Skip to main content

16S rRNA sequencing-based evaluation of the protective effects of Hua-Zhuo-Jie-Du on rats with chronic atrophic gastritis

Abstract

Background

Disturbance of the intestinal flora is a pathogenic factor for chronic atrophic gastritis (CAG). Hua-Zhuo-Jie-Du (HZJD) has been shown to be an effective Chinese herbal preparation for treating CAG. However, the effects of HZJD on the intestinal flora of CAG is unclear. In this study, we probed the regulating effects of HZJD on intestinal microbes in CAG rats using 16S rRNA gene sequencing.

Methods

High-performance liquid chromatography (HPLC) analysis was used to perform quality control of HZJD preparations. We then administered 1-methyl-3-nitro-1-nitrosoguanidine (200 μg/ml) to Sprague–Dawley rats to establish a CAG model. HZJD and vitacoenzyme were administered orally to these rats over a 10 week period. Hematoxylin and eosin (H&E) staining was performed to observe the histopathology of CAG rats. A rarefaction curve, species accumulation curve, Chao1 index, and ACE index were calculated to assess the alpha diversity. Principal component analysis (PCA), non-metric multi-dimensional scaling (NMDS), and unweighted pair group method with arithmetic mean (UPGMA) were conducted to examine the beta diversity. The LEfSe method was used to identify differential bacteria. Differential function analysis used PCA based on KEGG function prediction.

Results

HPLC showed that our HZJD preparation method was feasible. H&E staining showed that HZJD significantly improved the pathological state of the gastric mucosa in CAG rats. The rarefaction curve and species accumulation curve showed that the sequencing data were reasonable. The Chao1 and ACE indices were significantly increased in CAG rats compared to the N group. Following HZJD and vitacoenzyme treatment, the Chao1 and ACE indices were decreased. PCA, NMDS, and UPGMA results showed that the M group was separated from the N, HZJD, and V groups, and LEfSe results showed that the relative abundance of Akkermansia, Oscillospira, Prevotella, and CF231 were significantly higher in the N group. Proteobacteria and Escherichia were significantly enriched in the M group, Allobaculum, Bacteroides, Jeotgalicoccus, Corynebacterium, and Sporosarcina were significantly enriched in the V group, and Firmicutes, Lactobacillus, and Turicibacter were significantly enriched in the HZJD group.

Conclusion

HZJD exhibited a therapeutic effect on the intestinal flora of CAG rats.

Peer Review reports

Background

Gastric carcinoma (GC) is a serious global health problem. With the prevalence of GC rising, this disease is currently the fifth most common cancer and the third leading cause of cancer death worldwide [1, 2]. Chronic atrophic gastritis (CAG) is defined as the occurrence of crucial transitional lesions from the normal gastric mucosa to GC. Its pathological features include decreased gastric acid secretion, thinning of the gastric mucosa, and decrease in proper gland morphology, with or without intestinal metaplasia or gastric epithelial dysplasia. CAG's main clinical manifestations include upper abdominal pain, bloating, and loss of appetite, which is occasionally accompanied by weight loss, diarrhea, or secondary anemia. CAG has become an important area of research given that suppression or reversion of CAG may serve to lower the incidence of GC [3, 4]. Emerging studies have demonstrated that the gut microbiome contributes to the regulation of gastrointestinal function and participates in various physiological and pathological processes, including inflammation and cell proliferation. Changes in intestinal flora promote the progression from CAG to GC [5,6,7].

Currently, therapies such as folic acid and vitamin B (12), endoscopic mucosal resection, and anti-Hp treatment, are used in CAG treatment [8,9,10,11]. However, these treatments are often limited in clinical application and may have adverse effects. In China, many herbs based on traditional Chinese medicine (TCM) have been utilized to protect the gastric mucosa and reverse CAG [12, 13]. Hua-Zhuo-Jie-Du (HZJD) is a Chinese formula that is widely used for treating CAG that is based on the TCM theories of detoxicating and resolving dampness. HZJD consists of 11 herbs, including Artemisia capillaris Thunb. (Yinchen), Scutellaria baicalensis Georgi (Huangqin), Coptis chinensis Franch. (Huanglian), Scutellaria barbata D. Don (Banzhilian), Scleromitrion diffusum (Willd.) R. J. Wang (Baihuasheshecao), Isatis tinctoria L. (Banlangen), Pogostemon cablin (Blanco) Benth. (Guanghuoxiang), Lobelia chinensis Four. (Banbianlian), Sophora flavescens Aiton (Kushen), Eupatorium fortunei Turcz. (Peilan), and Gynostemma pentaphyllum (Thunb.) Makino (Jiaogulan). In previous research, through network pharmacology and experimental verification, the important components and possible targets of HZJD in the treatment of CAG were obtained [14]. Previous clinical experiments have also confirmed that HZJD had positive clinical effects [15]. In a recent study, it was shown that HZJD could inhibit the proliferation of precancerous gastric lesions and promote cell apoptos is by down-regulating lncRNA-517368 [16].

The effect of HZJD, however, on the intestinal flora in patients with CAG remains unclear. Therefore, in the current study, we sought to investigate whether HZJD inhibited the development of CAG by regulating the intestinal flora of CAG model rats.

