beads. Then fragments were transcribed into cDNA by using NEBNext Ultra RNA Library Prep Kit for Illumina (NEB #7530, New England Biolabs, Ipswich, MA, USA). The ligation reaction was purified and PCR amplified. The resulting cDNA library was sequenced using Illumina Novaseq6000 by Gene Denovo Biotechnology Co. (Guangzhou, China). RNAs differential expression analysis was performed by DESeq2[
23] software between two different groups. The transcripts with the parameter of false discovery rate (FDR) below 0.05 and absolute fold change ≥ 2 were considered differentially expressed transcripts. Gene set enrichment analysis (GSEA) was performed using software GSEA[
24] and MSigDB[
24] to identify whether a set of genes in specific KEGG pathway shows significant differences in two groups.
2.6. Gene expression analysis
Total RNA was extracted from kidney and colon tissue using the Animal Total RNA Isolation Kit (Foregene Co., Ltd, Chengdu, China) according to the manufacturer’s instructions. A reverse transcript enzyme, HiScript III RT SuperMix for qPCR (+gDNA wiper) (Vazyme Biotech Co., Ltd, Nanjing, China) was applied to obtain cDNA. The real-time quantitative PCR reactions were performed on the LightCycler480 (Roche Diagnostics International, Rotkreuz, Switzerland) using a ChamQ SYBR qPCR Master Mix (Vazyme, Nanjing, China). Relative quantification of target genes were calculated using the 2
-△△CT method.
Gapdh was used as a control gene. All the target gene primer sequences are listed in
Supplementary Table S2.
2.7. Protein expression and biochemical analysis
The kidney and colon tissue protein were extracted with a commercial RIPA lysis buffer (Epizyme Biomedical Technology Co., Ltd, Shanghai, China) adding 1% cocktail protease inhibitor (Meilunbio, Dalian, China). Western blot was performed with the primary antibodies for Occludin, ZO-1 (Abcam, Cambridge, MA, USA), Claudin-1, TNF-α, IL-1β (ImmunoWay, Suzhou, China), β-catenin (Proteintech, Wuhan, China) and β-actin (Beijing Ray Antibody Biotech, Beijing, China). Serum creatinine was determined by using a creatinine (Cr) Assay kit (sarcosine oxidase) (Nanjing Jiancheng Bioengineering institution Co., Ltd, Nanjing, China). The urinary protein, serum LPS and serum TNF-α level were detected manually with commercial ELISA Kit (MEIMIAN, Yancheng, China). Serum and urine Na+ concentrations were determined on an automatic biomedical analyzer (Roche Diagnostics International, Rotkreuz, Switzerland).
2.8. FD-4 permeability experiment
FITC Dextran 4-KD (FD-4, Sigma, Shanghai, China) was used to detect intestinal permeability in vivo. FD-4 was orally administered to mice at a dose of 0.6 mg/kg four hours before serum was harvested. The FD-4 level in serum was detected keep in the dark, based on a standard curve and spectrofluorometrically with an excitation wavelength of 485 nm and an emission wavelength of 530 nm in a microplate fluorescence reader (Infinite® M1000 PRO, TECAN, Switzerland).
2.9. Imaging of intestinal inflammation in vivo
L-012 (FUJIFILM Wako Pure Chemical corporation, Tokyo, Japan) was used to confirm intestinal inflammation
in vivo [
25]. The mice were anesthetized in an anesthesia chamber with 1.5–2.0% isoflurane–mixed gas and injected with 100 μL of a 20-mmol L-012 solution. 1 min later, IVIS Spectrum CT system was used to acquire bioluminescent images. For postacquisition analysis, the Living Image software was used to quantify the bioluminescent signals at standardized regions of interest (ROIs) defined on the abdomen.
