Next Article in Journal
Density and Home Range of Cats in a Small Inhabited Mediterranean Island
Previous Article in Journal
Perioperative Analgesic and Sedative Effects of Cannabidiol in Cats Undergoing Ovariohysterectomy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Clostridium butyricum Prevents Diarrhea Incidence in Weaned Piglets Induced by Escherichia coli K88 through Rectal Bacteria–Host Metabolic Cross-Talk

1
College of Life Science, Yulin University, Yulin 719000, China
2
College of Animal Science and Technology, Shihezi University, Shihezi 832003, China
3
College of Animal Science and Technology, Northwest A&F University, Xianyang 712100, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Animals 2024, 14(16), 2287; https://doi.org/10.3390/ani14162287
Submission received: 4 June 2024 / Revised: 15 July 2024 / Accepted: 30 July 2024 / Published: 6 August 2024
(This article belongs to the Section Animal Nutrition)

Abstract

:

Simple Summary

Following weaning, piglets exhibit an immature immune system, reduced capacity to digest and absorb nutrients, and encounter diverse stressors that significantly compromise their overall health. The supplementation of probiotics in animal feed has proven to be a beneficial strategy for enhancing the welfare of livestock and poultry. The findings of this research demonstrate that supplementing the diet with 5 × 108 CFU/kg of Clostridium butyricum enhances fecal microflora diversity and richness through the modulation of specific metabolites such as L-aspartic acid, 5-hydroxyindole-3-acetic acid, stearic acid, and adipic acid. This mechanism contributes to a reduction in the incidence of diarrhea in weaned piglets triggered by E. coli K88. Consequently, the study outcomes offer both empirical evidence and theoretical support for the utilization of Clostridium butyricum in combatting diarrhea in piglet rearing.

Abstract

This study aimed to evaluate the effects of Clostridium butyricum (C. butyricum) on the prevention of the diarrhea rates and growth performances of weaned piglets induced by Escherichia coli K88 (E. coli K88). Twenty-four weaned piglets (6.92 ± 0.11 kg) were randomly assigned to one of three treatment groups for a period of 21 days. Each group consisted of eight pigs, with each pig being housed in an individual pen. Group I received the control diet along with normal saline, Group II received the control diet along with E. coli K88, and Group III received the control diet supplemented with 5 × 108 CFU/kg of C. butyricum and E. coli K88. We examined alterations in rectal microbiota and metabolites, analyzed the incidence of diarrhea, and investigated the interactions between microbiota and metabolites through the application of Illumina MiSeq sequencing and liquid chromatography–mass spectrometry. The results showed that, from days 14 to 21, the diarrhea incidence in Group III decreased significantly by 83.29% compared to Group II (p < 0.05). Over the entire experimental duration, the average daily feed intake of Group III decreased significantly by 11.13% compared to Group I (p < 0.05), while the diarrhea incidence in Group III decreased by 71.46% compared to Group II (p < 0.05). The predominant microbial flora in the rectum consisted of Firmicutes (57.32%), Bacteroidetes (41.03%), and Proteobacteria (0.66%). Administering E. coli K88 orally can elevate the relative abundance of Megasphaera (p < 0.05). Conversely, the supplementation of C. butyricum in the diet reduced the relative abundance of Megasphaera (p < 0.05), while increasing the relative abundance of unclassified_f_Lachnospiraceae (p < 0.05). Rectal metabolomics analysis revealed that supplementing C. butyricum in the feed significantly altered the amino acids and fatty acids of the piglets infected with E. coli K88 (p < 0.05). The correlation analysis showed that the occurrence of diarrhea was inversely related to adipic acid (p < 0.05) and positively associated with (5-hydroxyindol-3-YL) acetic acid and L-aspartic acid (p < 0.05). Prevotella_1 exhibited a negative correlation with octadecanoic acid (p < 0.05). Prevotellaceae_UCG-005 showed a negative correlation with (5-hydroxyindol-3-YL) acetic acid (p < 0.05). The findings from this research study aid in probiotic development and the enhancement of healthy growth in weaned piglets.