Materials and methods

Experimental procedure

An experimental flowchart for isolation of HZJD components is shown in Fig. 1.

Fig. 1
figure 1

Experimental flow chart

Drugs and reagents

1-Methyl-3-nitro-1-nitrosoguanidine (MNNG) was purchased from Tokyo Chemical Industry, Japan (Lot. NH8JH-BM). Vitacoenzyme tablets were purchased from LEPUHENGJIUYUAN Co., Ltd., China (No. 20190803). Sodium salicylate was purchased from Sangon Biotech, China (Lot. EC11BA0022). The following formula granules were provided by Guangdong Yifang Pharmaceutical Co., Ltd.: Artemisia capillaris Thunb. (Yinchen), 15 g; Scutellaria baicalensis Georgi (Huangqin), 12 g; Coptis chinensis Franch.(Huanglian), 12 g; Scutellaria barbata D. Don (Banzhilian), 15 g; Scleromitrion diffusum (Willd.) R. J. Wang (Baihuasheshecao), 15 g; Isatis tinctoria L.(Banlangen), 15 g; Pogostemon cablin (Blanco) Benth.(Guanghuoxiang), 9 g; Lobelia chinensis Lour. (Banbianlian), 15 g; Sophora flavescens Aiton (Kushen), 10 g; Eupatorium fortunei Turcz.(Peilan), 9 g; and Gynostemma pentaphyllum (Thunb.) Makino (Jiaogulan), 15 g.

High-performance liquid chromatography (HPLC) analysis of HZJD

The quality and chemical constituents of our HZJD preparation were examined by HPLC. The condition optimization of the chemical fingerprint was as follows. An Agilent Eclipse XDB-C18 (5 μm, 4.6 × 250 mm), with methanol (A) and 0.1% phosphoric acid (B) as the mobile phase was used under full wavelength detection. Gradient elution was used for these solvents. For HPLC analysis, a 10 μl sample was injected into the column, with a column temperature of 35 °C. The detection was performed at 330 nm.

Animals

The animal care and experimental procedures performed in this study were approved by the Institutional Animal Care and Use Committee of the Hebei College of Traditional Chinese Medicine (DWLL2019012). Seventy male Sprague–Dawley (SD) rats (body weight 150 – 180 g) were supplied by Liaoning Changsheng Biotechnology Co., Ltd. The rats were raised in a temperature controlled environment (24 °C ± 4 °C) under a 12/12 h light–dark cycle, with free access to water and food. Following a week of adaptation, the rats were randomly divided into a normal group (N, n = 8) and a CAG model group (n = 62). The N group had free access to clean water and normal diet. This CAG rat model was generated by administration of MNNG (200 μg/ml) combined with irregular fasting (1 day fasting and 1 day feeding), and 2% sodium salicylate was intragastrically administered once per fasting day on an empty stomach, as described previously [12, 13, 17].

Thereafter, at the 12th, 16th, 20th, and 24th weeks, two rats from the model group were selected randomly and euthanized by intraperitoneal injection of 2% sodium pentobarbital (140 mg/kg). The stomach was removed for pathological examination to confirm successful establishment of a CAG model. Finally, the rats in the CAG model group were randomly divided into a model group (M, n = 8), a vitacoenzyme group (V, n = 8), and a HZJD group (HZJD, n = 8). The rats in the HZJD and V groups were treated with a daily dose of 14.81 g/kg/d HZJD and 0.08 g/kg/d vitacoenzyme. All animals were treated for 10 weeks, consecutively.

Sample collection

Fresh feces were collected before pathological examination. Following fasting but were allowed to drink water for 24 h, the SD rats were euthanized with 2% sodium pentobarbital (140 mg/kg i.p.) and stomach samples were collected immediately. The gastric tissues and feces were stored at − 80 °C until future use.

Pathological examination

Stomach samples were collected and then immersed in paraformaldehyde. Following tissue dehydration, the samples were embedded in paraffin and subsequently cut into 4-μm-thick sections. Hematoxylin–eosin (H&E) staining was performed for histological evaluation by light microscopy.

Extraction of genomic DNA

The CTAB/SDS method was used to extract genomic DNA from gastric mucosal tissues. The DNA concentration and purity were monitored on a 1% agarose gel. The DNA was then diluted to 1 ng/μl with sterile water.

Amplicon generation

Primers 16S V3-V4: 341F (5′-CCTAYGGGRBGCASCAG-3′) and 806R (5′-GGACTACNNGGGTATCTAAT-3′) were used for amplicon generation. The 16S rRNA gene was amplified using specific primers with barcodes. All PCR reactions were performed in a 30 μL reaction volume, with 15 μL Phusion® high-fidelity PCR Master Mix (New England Biolabs), 0.2 μM forward and reverse primers, and approximately 10 ng of template DNA. The thermal cycle included an initial denaturation at 98 °C for 1 min, followed by a cycle of denaturation at 98 °C for 30 s, annealing at 50 °C for 30 s, and extension at 72 °C for 60 s. Finally at 72 °C for 5 min.