2.10. Histological staining and analysis
The left kidney and colon tissue of mice were collected and fixed in 4% paraformaldehyde. The samples were then embedded in paraffin, sectioned at 4 μm thick and stained with hematoxylin and eosin (HE) or Masson’s trichrome. The kidney tissue histological damage was accessed according to previously described scoring method[
26]. The kidney fibrosis area was selected randomly from at least 5 points of cortical fields and quantified using image J software. Immunohistochemistry was performed using the primary antibodies for Occludin, ZO-1 (Abcam, Cambridge, MA, USA), Claudin-1 (ImmunoWay, Suzhou, China). Quantification of the average optical density was performed by automated image analysis in five randomly chosen × 200 fields of each sample. All images were scanned by a NanoZoomer Digital slide scanner and captured at × 200 or × 400 with an NDP. View2 Plus Image viewing software (Hamamatsu Photonics, Hamamatsu, Japan).
2.11. Immunofluorescence
Colon paraffin section samples were generated as above described, blocked for 1 h, and incubated at 4 °C with anti-β-catenin antibody (Proteintech, Wuhan, China) overnight. Then the slices were washed and stained with DAPI for 10 min. Images were captured with fluorescent microscopy (Olympus, Japan). Quantification of the fluorescence intensity was calculated by automated image analysis in five randomly chosen × 200 fields of each sample.
2.12. Statistical analysis
Results were represented as means ± standard error of the mean (SEM) and performed using GraphPad Prism software (version 8.0) (GraphPad Software, San Diego, Canada). Statistical analyses were performed using one-way analysis of variance (ANOVA) or repeated measurement ANOVA as indicated in the figure legends. A p-value < 0.05 was considered statistically significant.
4. Discussion
It is well known that excessive salt intake is associated with hypertension, which closely links cardiovascular disease and CKD. Therefore, how to relieve the potential risk about kidney injury brought by high salt has arisen the attention[
30,
31]. Previous review advocated non-pharmacological strategies such as dietary and lifestyle modulation as well as kidney disease-specific pharmacological interventions to achieve kidney function preservation[
32]. PLR is rich in nutrients including flavonoids, starch, saponins, and amino acids, and has always been a medicine and food homologous herb to relieve gastrointestinal and cardiovascular diseases[
13]. PLR is consistent with the principles of the CKD management guidelines that advocate dietary control. However, whether PLR can have effect on CKD remains unclear. In the present study, for the first time, we found that oral PLR administration could alleviate CKD and the protective effect was achieved, at least partly by remodeling gut microbiota. Besides, PLR intervention significantly alleviated the renal fibrosis and down-regulated the canonical Wnt / β-catenin signaling pathway in kidney.
Salt is one of the most common dietary elements and plays an important role in maintaining the water-salt balance of the body. However excessive salt intake has been shown to associate with the development of cardiovascular diseases and other metabolic disease. In our study, prolonged excessive high salt diet led to gut microbiota disruption, intestinal inflammation, and permeability increasement. We found that high salt diet induced both inflammatory and fibrotic damage to the kidneys, probably because of harmful bacterial metabolites, like LPS, and inflammatory factors, including TNF-α, originating from the gut entering the circulation and inducing damage to distant organs. However, the therapeutic intervention of PLR could protect against both intestinal and renal injury, which we hypothesized may be a result of its ability to reverse disturbances in gut microbiota. We also found that PLR intervention increased
Akkermansia,
Lactobacillus and
Bifidobacterium and decreased
Rikenella,
Prevotellaceae_UCG-001 and
Lachnoclostridium. In recent years, many clinical studies have found that gut microbiota can mediate associations between the gut and other organs, such as “gut-brain axis”, “gut-kidney axis”, etc. This suggests that gut microbiota may be a potential target for intervention in abenteric disease progression. Indeed, we confirmed that PLR exerted efficacy at least partially through gut microbiota by FMT experiment. PLR greatly increased the relative abundance of beneficial bacteria such as
Akkermansia.