1. Introduction

There are a large number of complex microorganisms in the gastrointestinal tract of animals, including bacteria, fungi, protozoa, and archaea [1]. The intestinal microbiota plays an important role in preventing the colonization of pathogens, regulating immune responses, and maintaining the stability of the gastrointestinal environment [1]. The structure and composition of the gut microbiota are influenced by factors such as genetics, diet, age, external environment, and stress [2]. Newborn piglets are prone to diarrhea in the early stages of gastrointestinal development [3]. Before birth, the fetal intestine is thought to be sterile, but newborn infants are colonized by a complex microbial community shortly after birth [4]. Imbalances or dysbiosis in the neonatal gut microbiota can lead to a higher risk of disease and have adverse effects on host health [5]. Increasing evidence suggests that an early colonization of the microbiota can affect its composition and immune maturation [6,7]. Therefore, early colonization with beneficial bacteria to establish a stable gut microbiota may have practical significance in improving the health of newborn piglets. Newborn piglets are often infected with pathogenic bacteria from maternal or environmental sources [8]. Enterotoxigenic Escherichia coli (ETEC) K88 is considered a major pathogen for post-weaning diarrhea in piglets [9]. ETEC K88 infection is one of the key causes of diarrhea in both newborn and post-weaning piglets [3], leading to slow growth in pig farms and causing significant economic losses [10]. The severity of diarrhea in suckling piglets can be as high as 50% [11]. ETEC produces enterotoxins, disrupts the gut microbiota, stimulates the loss of water and electrolytes, and causes diarrhea [12]. Additionally, when weaned piglets experience intestinal damage, there may be changes in the regulation of their metabolism [13].
Probiotics can promote the proliferation of anaerobic bacteria and inhibit or eliminate harmful bacteria through competing for nutrients and growth sites, thereby continuously reducing the number of harmful bacteria and rendering them incapable of causing disease in the body [14]. Clostridium butyricum (C. butyricum) is an important component of probiotics, mainly residing in the distal small intestine and colon of animals [15], and has been used to prevent or treat gastrointestinal diseases in animals [16]. Studies have shown that adding C. butyricum to feed can regulate the gut microbiota of broiler chickens, increasing the numbers of certain beneficial bacteria and reducing the numbers of harmful bacteria in the chicken gut [17,18]. Furthermore, the liver-protective effect of C. butyricum in mice is believed to be related to its reshaping of the gut microbiota composition [19]. In recent years, metabolomics has been introduced into the study of the nutritional metabolism of post-weaning piglets [20] and finishing pigs [21]. This study advances a scientific hypothesis suggesting that C. butyricum is capable of colonizing the intestinal tract and inhibiting the growth of detrimental bacteria, thereby preserving the intestinal microecological balance and enhancing the healthy growth of piglets. Building on the previous research of our research group, this investigation aims to establish an E. coli K88 diarrhea model to explore the diversity and changes in the fecal microbiota of diarrhea piglets mediated by E. coli K88, the co-variation characteristics of the microbiota and host metabolism, and to identify important fecal bacterial types and biomarkers closely associated with host diarrhea.

2. Materials and Methods

2.1. Animals, Diet, and Management

A total of 24 weaning piglets [(Yorkshire × Landrace) × Duroc] (half male, half female) were weaned at 28 days of age (6.92 ± 0.11 kg BW) and randomly divided into three groups for 21 days, with eight pigs per group. Each pig was fed in a single pen, receiving a control diet (Group I), a control diet (Group II), or a diet supplemented with 5 × 108 CFU/kg C. butyricum (Group III). On the 18th day of the experiment, piglets in Group I were gavaged (force-fed) sterile saline, and the other two groups’ piglets were gavaged E. coli K88 for three consecutive days. The diets were a highly digestible basal diet, including highly digestible carbohydrate ingredients and low, anti-nutritional protein ingredients. The composition and nutrient content of the treatment diets are presented in Table 1. The C. butyricum culture was provided by Zhejiang Huijia Biological Technology Co., Ltd. (Anji, Zhejiang, China). The commercial product contained a viable count of 5.0 × 108 CFU/g.
The pigs were housed in a climate-controlled facility with regulated temperature and humidity to minimize environmental stress. The room temperature was maintained at 28–30 °C for the first week and then gradually reduced to 22–24 °C. Humidity was kept between 55 and 65%. All pigs were subjected to a 12 h light/dark cycle and had ad libitum access to water and their respective diets. Regular cleaning and disinfection protocols were strictly followed to maintain hygiene and reduce the risk of disease. This experiment was carried out at Fushun Breeding Farmer Professional Cooperative, a pig farm located in Shihezi, Xinjiang.