Quantification and qualification of PCR products

An equal volume of 1X loading buffer (containing SYB green) was mixed with PCR products, and electrophoresis on a 2% agarose gel was performed for detection. We selected samples with bright main bars between 400 and 450 bp for further experiments.

Mixing and purification of PCR products

PCR products were mixed at an equivalent ratio before purifying with an AxyPrepDNA Gel Extraction Kit (AXYGEN).

Library preparation and sequencing

Following the manufacturer’s recommendations, a sequencing library was generated using the NEB Next® Ultra™ DNA Library Preparation Kit for Illumina (NEB, USA), and an index sequence was added. The library quality was evaluated using a Qubit@2.0 Fluorometer (Thermo Scientific) and an Agilent Bioanalyzer 2100 system. Finally, the library was sequenced on the Illumina MiSeq/HiSeq 2500 platform, and 250 bp/300 bp paired end reads were generated.

Data analysis

The raw data obtained by sequencing were spliced, filtered, and de-chimerized to obtain clean data to improve the accuracy and reliability of our results. We next divided the filtered sequences with ≥ 97% similarity into the same operational taxonomic unit (OTU). A rarefaction curve, species accumulation curve, Chao1 index, and ACE index were calculated to assess the alpha diversity of the species present in each sample and the depth of sequencing between groups. Based on this taxonomic information, a statistical analysis of community structure was conducted at each taxonomic level (phylum, class, order, family, genus, and species). Beta diversity was used to investigate the similarity of the bacterial community structures among the groups. Principal component analysis (PCA) and non-metric multi-dimensional scaling analysis (NMDS) were used to show beta diversity. The LEfSe uses linear discriminant analysis (LDA) to screen for differential flora. Finally, differential function analysis used PCA based on KEGG function prediction.

Results

HPLC profile for HZJD

Our previous clinical trials and animal experiments revealed that HZJD had a good inhibitory effect on CAG. We also examined the main components of HZJD granules to ensure the quality of their preparation. Fig. 2 shows the HPLC chromatograms of an HZJD reference sample (A) and of a test sample (B). The digital identification of the chromatographic peak on the picture is its retention time. From left to right, the index components are scutellarin, berberine, palmatine, baicalin, wogonin, and baicalein. The results showed that our preparation method for HZJD was both stable and feasible, which provides a reference for its quality control.

Fig. 2
figure 2

High-performance liquid chromatography (HPLC) chromatogram of a HZJD reference sample (A) and a test sample (B). From left to right, the index components are scutellarin, berberine, palmatine, baicalin, wogonin, and baicalein

Morphological changes of the gastric mucosa

Figure 3 shows the histopathological changes of the gastric mucosa. H&E staining showed that in the N group rats, the gastric mucosal epithelial cells and the glands had preserved integrity and were arranged regularly. No expansion or hyperemia was observed. Additionally, no inflammatory cellular infiltration was observed in the submucosa and muscularis. In contrast, H&E staining showed that in the M group rats, the gastric glands were reduced and disordered, and the gastric mucosa layer was significantly reduced, which was accompanied by inflammatory cellular infiltration. These results suggested that a CAG rat model had been successfully established. Compared to the M group, H&E staining of V group rats showed that the arrangements of gastric glands were irregular, the structures of gastric glands were lacking, and the base layer of the mucosa was thicker, with no obvious improvement. However, the gastric mucosa morphology was significantly improved in the HZJD group compared to that in the M group. H&E staining indicated that the glands were neatly arranged and the complete glandular tube structure was clearly observable. No abnormal thickening of the muscularis mucosa was noted. Based on these pathological manifestations, HZJD was considered to be an effective treatment for CAG.

Fig. 3
figure 3

Representative images of the gastric mucosa of rats in different groups. (H&E × 100)

Sequencing data preprocessing and quality control

A total of 2,881,791 raw sequences were obtained after 16S rRNA sequencing of the four groups of rats. After splicing, filtering, and de-chimerizing the raw data, 2,030,883 clean data sequences were obtained, with each sample containing 56,821 to 68,687 sequences (S1).

Venn diagram

A Venn diagram was generated to define the core microbiome detected in most samples at the species level. We identified 2,975, 3,299, 2,441, and 2,148 species in the N, M, HZJD, and V groups, respectively (Fig. 4). Among these, 1,093 species were uniform and the other species were not shared in all groups; 444 species were detected in the N group only, whereas 811 species were detected in the M group only. Similarly, 332 species were found in the HZJD group only, and 159 species were found in the V group only.

Fig. 4
figure 4

Venn diagram. Different colors represent different groups. The overlapping portions represent the common taxa between groups, and the non-overlapping portions represent unique taxa in each group

Alpha diversity

A fixed amount of sequencing data from each sample was extracted randomly and used to construct a rarefaction curve with the corresponding number of species. A rarefaction curve reflects the rationality of the amount of sequencing data. When the curve tends to be flat, it indicates that the amount of sequencing data is reasonable. As shown in Fig. 5A, the rarefaction curve gradually became flat, indicating that the sequencing data were reasonable in this study. A species accumulation curve can be used to fully determine whether a sample size is sufficient. A sharp rise in the position of the box chart indicates that the sample size is insufficient and the sample size needs to be increased, while the opposite indicates that the sample is sufficient and data analysis can be conducted. As shown in Fig. 5B, as the sample size increased, the box plot tended to be flat, indicating that the sample size was sufficient. We also calculated the Chao1 and ACE indices to assess the microbial diversity. Gut microbiota from the CAG rats demonstrated that the Chao1 and ACE indices were significantly increased compared to the N group. Following HZJD and vitacoenzyme treatment, the Chao1 and ACE indices were significantly decreased compared to the M group (Fig. 5C and D).