Akkermansia muciniphila (
A. muciniphila) is considered one of the key players in colonic mucus-associated microbiota and is necessary for gut to produce mucus to maintain a healthy mucus layer and thickness of the intestinal wall[
33,
34]. Previous study has demonstrated that increased colonization of
A. muciniphila in the colon of mice mitigated gut barrier leakage and blood endotoxemia in experimental colitis[
35]. Furthermore, previous clinical research showed the relative abundance of
Akkermansia in the CKD patients was significantly lower than that in healthy group[
36]. In addition,
Akkermansia may be the driving factor to influence butanoate and tryptophan metabolism to alleviate renal fibrosis in CsA-induced chronic nephrotoxicity mice model[
37]. Further, oral gavage of mice with
A. muciniphila protected against HFD/CCl
4-induced liver and kidney fibrosis by modulating inflammatory response[
38]. Moreover,
A. muciniphila administration suppressed epithelial-mesenchymal transition and reduced renal interstitial fibrosis in 5/6 nephrectomy rats[
39]. This suggested that besides repairing the intestinal barrier,
Akkermansia was also closely related to alleviating the damage of CKD and renal fibrosis. Similarly, in our study, our results also revealed that the relative abundances of
Akkermansia were negatively correlated with nephritic histopathological fibrotic degree and biochemical indexes of kidney injury. Therefore, we hypothesized that the alleviating effect of PLR on CKD and renal fibrosis may be associated with increased colonization of
Akkermansia in the intestine.
The gut microbiota is a complex ecosystem in which various strains of bacteria can interact with each other through resource competition and nutrient symbiosis[
13], besides the protective effect of a single bacterium, symbiotic relationship between beneficial bacteria in intestinal homeostasis maintenance and disease protection should not be overlooked as well[
40]. In our study, we found that PLR also increased the relative abundance of
Bifidobacterium and
Lactobacillus, which have already been used as industrial production probiotics[
41], are capable of enhancing the intestinal mucus layer and goblet cell function thus protecting the integrity of the intestinal barrier[
42,
43]. Similarly, in our previous study, we found that PLR induced concomitant increases in
Akkermansia and
Bifidobacterium in the treatment of ischemic stroke[
16], which led us to hypothesize that both
Akkermansia and
Bifidobacterium were biomarkers for the efficacy of the protective efficacy of PLR in CKD. In vitro experiments demonstrated that
A. muciniphila can stimulate the growth and of change the gene expression profile of
Lactobacillus[
44,
45]. Studies have also shown that
Bifidobacterium and
Lactobacillus have potential benefit to reduce levels of uraemic toxins and protect against CKD[
46,
47,
48]. PLR is rich in macromolecules such as starch, cellulose, and lignin[
40,
49]. These macromolecules reached the colon and provided energy for the synergistic growth of carbohydrate-utilizing bacteria such as
Bifidobacterium and
Lactobacillus[
13,
50,
51]. In addition, above mentioned probiotics, together with
Faecalibaculum, which could be enriched by PLR intervention in our study, have been reported as short-chain fatty acids-producing bacteria[
52,
53,
54], which could increase SCFAs production and thus create an anti-inflammatory and anti-oxidative environment in intestinal. Moreover, such non-inflammatory stable state reduced gut barrier damage and permeation of harmful metabolites into plasma, thus decreased the adverse impact on abenteric organs. Together, the gut microbiota molded by PLR, characterized by
Akkermansia,
Bifidobacterium and
Lactobacillus, is a key factor in its enteroprotective and nephroprotective effects.