2.2. Oral Challenge

The E. coli K88 strain was obtained from the Jiangsu Academy of Agricultural Sciences (Nanjing, China) and grown at 37 °C. The piglets fasted for 12 h before modeling. Firstly, 10 mL of 1% sodium bicarbonate solution was gavaged, and, after one hour, E. coli K88 10 mL (1.0 × 1010 CFU/mL) was gavaged once a day for three consecutive days. Group I was treated with sterile saline in the same way.

2.3. Sample Collection

Body weight per pen was recorded on days 1, 7, 14, and 21 to calculate average daily gain (ADG). Feed was weighed and provided daily, and the feed consumption was determined on day 7, day 14, and day 21 by weighing the remaining feed in the feeder. The average daily feed intake (ADFI) and feed-to-gain ratio (F/G) of each phase were calculated. In the experimental period, the piglets were observed daily for the presence of diarrhea: the number of piglets experiencing diarrhea per day in each pen was recorded, and the consistency of the feces was simultaneously evaluated according to 4 levels: 0, normal; 1, pasty; 2, semi-liquid; and 3, liquid [22]. Diarrhea was defined as fecal output at level 2 or 3 for two consecutive days. Fecal consistency was visually assessed daily. Diarrhea incidence of each group (%) = the total number of days of diarrhea of piglets in the group during the experiment/(the number of piglets in the group × the number of days of the experiment) × 100%
On day 21 of the experiment, six piglets in each group were randomly selected. About 20 g of fresh feces was collected by rectal stimulation method and stored in sterile cryopreservation tubes, which were immediately placed in liquid nitrogen, and stored at −80 °C in a refrigerator for further analysis. Six samples were employed for the analysis of metabolites, and, from these, three samples were randomly chosen for 16S rRNA high-throughput sequencing.

2.4. DNA Extraction

Using a TIANamp Stool QIAamp PowerFecal DNA Kit (QIAGEN, Hilden, Germany), microbial DNA was extracted from fecal samples according to the instructions in the kit. NanoDrop 2000 UV-VIS spectrophotometer (Thermo Scientific, Wilmington, NC, USA) DNA concentration and purity were determined, and DNA quality and concentration were determined by 1% agarose gel electrophoresis.

2.5. Illumina MiSeq Sequencing

Using a PCR thermocycling system (GeneAmp 9700, ABI, Los Angeles, CA, USA), the samples of bacterial 16S rRNA hypervariable region were amplified with primers 515F (5′-GTGCCAGCMGCCGCGGTAA-3′) and 907R (5′-CCGTCAATTCCTTTGAGTTT-3′) and sent to Shanghai Magi Biomedical Technology Co., Ltd., Shanghai, China. Illumina sequencing was performed on the MiSeq platform (Illumina, San Diego, CA, USA). The raw sequencing data were deposited in the NCBI Sequence Read Archive (SRA, http://www.ncbi.nlm.nih.gov/sra) under the accession number PRJNA679230 accessed on 18 November 2020.

2.6. Data Processing

Raw FASTQ files were quality-filtered by Trimmomatic software (Version 0.36) and combined by Quick Length Adjustment (FLASH) for short reads according to the following criteria. UPARSE software (Version 7.1, http://drive5.com/uparse/, accessed on 18 November 2020), clustering, was used according to the similarity of the 97% cutoff value for OTU. The 16S rRNA gene sequences were classified and analyzed using the Ribosomal Database Project (RDP) classification algorithm (http://rdp.cme.msu.edu/, accessed on 18 November 2020) and SILVA 16S rRNA gene reference database (https://www.arb-silva.de/, accessed on 18 November 2020), with a confidence threshold of 70%.

2.7. Extraction of Metabolites

First, 100 mg of feces was placed in a 5 mL tube, and 500 μL of dd H2O was added at 4 °C and thoroughly mixed for 1 min. In the second step, 1 mL of methanol at −20 °C was added to vortex for 30 s. The third step is to place the ultrasonic machine at room temperature for 10 min and then place it on ice for 30 min. The fourth step was to centrifuge at 14,000 rpm at 4 °C for 10 min; 1200 μL of supernatant was taken and transferred to a new 1.5 mL centrifuge tube. The sample was concentrated with a vacuum centrifuge concentrator. In the fifth step, the sample was dissolved in 400 μL of methanol solution at 4 °C in a ratio of 1:1 and filtered with a 0.22 μm membrane to obtain the sample for testing. Then, transfer 20 μL from each sample to be tested and mix it into the QC sample (QC: quality control, used to correct the deviation of the analysis’ result of the mixed sample and the error caused by the analytical instrument itself). Finally, the remaining samples were tested using LC–MS.