Fig. 5
figure 5

Alpha diversity indices from different groups. A Rarefaction curve. B Species accumulation curve. C Chao1 result. D ACE result. Different colors represent different groups

Beta diversity

PCA can be used to show the similarity or difference in community composition between samples. Samples with high community structure similarity cluster together, while samples with large community structure differences become separated. The PCA score revealed that the community composition of the M group was far from that of the HZJD and V groups (Fig. 6A). The samples of the N, HZJD, and V groups were distributed on the right side of the t1 axis, which explained 13% of the total variations, indicating an influence of the HZJD constituents on the gut microbial composition. NMDS analysis is based on the species information contained in the sample in the form of points reflected in the multi-dimensional space, and the degree of difference between different samples is reflected by the distance between points, which can reflect the differences between groups and within groups of samples. The NMDS results also showed a separation between the HZJD and M groups (Fig. 6B). Moreover, to compare the similarity between different samples, the unweighted pair group method with arithmetic mean (UPGMA) was used. The results demonstrated that the majority of the gut microbes were clustered in their own groups, which was consistent with our PCA and NMDS results (Fig. 6C).

Fig. 6
figure 6

Beta diversity. A PCA score plot. B NMDS result. C UPGMA result

Analysis of horizontal community structures across groups

The top 10 abundant species among the 4 groups of rats were selected based on the results of species annotations, and a columnar cumulative chart of species relative abundance was generated to visually display the species with higher relative abundance and their proportions. At the phylum level, the top 10 abundant species were Firmicutes, Bacteroidetes, Proteobacteria, Actinobacteria, Verrucomicrobia, Tenericutes, Spirochetes, Cyanobacteria, Elusimicrobia, and Acidobacteria (Fig. 7A). At the genus level, the top 10 abundant species were Lactobacillus, Oscillospira, Ruminococcus, Allobaculum, Prevotella, Escherichia, [Prevotella], Akkermansia, Desulfovibrio, and Bacteroides (Fig. 7E). The top 10 species of the other levels are shown in Fig. 7B, C, D and F.

Fig. 7
figure 7

The top 10 abundant species at different levels A Phylum. B Class. C Order. D Family. E Genus. F Species

LEfSe analysis

We used the LEfSe method to identify the specific bacterial phylotypes that were differentially altered between the N, M, HZJD, and V groups. A cladogram was obtained from the LEfSe method (LDA > 3.0), and the results showed that the abundances of the genera Akkermansia, Oscillospira, Prevotella, and CF231 were significantly higher in the N group. The phylum Proteobacteria and the genus Escherichia were significantly enriched in the M group (Fig. 8A and B). Following HZJD and vitacoenzyme treatment, the abundance of the phylum Proteobacteria and the genus Escherichia were decreased, and the therapeutic effect of HZJD was better than that of vitacoenzyme (Fig. 8C). The phylum firmicutes and the genera Lactobacillus and Turicibacter were significantly enriched in the HZJD group, and the genera Allobaculum, Bacteroides, Jeotgalicoccus, Corynebacterium, and Sporosarcina were significantly enriched in the V group (Fig. 8A and B).

Fig. 8
figure 8

The potential biomarkers defined by LEfSe. A Cladogram. Each circle represents the classified level from phylum to species. The diameter of the small circle is proportional to the relative abundance of gut microbiota. B Histogram. The threshold on the logarithmic LDA score for discriminative features was set to 3.0. C Abundance of Escherichia and Proteobacteria in each group

PCA KEGG function prediction

KEGG function prediction analysis is an effective method to study the changes in metabolic function of community samples to adapt to environmental changes. PCA was thus used to explore the similarity or difference of the functions of different groups at an overall level. Fig. 9 shows that the samples of the N, HZJD, and V groups clustered together, the M group was separated from the N, HZJD, and V groups.

Fig. 9
figure 9

PCA KEGG function prediction result

Discussion

GC comprises the following precancerous steps: chronic active gastritis → chronic atrophic gastritis → gastric intestinal metaplasia → gastric epithelial dysplasia [18]. Inflammation, cell proliferation and apoptosis, autophagy dysfunction, energy metabolism disorders, and other factors may contribute to the development process, although the exact pathogenesis is still unclear [14, 17, 19, 20]. The common therapies used for CAG focus on symptomatic treatment and are often not completely effective. Strategies involving TCM are, however, becoming increasingly accepted. In TCM, turbid toxicity is an important cause of CAG, leading to disharmony between the spleen and the stomach and increases endogenous dampness and heat. The use of HZJD is based on turbid toxic syndrome. It functions to clear away heat, and detoxify and invigorate the spleen to remove dampness and has achieved good results in the treatment of CAG [14, 19]. However, the regulating effect of HZJD on the intestinal flora is still unclear. In this study, we used HPLC to control the quality of the HZJD decoction studied, H&E staining to evaluate the therapeutic effect of HZJD on CAG pathology, and 16S rRNA sequencing to evaluate the regulating effect of HZJD on the intestinal flora of CAG rats.