Continuous intake of high salt needed to be excreted in urine to maintain the balance of water-salt in the plasma, which might be a sustained stimulation towards kidney in urine production that caused pathological damage and fibrosis of the kidneys. The Wnt signaling pathway is more accepted to influence high salt-related CKD at present[
55]. Previous literature indicated that transient activation of Wnt / β-catenin facilitates kidney tissue generation after acute kidney injury, whereas sustained activation stimulates kidney fibrosis in CKD[
56]. With the presence of high salt load, mice presented with more fibrosis and upregulated of the Wnt / β-catenin signal in heart and kidney[
57,
58]. In the present study, the intervention of PLR significantly alleviated the degree of renal fibrosis in high salt induced CKD. More importantly, gene set enrichment analysis showed the downregulation of canonical Wnt signaling pathway in PLR group. This might explain a potential pathway by which the PLR protected kidney from fibrosis. In our study, we inspected the Tax4Fun prediction analysis from the KEGG database and found that the Wnt signaling pathway was correlated with gut microbial metabolism. Besides, our results showed that the expression of the Wnt / β-catenin pathway as well as the level of pathological damage in kidney were in significantly negative correlation with the relative abundance of beneficial bacteria increased by PLR intervention, such as
Akkermansia,
Lactobacillus and
Bifidobacterium, which led us to hypothesize that the gut microbiota may be effective in regulating the expression of the Wnt signaling pathway and subsequently slow down the process of renal fibrosis. Many studies have found that the Wnt signaling pathway can be regulated by gut microbiota. Treatment of mice with
A. muciniphila is reported to significantly suppress EMT and reduced renal interstitial fibrosis[
39]. Furthermore, treatment of
Bifidobacterium bifidum and
Lactobacillus gasseri with quercetin could exert inhibition of the canonical Wnt / β-catenin signaling pathway to protect against colorectal cancer in mice[
59]. Both PLR and PLR-FMT intervention could significantly downregulated the Wnt signaling pathway. From this we speculated that alterations in the Wnt signaling pathway and alleviation of renal fibrosis by PLR intervention was associated with gut microbiota and their metabolism. On the other hand, PLR is rich in flavonoids, such as puerarin and daidzin. Study showed that combined use of vitamin D and
Puerarin protected against hepatic fibrosis probably via silencing the Wnt1 / β-catenin pathway[
60]. This suggested that in addition to manipulating the gut microbiota, the nephroprotection of PLR may be partially attributed to the antifibrotic effects of its small molecule active ingredients.
Inflammation and other signaling pathways can also contribute to CKD. In our study, significantly elevated protein level of TNF-α was found in serum and kidney of CKD mice. Increased TNF-α is one of the upstream targets for triggering β-catenin activation in kidney[
61,
62], the reduction of β-catenin has been shown to result in a lower expressions of fibrosis markers including fibronectin, Col1a1 and Col3a1[
63,
64,
65,
66]. However, the elevation of TNF-α and β-catenin were significantly suppressed by the intervention of PLR in the present study. This added to the possible targets for PLR exerting its nephroprotective effects. In addition, VEGF signaling pathway was another pathway that could be downregulated by PLR intervention in our study. Previous studies have shown that many therapies targeted at HIF-1α/VEGF signaling pathway to relieve liver fibrosis[
67,
68]. VEGF signaling pathway was also reported to have synergistic effect with Wnt / β-catenin signaling pathway in angiogenesis and fibrosis[
69,
70]. In our study, they may have corporate effects on renal fibrosis after PLR intervention.
PLR, as a traditional Chinese medicine and food homologous herb, has multi-target and multi-pathway pharmacodynamic routes of action. In the present study, we confirmed that PLR could alleviate CKD by remodeling the gut microbiota, repairing intestinal epithelial barrier, and downregulating the Wnt signaling pathway in the kidney, we identified for the first time that PLR relieved high salt diet-induced CKD by modulating the gut-kidney axis. However, our study definitely has some limitations; this study focused on gut microbiota entirety and their beneficial effect on CKD, and single strains of bacteria were not accessed. In addition, we focused on Wnt1, Wnt3 and Wnt4 in the canonical Wnt / β-catenin signaling pathway, this did not mean the other Wnt gene have no influence[
71]. Previous studies have revealed that upregulation of Wnt4 and Wnt5 gene could activate the noncanonical Wnt pathway in hepatic stellate cells of fibrotic livers[
72]. Additionally, Wnt6 is involved in epithelialization and loss of Wnt6 expression contributes to tubular injury and fibrosis in animal models[
73]. Also, it was certainly that the beneficial pharmaceutical effects of PLR must mediate through more than one mechanism, the RNA-seq results also showed a differential KEGG enrichment in HIF-1 signaling pathway, Ferroptosis and MAPK signaling pathway, the potential role of which cannot be completely ruled out is a major limitation of this study. In the future, more deep understanding about the remodeling ability of PLR towards gut microbiota should be further explored. Overall, our study provides evidence for new function of PLR on kidney protection and a novel direction for the treatment of kidney disease.
Figure 1.