2.8. Chromatographic Condition

The Waters ACQUITY UPLC (ACQUITY UPLC)® HSS T3 1.8 μm (2.1 × 150 mm) column was utilized for analysis. The automatic sampler temperature was maintained at 4 °C, with a flow rate set to 0.25 mL/min, while the column temperature was kept at 40 °C during the 40 μL gradient elution. The mobile phase consisted of 0.1% formic acid in water (A) and 0.1% formic acid in acetonitrile (B). The gradient elution profile included the following steps: 0–1 min, 2% B; 1–9.5 min, 2–0% B; 9.5–14 min, 50–98% B; 14–15 min, 98% B; 15–15.5 min, 98–2% B; 15.5–17 min, 2%.

2.9. Mass Spectrometry Conditions

Thermo LTQ Orbitrap XL, electrospray ion source (ESI), positive and negative ionization mode, positive ion spray voltage 4.80 kV, negative ion spray voltage 4.50 kV, sheath gas at 45 arb, and auxiliary gas at 15 arb were applied. The capillary temperature was 325 °C, the capillary voltage was 35 V/−15 V, and the tube lens voltage was 50 V/−50 V. The full scan was carried out with a resolution of 60,000, and the scanning range was 50~1000. Secondary cracking was carried out with CID, and the collision voltage was 30 eV. At the same time, the unnecessary MS/MS information was removed by dynamic exclusion (the repeat count was 2), and the dynamic exclusion time was set to 15 s.

2.10. Data Processing

ProteoWizard software (Version 3.0.8789) was used to change the original file to MzXML format (XCMS input file format). R’s XCMS package (Version 3.3.2) was used for peak detection peak filtering and peak alignment. Then, a two-dimensional data matrix consisting of the mass/charge ratio (m/z) residence time (RT) and peak area of metabolites were obtained. SIMCA-P 13.0 software (Umetrics AB, Umea, Sweden) was introduced for multivariate statistical analysis. Partial least-squares discriminant analysis (PLS-DA) was used for predictive description modeling and discriminant variable selection. The significant variables under differential multiples (FC ≥ 1.5 or FC ≤ 0.667) obtained by OPLS-DA and t test (p < 0.05) were used to analyze the metabolites differentially expressed in the two groups. Then, accurate MS/MS fragment information was searched for in the human metabolome database (HMDB, http://www.hmdb.ca/, accessed on 18 November 2020), Metlin database (http://metlin.scripps.edu/, accessed on 18 November 2020), and KEGG database (http://www.genome.jp/kegg/, accessed on 18 November 2020) for screening and identification of potential biomarkers. Metabolic pathways were analyzed by MetaboAnalyst R 3.0, available at http://www.metaboanalyst.ca, accessed on 18 November 2020.

2.11. Bioinformatics and Statistical Analyses

The growth performance data and alpha diversity index data were reported as mean ± standard deviation (SD), with one-way analysis of variance (ANOVA) with Duncan’s multiple range test. The diarrhea rate data were analyzed using the chi-square test. The rank–abundance and rarefaction curves were made by using R language tools based on Majorbio Cloud (https://cloud.majorbio.com/, accessed on 18 November 2020). The software Mothur (version v.1.30.2; https://mothur.org/wiki/calculators/, accessed on 18 November 2020) was used to perform α-diversity index analysis. Community composition analysis involved plotting with R language tools (version 3.3.1). Heat maps were generated using the VEGAN R package (Version 3.3.1), and the VEGAN package facilitated non-metric multidimensional scaling (NMDS) analysis. Graphical representations, like histograms and Venn diagrams, were created through Origin 8.0 software by OriginLab. Spearman’s Rho test assessed the relationship between bacteria displaying significant changes and their metabolites (with FC exceeding 0.667 or 1.5, after adjustment, and p < 0.05). The correlation map illustrating the link between microbiota and metabolites was produced using the Jidiao cloud platform available at https://www.omicshare.com, accessed on 18 November 2020. The statistical analysis for this experiment utilized SPSS version 22.0. A significance level of p ≤ 0.01 indicated a highly significant difference, p ≤ 0.05 indicated a significant difference, and p > 0.05 signified an insignificant difference.