There is little doubt that the intestinal flora plays an important role in the development and progression of gastric diseases. Disturbances of the intestinal microbiome can induce inflammation, abnormal cell proliferation and apoptosis, and oxidative stress, which all may be associated with the development of CAG [21, 22]. A recent study has shown that there are significant differences in the microbiome between patients with GC, intestinal metaplasia, and chronic gastritis [23,24,25]. Thus, understanding what constitutes a health-promoting (eubiotic) or disease-promoting (dysbiotic) microbial community has become the focus of recent research [26].

The Venn diagram showed that 1,093 species were uniform between the four groups. These species may play an important role in the stability and function of a CAG rat intestinal microecological environment. However, 811 species were only found in the CAG model group, and these species may be related to the occurrence of CAG and are worthy of further exploration.

A rarefaction curve and species accumulation curve were generated and demonstrated the reliability of the sequencing data in this study. The Chao1 and ACE indices were significantly increased in the M group, indicating that the intestinal flora of CAG rats had a higher diversity. Prior reports have suggested that changes in microbial composition and diversity are essential to promote inflammation, proliferation, and tumor progression [27]. Moreover, our previous research demonstrated that abnormal cell proliferation and apoptosis mechanisms play an important role in the malignant transformation of the gastric mucosa [14, 28]. Therefore, we speculated that changes in the diversity of the intestinal flora in CAG may be an important factor for the occurrence of CAG. Following HZJD treatment, the Chao1 and ACE indices were significantly decreased, with no statistical significance compared to the N group, indicating that HZJD could effectively regulate the diversity of the CAG intestinal flora. PCA, NMDS and UPGMA results indicated that the intestinal flora microbial composition in CAG rats had changed, and the HZJD group exhibited a therapeutic effect on the community structure of the intestinal flora in CAG rats.

Akkermansia and Oscillospira are normal bacteria in the intestine and are crucial in maintaining the integrity and anti-inflammatory state of the gastrointestinal tract, while Prevotella is a dominant bacterial genus within the gut [29, 30]. The abundance of these bacteria were significantly enriched in the N group, which may be contribute to gastrointestinal health. The presence of Proteobacteria is a microbial symbol of intestinal flora imbalance, and an increase in Proteobacteria can cause inflammation of the gastrointestinal tract and destroy intestinal homeostasis [31]. Escherichia is one of the most common causes of several bacterial infections in humans and animals, is the prominent cause of gastroenteritis, and can induce apoptosis in gastric cells [32, 33]. In this study, the abundance of Escherichia and Proteobacteria increased significantly in the M group, which may be a cause of gastrointestinal inflammation and gastric mucosal atrophy. Following HZJD treatment, the abundance of the phylum Proteobacteria and the genus Escherichia decreased, indicating that HZJD could alleviate gastrointestinal inflammation by regulating the abundance of Proteobacteria and Escherichia. Firmicutes interact with endogenous beneficial microbes to restore intestinal homeostasis and are beneficial to a host’s intestinal environment [34]. Lactobacillus has been characterized as a type of probiotic in the digestive tract, and can maintain the microecological balance of the gastrointestinal tract. Moreover, dynamic monitoring of Lactobacillus levels has important clinical significance for predicting the development of GC [6]. Lactobacillus can also regulate the immune system, reducing the risk of allergies and cancer, and can reduce free radical oxidizing substances [35]. Studies have found that Lactobacillus can reduce the adhesion of Helicobacter pylori (Hp), inhibit Hp infection, and alleviate Hp-related gastritis [36,37,38]. Turicibacter is an important member of the gut microbiota and is considered to be a healthy bacterial genera with anti-inflammatory effects [39,40,41]. CAG is a process of inflammatory cancer conversion, in which continuous inflammatory stimulation promotes the growth of GC cells and increases the chance of gene mutation. Imbalance in the intestinal flora can lead to the production of inflammatory factors and can cause gastric mucosal damage [42, 43]. In this study, the abundance of Firmicutes, Lactobacillus, and Turicibacter was significantly increased in the HZJD group, indicating that HZJD can increase the abundance of healthy bacterial genera, improving the anti-inflammatory ability of the gastrointestinal tract, and therefore protecting against the development of CAG. The genera Allobaculum, Bacteroides, Jeotgalicoccus, Corynebacterium, and Sporosarcina were significantly enriched in the V group. Allobaculum can produce short-chain fatty acids (SCFAs), and bacteria that produce SCFAs have previously been shown to protect mucous membranes from damage from pathogens by providing colonic cell nutrition and reducing inflammation, which are beneficial to the host [44]. However, conditional pathogenic bacteria, such as Bacteroides and Sporosarcina, were also found to be present in the V group.