PLR alleviated kidney injuries induced by high salt diet and down-regulated the Wnt / β-catenin pathway in kidney of HS mice. (A) SBP change tendency of groups were evaluated noninvasively in the course of experiment. (B) Serum creatinine after 8 weeks. (C) Urine protein after 8 weeks. (D) Representative gross anatomy pictures of the kidney. (E) Kidney coefficient. (F) HE and MASSON staining of the kidney and the pathology scores and histological analysis. HE staining scale bar = 100 μm, MASSON staining scale bar = 50 μm. (G) RT-qPCR analysis of kidney injuries related genes. (H) Urine Na+ concentration. (I) Serum Na+ concentration. (J) RT-qPCR analysis of inflammatory factors genes in kidney. (K, L) GSEA performed on the RNA seq data (n = 3). (M) RT-qPCR analysis of Wnt1, Wnt3, Wnt4 and β-catenin genes in kidney (n = 5-6). (N) β-catenin and TNF-α protein levels in the kidney (n = 4). (O) Representative in situ detection of β-catenin in renal cortex was measured by immunofluorescent staining. Kidney tissue sections were stained with DAPI (blue) and probed with β-catenin (red). Scale bar = 50 μm (n = 4). Results are expressed as the mean ± SEM. n = 6 for each group. * p<0.05, ** p<0.01 and *** p<0.001, versus the CON group. # p<0.05, ## p<0.01 and ### p<0.001, versus the HS group. & p<0.05, && p<0.01 and &&& p<0.001, versus the PLR-L group. $ p<0.05, $$ p<0.01 and $$$ p<0.001, versus the PLR-M group in A-E. ***p<0.001, **p<0.01, *p<0.05 in G-J, M-O. PLR, Puerariae Lobatae Radix; HS, high salt; SBP, systolic blood pressure; HE, hematoxylin-eosin; RT-qPCR, real-time quantitative PCR, GSEA, gene-set enrichment analyses.
Figure 1.
PLR alleviated kidney injuries induced by high salt diet and down-regulated the Wnt / β-catenin pathway in kidney of HS mice. (A) SBP change tendency of groups were evaluated noninvasively in the course of experiment. (B) Serum creatinine after 8 weeks. (C) Urine protein after 8 weeks. (D) Representative gross anatomy pictures of the kidney. (E) Kidney coefficient. (F) HE and MASSON staining of the kidney and the pathology scores and histological analysis. HE staining scale bar = 100 μm, MASSON staining scale bar = 50 μm. (G) RT-qPCR analysis of kidney injuries related genes. (H) Urine Na+ concentration. (I) Serum Na+ concentration. (J) RT-qPCR analysis of inflammatory factors genes in kidney. (K, L) GSEA performed on the RNA seq data (n = 3). (M) RT-qPCR analysis of Wnt1, Wnt3, Wnt4 and β-catenin genes in kidney (n = 5-6). (N) β-catenin and TNF-α protein levels in the kidney (n = 4). (O) Representative in situ detection of β-catenin in renal cortex was measured by immunofluorescent staining. Kidney tissue sections were stained with DAPI (blue) and probed with β-catenin (red). Scale bar = 50 μm (n = 4). Results are expressed as the mean ± SEM. n = 6 for each group. * p<0.05, ** p<0.01 and *** p<0.001, versus the CON group. # p<0.05, ## p<0.01 and ### p<0.001, versus the HS group. & p<0.05, && p<0.01 and &&& p<0.001, versus the PLR-L group. $ p<0.05, $$ p<0.01 and $$$ p<0.001, versus the PLR-M group in A-E. ***p<0.001, **p<0.01, *p<0.05 in G-J, M-O. PLR, Puerariae Lobatae Radix; HS, high salt; SBP, systolic blood pressure; HE, hematoxylin-eosin; RT-qPCR, real-time quantitative PCR, GSEA, gene-set enrichment analyses.
Figure 2.