3. Results

3.1. Growth Performance and Diarrhea Incidence

At 0–7 and 7–14 days post-weaning, there were no significant differences in feed conversion efficiency and diarrhea incidence among all experimental groups (p > 0.05). However, from 14 to 21 days after weaning, the average daily feed intake (ADFI) of group III decreased significantly by 17.47% compared to group II (p < 0.05), while the occurrence of diarrhea in group III decreased significantly by 83.29% compared to group II (p < 0.05). Over the entire duration of the experiment, the ADFI in group III decreased significantly by 11.13% compared to group I (p < 0.05), and the incidence of diarrhea in group III decreased significantly by 71.46% compared to group II (Table 2).

3.2. The Fecal Microbial Community Structure in Weaned Piglets Challenged with E. coli K88

Based on a 97% similarity threshold, all sequences were clustered into operational taxonomic units (OTUs), resulting in the identification of a total of 456 OTUs. Notably, 349 OTUs were shared among the three treatment groups, representing 76.54% of the total OTUs, while 82 OTUs were common between the two treatment groups, accounting for 17.98% of the total OTUs (Figure 1).
The sequencing coverage rate for each treatment group exceeded 99%, suggesting a high probability of sequence detection in the samples and a low likelihood of missed detection. These sequencing results accurately reflect the microbial composition of the samples. The Sobs index in group I and group III increased significantly by 22.04% and 23.23%, respectively, compared to group II (p < 0.05). Furthermore, the Shannon index in group I showed a significant increase of 20.35% compared to group II (p < 0.05). No significant differences were observed in the Ace and Chao indices among the various treatments (p > 0.05), indicating that the C. butyricum (CB) treatment group displayed high richness and diversity (Table 3).
In Figure 2A, each sample from the three groups is distributed and clustered at the operational taxonomic unit (OTU) level with relatively minor discrepancies. The partial least-squares discriminant analysis (PLS-DA) effectively distinguishes the observed values between groups and identifies the influential variables contributing to between-group differences. The distinct separation of the three treatment groups without sample overlap is evident in Figure 2B. The suspected influencing factor for microbial compositional divergence between group I and group II was Comp1, with the explanatory weight ratio of each dimension on the outcomes. Additionally, the suspected influential factor associated with group II and group III was C. butyricum, denoted as Comp2. The combination of the sample information revealed ETEC K88 as a potential influential factor for group I (Figure 2).
Figure 3A displays three categories of microorganisms—Firmicutes, Bacteroidetes, and Proteobacteria—with average abundances of 57.32%, 41.03%, and 0.66%, respectively. At the genus level (Figure 3B), 141 genera were identified, including Lactobacillus (15.68%), Prevotella_9 (13.72%), and Megasphaera (12.38%). The relative abundance of unclassified_f_Lachnospiraceae in group III significantly exceeded that in group II (p < 0.001), while the relative abundance of Megasphaera in group III notably decreased compared to group II (p = 0.047) (Figure 3).
Figure 4 illustrates the bacterial communities of the three treatment groups, grouping them distinctly: group II forms one cluster, while group I and group III constitute another, signifying the high similarity and separate clustering of the bacterial communities in group I and group III from group II (Figure 4).