Based on the KEGG database, PICRUSt software was used for function prediction. PCA result suggested that the M group had significant differences in the functions of the intestinal flora, while the N, HZJD, and V groups had similar enrichments of functions, which also proved the regulating effect of HZJD on the intestinal flora.

Conclusion

In conclusion, these results indicated that HZJD exhibited a therapeutic effect on the diversity, community structure, and relative abundance of the intestinal flora in CAG rats, which may represent one of the mechanisms underlying the treatment effects of HZJD in CAG.

Availability of data and materials

The datasets used and analyzed in the current study are available from the corresponding author upon reasonable request. All data generated or analyzed in this study are included in this article.

Abbreviations

CAG:

Chronic atrophic gastritis

GC:

Gastric carcinoma

TCM:

Traditional Chinese medicine

HZJD:

Hua-Zhuo-Jie-Du

MNNG:

1-Methyl-3-nitro-1-nitrosoguanidine

V:

Vitacoenzyme

M:

Model

N:

Normal

SD:

Sprague–Dawley

HPLC:

High-performance liquid chromatography

H&E:

Hematoxylin–Eosin

KEGG:

Kyoto encyclopedia of genes and genomes

PCA:

Principal component analysis

LEfSe:

LDA effect size

UPGMA:

Unweighted pair group method with arithmetic mean

References

  1. Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D, Bray F. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015;136(5):E359-386.

    Article  CAS  PubMed  Google Scholar 

  2. Luo G, Zhang Y, Guo P, Wang L, Huang Y, Li K. Global patterns and trends in stomach cancer incidence: age, period and birth cohort analysis. Int J Cancer. 2017;141(7):1333–44.

    Article  CAS  PubMed  Google Scholar 

  3. Zeng J, Guo J, Gong D, Zhang Y, You F, Liang C, Pan H, Cai T, Chen X, Chen L, et al. Weipixiao ameliorates gastric precancerous lesions in a rat’s model by regulating GSK3¦Â and C-myc. J Tradit Chin Med. 2018;38(5):705–13.

    Article  PubMed  Google Scholar 

  4. Liu W, Pan HF, Wang Q, Zhao ZM. The application of transgenic and gene knockout mice in the study of gastric precancerous lesions. Pathol Res Pract. 2018;214(12):1929–39.

    Article  CAS  PubMed  Google Scholar 

  5. Pimentel-Nunes P, Barros A, Pita I, Miranda I, Conceição G, Borges-Canha M, Leite-Moreira AF, Libânio D, Dinis-Ribeiro M. Gastric microbiome profile throughout gastric carcinogenesis: beyond helicobacter. Scand J Gastroenterol. 2021;56(6):708–16.

  6. Yu C, Su Z, Li Y, Li Y, Liu K, Chu F, Liu T, Chen R, Ding X. Dysbiosis of gut microbiota is associated with gastric carcinogenesis in rats. Biomed Pharmacother. 2020;126:110036.

    Article  CAS  PubMed  Google Scholar 

  7. Ndegwa N, Ploner A, Andersson AF, Zagai U, Andreasson A, Vieth M, Talley NJ, Agreus L, Ye W. Gastric microbiota in a low-helicobacter pylori prevalence general population and their associations with gastric lesions. Clin Transl Gastroenterol. 2020;11(7):e00191.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Li Z, Wu C, Li L, Wang Z, Xie H, He X, Feng J. Effect of long-term proton pump inhibitor administration on gastric mucosal atrophy: a meta-analysis. Saudi J Gastroenterol. 2017;23(4):222–8.

    PubMed  PubMed Central  Google Scholar 

  9. Lewerin C, Jacobsson S, Lindstedt G, Nilsson-Ehle H. Serum biomarkers for atrophic gastritis and antibodies against Helicobacter pylori in the elderly: Implications for vitamin B12, folic acid and iron status and response to oral vitamin therapy. Scand J Gastroenterol. 2008;43(9):1050–6.

    Article  CAS  PubMed  Google Scholar 

  10. Ohkusa T, Fujiki K, Takashimizu I, Kumagai J, Tanizawa T, Eishi Y, Yokoyama T, Watanabe M. Improvement in atrophic gastritis and intestinal metaplasia in patients in whom Helicobacter pylori was eradicated. Ann Intern Med. 2001;134(5):380–6.

    Article  CAS  PubMed  Google Scholar 

  11. Rakic S, Pesko P, Milicevic M, Gerzic Z. Atrophic chronic gastritis and esophagogastric anastomotic leak after resection and reconstruction for esophageal carcinoma. J Surg Oncol. 1992;51(1):19–21.

    Article  CAS  PubMed  Google Scholar 

  12. Cai T, Zhang C, Zeng X, Zhao Z, Yan Y, Yu X, Wu L, Lin L, Pan H. Protective effects of Weipixiao decoction against MNNG-induced gastric precancerous lesions in rats. Biomed Pharmacother. 2019;120:109427.