PLR reduced intestinal inflammation and protected against intestinal barrier damage in HS mice. (A) Representative gross anatomy pictures of the colon and the colon length measurement. (B) RT-qPCR analysis of inflammatory factors genes in colon. (C) Representative L-012 fluorescent staining and animal fluorescence imaging (n = 5-6). (D, E) Relative serum TNF-α and LPS level. (F) FD-4 level in the plasma. (G, H) HE staining and ZO-1, Occludin and Claudin-1 immunohistochemical staining in colon tissues. Scale bar = 100 μm (n = 3-6). (I) RT-qPCR analysis of tight junction proteins genes in colon (n = 5-6). (J) ZO-1 and Occludin protein levels in the colon (n = 3). Results are expressed as the mean ± SEM. n = 6 for each group. ***p<0.001, **p<0.01, *p<0.05 were determined by One-way ANOVA. LPS, lipopolysaccharide; FD-4, FITC-dextran 4-KD; HE, hematoxylin-eosin.
Figure 2.
PLR reduced intestinal inflammation and protected against intestinal barrier damage in HS mice. (A) Representative gross anatomy pictures of the colon and the colon length measurement. (B) RT-qPCR analysis of inflammatory factors genes in colon. (C) Representative L-012 fluorescent staining and animal fluorescence imaging (n = 5-6). (D, E) Relative serum TNF-α and LPS level. (F) FD-4 level in the plasma. (G, H) HE staining and ZO-1, Occludin and Claudin-1 immunohistochemical staining in colon tissues. Scale bar = 100 μm (n = 3-6). (I) RT-qPCR analysis of tight junction proteins genes in colon (n = 5-6). (J) ZO-1 and Occludin protein levels in the colon (n = 3). Results are expressed as the mean ± SEM. n = 6 for each group. ***p<0.001, **p<0.01, *p<0.05 were determined by One-way ANOVA. LPS, lipopolysaccharide; FD-4, FITC-dextran 4-KD; HE, hematoxylin-eosin.
Figure 3.
PLR reversed intestinal microbial dysbiosis in HS mice and remodeled gut microbiota by increase the relative abundance of probiotics. (A) Relative bacterial abundance at the genus level in the feces of mice. (B) Histogram of the LDA score showing the biomarker at the genus level of each group. (C) Relative abundance of indicator species at the genus level showing the enriched bacteria in the gut microbiome among groups. (D) Microbial Dysbiosis index of each group. (E, F) PCoA and NMDS based on the weighted UniFrac analysis of operational taxonomic units (OTUs). (G) Correlation heatmap of major indicator species and biomarkers based on LDA score and major injury indicators. (H) KEGG pathway analysis of function distribution and difference analysis based on Tax4Fun prediction results. Results are expressed as the mean ± SEM. n = 6-10 for each group. ***p<0.001, **p<0.01, *p<0.05 were determined by One-way ANOVA or Kruskal–Wallis test in C, D, adonis analysis and anosim analysis in E, Spearman analysis in G, Tukey HSD test in H. LDA, linear discriminant analysis; PCoA, principal coordinates analysis; NMDS, nonmetric multidimensional scaling; OTUs, operational taxonomic units; KEGG, kyoto encyclopedia of genes and genomes.
Figure 3.
PLR reversed intestinal microbial dysbiosis in HS mice and remodeled gut microbiota by increase the relative abundance of probiotics. (A) Relative bacterial abundance at the genus level in the feces of mice. (B) Histogram of the LDA score showing the biomarker at the genus level of each group. (C) Relative abundance of indicator species at the genus level showing the enriched bacteria in the gut microbiome among groups. (D) Microbial Dysbiosis index of each group. (E, F) PCoA and NMDS based on the weighted UniFrac analysis of operational taxonomic units (OTUs). (G) Correlation heatmap of major indicator species and biomarkers based on LDA score and major injury indicators. (H) KEGG pathway analysis of function distribution and difference analysis based on Tax4Fun prediction results. Results are expressed as the mean ± SEM. n = 6-10 for each group. ***p<0.001, **p<0.01, *p<0.05 were determined by One-way ANOVA or Kruskal–Wallis test in C, D, adonis analysis and anosim analysis in E, Spearman analysis in G, Tukey HSD test in H. LDA, linear discriminant analysis; PCoA, principal coordinates analysis; NMDS, nonmetric multidimensional scaling; OTUs, operational taxonomic units; KEGG, kyoto encyclopedia of genes and genomes.
Figure 4.