    Article  CAS  PubMed  Google Scholar 

  13. Li S, Huang M, Chen Q, Li S, Wang X, Lin J, Zhong G, Lin P, Asakawa T. Confirming the effects of qinghuayin against chronic atrophic gastritis and a preliminary observation of the involved inflammatory signaling pathways: an in vivo study. Evid Based Complement Alternat Med. 2018;2018:4905089.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Hao X, Liu Y, Zhou P, Jiang Q, Yang Z, Xu M, Liu S, Zhang S, Wang Y. Integrating network pharmacology and experimental validation to investigate the mechanisms of huazhuojiedu decoction to treat chronic atrophic gastritis. Evid Based Complement Alternat Med. 2020;2020:2638362.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Lyu JJ, Wang YG, Lyu XY. Effect of Huazhuo Jiedu formula on HIF-1α, VEGF, and PTEN in chronic erosive gastritis patients with syndrome of accumulation of turbidity and toxicity. Zhongguo Zhong Yao Za Zhi. 2018;43(11):2384–90.

    PubMed  Google Scholar 

  16. Hao X, Zhou P, Yang Z, Yang T, Wang Y. The therapeutic effect of Huazhuojiedu decoction on precancerous lesions in a gastric cancer model via the regulation of lnc 517368. J Ethnopharmacol. 2021;283:114635.

    Article  PubMed  Google Scholar 

  17. Zeng J, Yan R, Pan H, You F, Cai T, Liu W, Zheng C, Zhao Z, Gong D, Chen L, et al. Weipixiao attenuate early angiogenesis in rats with gastric precancerous lesions. BMC Complement Altern Med. 2018;18(1):250.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Correa P. A human model of gastric carcinogenesis. Cancer Res. 1988;48(13):3554–60.

    CAS  PubMed  Google Scholar 

  19. Zhou P, Hao X, Liu Y, Yang Z, Xu M, Liu S, Zhang S, Yang T, Wang X, Wang Y. Determination of the protective effects of Hua-Zhuo-Jie-Du in chronic atrophic gastritis by regulating intestinal microbiota and metabolites: combination of liquid chromatograph mass spectrometer metabolic profiling and 16S rRNA gene sequencing. Chin Med. 2021;16(1):37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Wang T, Liu K, Wen L, Yang Y, Yin X, Liu K, Chen Y, He Y, Yang M, Wei Y, et al. Autophagy and gastrointestinal diseases. Adv Exp Med Biol. 2020;1207:529–56.

    Article  CAS  PubMed  Google Scholar 

  21. Greer JB, O’Keefe SJ. Microbial induction of immunity, inflammation, and cancer. Front Physiol. 2011;1:168.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Vipperla K, O’Keefe SJ. Diet, microbiota, and dysbiosis: a “recipe” for colorectal cancer. Food Funct. 2016;7(4):1731–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Zhang S, Shi D, Li M, Li Y, Wang X, Li W. The relationship between gastric microbiota and gastric disease. Scand J Gastroenterol. 2019;54(4):391–6.

    Article  PubMed  Google Scholar 

  24. Zhang X, Li C, Cao W, Zhang Z. Alterations of gastric microbiota in gastric cancer and precancerous stages. Front Cell Infect Microbiol. 2021;11:559148.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Wang Z, Gao X, Zeng R, Wu Q, Sun H, Wu W, Zhang X, Sun G, Yan B, Wu L, et al. Changes of the gastric mucosal microbiome associated with histological stages of gastric carcinogenesis. Front Microbiol. 2020;11:997.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Gorvitovskaia A, Holmes SP, Huse SM. Interpreting prevotella and bacteroides as biomarkers of diet and lifestyle. Microbiome. 2016;4:15.

    Article  PubMed  PubMed Central  Google Scholar 

  27. O’Keefe SJ. Diet, microorganisms and their metabolites, and colon cancer. Nat Rev Gastroenterol Hepatol. 2016;13(12):691–706.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Xu J, Zheng X, Cheng KK, Chang X, Shen G, Liu M, Wang Y, Shen J, Zhang Y, He Q, et al. NMR-based metabolomics reveals alterations of electro-acupuncture stimulations on chronic atrophic gastritis rats. Sci Rep. 2017;7:45580.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Macchione IG, Lopetuso LR, Ianiro G, Napoli M, Gibiino G, Rizzatti G, Petito V, Gasbarrini A, Scaldaferri F. Akkermansia muciniphila: key player in metabolic and gastrointestinal disorders. Eur Rev Med Pharmacol Sci. 2019;23(18):8075–83.

    CAS  PubMed  Google Scholar 

  30. Gálvez EJC, Iljazovic A, Amend L, Lesker TR, Renault T, Thiemann S, Hao L, Roy U, Gronow A, Charpentier E, et al. Distinct polysaccharide utilization determines interspecies competition between intestinal prevotella spp. Cell Host Microbe. 2020;28(6):838-852.e836.

    Article  Google Scholar 

  31. Shin NR, Whon TW, Bae JW. Proteobacteria: microbial signature of dysbiosis in gut microbiota. Trends Biotechnol. 2015;33(9):496–503.

    Article  CAS  PubMed  Google Scholar 

  32. Durkin E, Moran AP, Hanson PJ. Apoptosis induction in gastric mucous cells in vitro: lesser potency of Helicobacter pylori than Escherichia coli lipopolysaccharide, but positive interaction with ibuprofen. J Endotoxin Res. 2006;12(1):47–56.