Gut microbiota from mice treated with PLR improved kidney tissue damage induced by high salt diet and down-regulated the Wnt / β-catenin pathway. (A) Flow chart of FMT experimental design. 8-week-old male C57BL/6 mice were given drinking water containing 2% w/v NaCl for 8 weeks. (B) SBP change tendency was evaluated noninvasively during experiment. aa indicates p<0.01 and aaa indicates p<0.001 for the CON group versus the HS-FMT group on the corresponding week. bb indicates p<0.01 and bbb indicates p<0.001 for the PLR-FMT group versus the HS-FMT group on the corresponding week. cc indicates p<0.01 and ccc indicates p<0.001 for the PLR-FMT group versus the CON group on the corresponding week. Repeated measurement ANOVA was used for statistical analysis. (C) Serum creatinine after 8 weeks. (D) Urine protein after 8 weeks. (E) Representative gross anatomy pictures of the kidney. (F) Kidney coefficient. (G) HE and MASSON staining of the kidney and the pathology scores and histological analysis. HE staining scale bar = 100 μm, MASSON staining scale bar = 50 μm (n = 4-8). (H) RT-qPCR analysis of kidney injuries related genes. (I) RT-qPCR analysis of inflammatory factors genes in kidney. (J) RT-qPCR analysis of Wnt1, Wnt3, Wnt4 and β-catenin genes in kidney (n = 5-8). (K-M) β-catenin and TNF-α protein levels in the kidney. Results are expressed as the mean ± SEM. n = 5-8 for each group. ***p<0.001, **p<0.01, *p<0.05 were determined by One-way ANOVA in C, D, F-I. PLR, Puerariae Lobatae Radix; HS, high salt; FMT, fecal microbiota transplantation; ABX, antibiotic; SBP, systolic blood pressure; HE, hematoxylin-eosin; RT-qPCR, real-time quantitative PCR.
Figure 4.
Gut microbiota from mice treated with PLR improved kidney tissue damage induced by high salt diet and down-regulated the Wnt / β-catenin pathway. (A) Flow chart of FMT experimental design. 8-week-old male C57BL/6 mice were given drinking water containing 2% w/v NaCl for 8 weeks. (B) SBP change tendency was evaluated noninvasively during experiment. aa indicates p<0.01 and aaa indicates p<0.001 for the CON group versus the HS-FMT group on the corresponding week. bb indicates p<0.01 and bbb indicates p<0.001 for the PLR-FMT group versus the HS-FMT group on the corresponding week. cc indicates p<0.01 and ccc indicates p<0.001 for the PLR-FMT group versus the CON group on the corresponding week. Repeated measurement ANOVA was used for statistical analysis. (C) Serum creatinine after 8 weeks. (D) Urine protein after 8 weeks. (E) Representative gross anatomy pictures of the kidney. (F) Kidney coefficient. (G) HE and MASSON staining of the kidney and the pathology scores and histological analysis. HE staining scale bar = 100 μm, MASSON staining scale bar = 50 μm (n = 4-8). (H) RT-qPCR analysis of kidney injuries related genes. (I) RT-qPCR analysis of inflammatory factors genes in kidney. (J) RT-qPCR analysis of Wnt1, Wnt3, Wnt4 and β-catenin genes in kidney (n = 5-8). (K-M) β-catenin and TNF-α protein levels in the kidney. Results are expressed as the mean ± SEM. n = 5-8 for each group. ***p<0.001, **p<0.01, *p<0.05 were determined by One-way ANOVA in C, D, F-I. PLR, Puerariae Lobatae Radix; HS, high salt; FMT, fecal microbiota transplantation; ABX, antibiotic; SBP, systolic blood pressure; HE, hematoxylin-eosin; RT-qPCR, real-time quantitative PCR.
Figure 5.