    Article  CAS  PubMed  Google Scholar 

  33. Allocati N, Masulli M, Alexeyev MF, Di Ilio C. Escherichia coli in Europe: an overview. Int J Environ Res Public Health. 2013;10(12):6235–54.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Kleerebezem M, Vaughan EE. Probiotic and gut lactobacilli and bifidobacteria: molecular approaches to study diversity and activity. Annu Rev Microbiol. 2009;63:269–90.

    Article  CAS  PubMed  Google Scholar 

  35. Slattery C, Cotter PD, O’Toole PW. Analysis of health benefits conferred by lactobacillus species from kefir. Nutrients. 2019;11(6):1252.

    Article  CAS  PubMed Central  Google Scholar 

  36. Sakarya S, Gunay N. Saccharomyces boulardii expresses neuraminidase activity selective for α2,3-linked sialic acid that decreases Helicobacter pylori adhesion to host cells. APMIS. 2014;122(10):941–50.

    Article  CAS  PubMed  Google Scholar 

  37. Yang YJ, Sheu BS. Probiotics-containing yogurts suppress Helicobacter pylori load and modify immune response and intestinal microbiota in the Helicobacter pylori-infected children. Helicobacter. 2012;17(4):297–304.

    Article  CAS  PubMed  Google Scholar 

  38. Francavilla R, Lionetti E, Castellaneta SP, Magistà AM, Maurogiovanni G, Bucci N, De Canio A, Indrio F, Cavallo L, Ierardi E, et al. Inhibition of Helicobacter pylori infection in humans by Lactobacillus reuteri ATCC 55730 and effect on eradication therapy: a pilot study. Helicobacter. 2008;13(2):127–34.

    Article  PubMed  Google Scholar 

  39. Terzo S, Mulè F, Caldara GF, Baldassano S, Puleio R, Vitale M, Cassata G, Ferrantelli V, Amato A. Pistachio consumption alleviates inflammation and improves gut microbiota composition in mice fed a high-fat diet. Int J Mol Sci. 2020;21(1):365.

    Article  CAS  PubMed Central  Google Scholar 

  40. Cuív P, Klaassens ES, Durkin AS, Harkins DM, Foster L, McCorrison J, Torralba M, Nelson KE, Morrison M. Draft genome sequence of Turicibacter sanguinis PC909, isolated from human feces. J Bacteriol. 2011;193(5):1288–9.

    Article  PubMed  Google Scholar 

  41. Auchtung TA, Holder ME, Gesell JR, Ajami NJ, Duarte RT, Itoh K, Caspi RR, Petrosino JF, Horai R, Zárate-Bladés CR. Complete genome sequence of Turicibacter sp. strain H121, isolated from the feces of a contaminated germ-free mouse. Genome Announc. 2016;4(2):e00114-16.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Park YH, Kim N. Review of atrophic gastritis and intestinal metaplasia as a premalignant lesion of gastric cancer. J Cancer Prev. 2015;20(1):25–40.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Oliphant K, Allen-Vercoe E. Macronutrient metabolism by the human gut microbiome: major fermentation by-products and their impact on host health. Microbiome. 2019;7(1):91.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Zhang X, Zhao Y, Xu J, Xue Z, Zhang M, Pang X, Zhang X, Zhao L. Modulation of gut microbiota by berberine and metformin during the treatment of high-fat diet-induced obesity in rats. Sci Rep. 2015;5:14405.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We would like to thank Shanghai Applied Protein Technology Co. Ltd for technical assistance with 16S rRNA sequencing and analysis. We thank LetPub (www.letpub.com) for its linguistic assistance during the preparation of this manuscript.

Funding

The current study was supported by the Natural Science Foundation of Hebei (No. H2020423207), the Hebei University of Chinese Medicine Science and Technology Ability Improvement Project (KTZ2019030), and the Postgraduate Innovation Funding Project of Hebei University of Chinese Medicine (XCXZZBS2021012).

Author information

Authors and Affiliations

Authors

Contributions

PPZ designed the study and contributed to the initial data analysis. PPZ and MCX performed the experiments. TXY, YJZ, and PPS provided helpful advice for the preparation of the manuscript. YGW critically reviewed and revised the manuscript and approved the final version for submission. All authors met the ICMJE criteria for authorship. The author(s) read and approved the final manuscript.

Corresponding author

Correspondence to Yangang Wang.

Ethics declarations

Ethics approval and consent to participate

Animal care and experimental procedures performed in this study were approved by the Institutional Animal Care and Use Committee of the Hebei University of Chinese Medicine (DWLL2019012). We confirm that all methods were performed in accordance with relevant guidelines and regulations. We also confirm that the study was carried out in compliance with ARRIVE guidelines.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhou, P., Yang, T., Xu, M. et al. 16S rRNA sequencing-based evaluation of the protective effects of Hua-Zhuo-Jie-Du on rats with chronic atrophic gastritis. BMC Complement Med Ther 22, 71 (2022). https://doi.org/10.1186/s12906-022-03542-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12906-022-03542-z

Keywords