FMT from mice treated with PLR reduced intestinal inflammation and protected intestinal barrier function. (A) Representative gross anatomy pictures of the colon and the colon length measurement. (B) RT-qPCR analysis of inflammatory factors genes in colon. (C) Representative L-012 fluorescent staining and animal fluorescence imaging (n = 5-6). (D, E) Relative serum TNF-α and LPS level. (F) FD-4 level in the plasma. (G, H) HE staining and ZO-1, Occludin and Claudin-1 immunohistochemical staining in colon tissues. Scale bar = 100 μm (n = 3-6). (I) RT-qPCR analysis of tight junction proteins genes in colon (n = 5-6). (J) ZO-1, Occludin and Claudin-1 protein levels in the colon (n = 4). Results are expressed as the mean ± SEM. n = 4-8 for each group. ***p<0.001, **p<0.01, *p<0.05 were determined by One-way ANOVA. LPS, lipopolysaccharide; FD-4, FITC-dextran 4-KD; HE, hematoxylin-eosin.
Figure 5.
FMT from mice treated with PLR reduced intestinal inflammation and protected intestinal barrier function. (A) Representative gross anatomy pictures of the colon and the colon length measurement. (B) RT-qPCR analysis of inflammatory factors genes in colon. (C) Representative L-012 fluorescent staining and animal fluorescence imaging (n = 5-6). (D, E) Relative serum TNF-α and LPS level. (F) FD-4 level in the plasma. (G, H) HE staining and ZO-1, Occludin and Claudin-1 immunohistochemical staining in colon tissues. Scale bar = 100 μm (n = 3-6). (I) RT-qPCR analysis of tight junction proteins genes in colon (n = 5-6). (J) ZO-1, Occludin and Claudin-1 protein levels in the colon (n = 4). Results are expressed as the mean ± SEM. n = 4-8 for each group. ***p<0.001, **p<0.01, *p<0.05 were determined by One-way ANOVA. LPS, lipopolysaccharide; FD-4, FITC-dextran 4-KD; HE, hematoxylin-eosin.
Figure 6.
FMT from mice treated with PLR relieved intestinal microbial dysbiosis and rebuilt healthy microbiota environment. (A) Relative bacterial abundance at the genus level in the feces of mice. (B) Histogram of the LDA score showing the biomarker at the genus level between the HS-FMT group and the PLR-FMT group. (C) Relative abundance of indicator species at the genus level showing the enriched bacteria in the gut microbiome among groups. (D) Microbial Dysbiosis index of each group. (E, F) PCoA and NMDS based on the weighted UniFrac analysis of operational taxonomic units (OTUs). (G) Correlation heatmap of major indicator species and biomarkers based on LDA score and major injury indicators. (H) KEGG pathway analysis of function distribution and difference analysis based on Tax4Fun prediction results. Results are expressed as the mean ± SEM. n = 5-8 for each group. ***p<0.001, **p<0.01, *p<0.05 were determined by One-way ANOVA or Kruskal–Wallis test in C, D, adonis analysis and anosim analysis in E, Spearman analysis in G, Tukey HSD test in H. LDA, linear discriminant analysis; PCoA, principal coordinates analysis; NMDS, nonmetric multidimensional scaling; OTUs, operational taxonomic units; KEGG, kyoto encyclopedia of genes and genomes.
Figure 6.
FMT from mice treated with PLR relieved intestinal microbial dysbiosis and rebuilt healthy microbiota environment. (A) Relative bacterial abundance at the genus level in the feces of mice. (B) Histogram of the LDA score showing the biomarker at the genus level between the HS-FMT group and the PLR-FMT group. (C) Relative abundance of indicator species at the genus level showing the enriched bacteria in the gut microbiome among groups. (D) Microbial Dysbiosis index of each group. (E, F) PCoA and NMDS based on the weighted UniFrac analysis of operational taxonomic units (OTUs). (G) Correlation heatmap of major indicator species and biomarkers based on LDA score and major injury indicators. (H) KEGG pathway analysis of function distribution and difference analysis based on Tax4Fun prediction results. Results are expressed as the mean ± SEM. n = 5-8 for each group. ***p<0.001, **p<0.01, *p<0.05 were determined by One-way ANOVA or Kruskal–Wallis test in C, D, adonis analysis and anosim analysis in E, Spearman analysis in G, Tukey HSD test in H. LDA, linear discriminant analysis; PCoA, principal coordinates analysis; NMDS, nonmetric multidimensional scaling; OTUs, operational taxonomic units; KEGG, kyoto encyclopedia of genes and genomes.