3.3. Fecal Metabolites

Figure 5A,B present PLS-DA models for dimension reduction analysis. In each figure, every point represents a sample. The PLS-DA model yields the following results: R2X (cum) = 0.531, R2Y (cum) = 0.992, and Q2 (cum) = 0.933 in positive ion mode; and R2X (cum) = 0.563, R2Y (cum) = 0.993, and Q2 (cum) = 0.937 in negative ion mode. The Permutations Plot aids in effectively assessing the potential overfitting of the current PLS-DA model. An essential criterion is that all blue Q2 values should be lower from left to right compared to the original blue Q2, or the regression line of Q2 should be at or below zero at the vertical coordinate intersection. Figure 5C (positive ion mode) and Figure 5D (negative ion mode) support the reliability and effectiveness of this model. An additional OPLS-DA analysis was conducted to further diminish intra-group differences and accentuate inter-group distinctions. Illustrated in Figure 5E,F, the model yields the following parameters: R2X (cum) = 0.415, R2Y (cum) = 0.996, and Q2 (cum) = 0.831 in positive ion mode; and R2X (cum) = 0.335, R2Y (cum) = 0.991, and Q2 (cum) = 0.803 in negative ion mode. The fecal metabolite score maps, constructed within the 95% Hotelling T2 confidence ellipse, display clear separation, indicating a strong identification and prediction accuracy of the model. The scatter plot division among the three treatment groups indicates significant differences in the composition and concentration of the variables or molecules present in the samples (Figure 5).
Table 4 illustrates two distinct metabolites between group I and group II: L-aspartic acid and (5-hydroxyindol-3-YL) acetic acid. Group I and group III exhibited four metabolites that varied, identified as L-phenylalanyl-L-proline, 2-keto-3-deoxy-D-gluconic acid, octadecanoic acid, and CMP. The comparison between group II and group III revealed four differential metabolites, specifically L-aspartic acid, (5-hydroxyindol-3-YL) acetic acid, octadecanoic acid, and adipic acid. Significantly higher levels of L-aspartic acid and (5-hydroxyindol-3-YL) acetic acid were observed in group II compared to group III (p < 0.05), while octadecanoic acid and adipic acid levels notably decreased (p < 0.05).
MetaboAnalystR 3.0 was utilized for the pathway enrichment profiling of the distinct metabolites. The metabolites in group I and group II were primarily associated with bacterial chemotaxis, biosynthesis of alkaloids for tannin production, lysine, nicotinic acid, and arginine biosynthesis pathways (Figure 6A). Differential metabolites within group I and group III were predominantly linked to photoconduction, caprolactam degradation, and pentose phosphate pathway metabolism (Figure 6B). In contrast, the differential metabolites between group II and group III were primarily involved in metabolic pathways such as apoptosis, necrotic apoptosis, sphingolipid signaling, bacterial chemotaxis, ABC transporters, and interactions in neural tissues (Figure 6C).

3.4. Analysis of the Correlation between Diarrhea Incidence, Body Weight, Fecal Microbial Community, and Fecal Metabolites

The Spearman correlation analysis results are depicted in Figure 7. The diarrhea rate exhibited a negative correlation with adipic acid (p < 0.05) and was positively correlated with 5-hydroxyindole-3-acetic acid, 2-keto-3-deoxy-D-gluconic acid, and L-aspartic acid (p < 0.05) (Figure 7B). Furthermore, Prevotella demonstrated a positive correlation with cytidine acid (p < 0.05) and a negative correlation with stearic acid (p < 0.05). Additionally, Prevotellaceae showed a negative correlation with 5-hydroxyindole-3-acetic acid (p < 0.05) (Figure 7C).

4. Discussion

4.1. The Impact of C. butyricum on the Growth Performance and Diarrhea Rate Induced by Escherichia coli K88 in Weaned Piglets

Post-weaning diarrhea presents a significant threat to piglet production, associated with the intestinal colonization of enterotoxigenic Escherichia coli (ETEC), leading to gut dysbiosis, inflammation, growth retardation, and potential mortality [23]. ETEC poses a crucial challenge in the swine industry, impacting post-weaning performance and piglet survival rates [24], possibly through immune-barrier enhancement and gut microbiota alterations. Probiotics and organic acids can regulate cytokine production by inhibiting ETEC K88 proliferation and toxin release, thereby ameliorating the inflammatory response [25,26]. Research suggests that supplementing weaned piglet diets with C. butyricum has been shown to enhance growth performance in pigs fed less digestible diets. There was a noticeable trend towards a reduction in the feed-to-gain ratio (F/G), which could potentially lead to cost savings in pig production. The positive effects may be attributed to a decrease in post-weaning diarrhea, which is associated with improvements in intestinal morphology, intestinal microflora composition, and immune function [27]. A study by Han [28] demonstrated that supplementing feed with C. butyricum enhances growth performance in weaned piglets, improves feed efficiency, reduces the feed-to-gain ratio significantly, and decreases diarrhea incidence. Numerous studies have demonstrated that C. butyricum can enhance growth performance and optimize nutrient utilization in animals [29,30,31]. However, some research findings have not observed an impact on growth performance [32,33,34]. In this study, the incorporation of C. butyricum in feed had no significant impact on feed efficiency but notably reduced the diarrhea rate in piglets, likely attributed to the production of volatile fatty acids like butyric acid, which lower intestinal pH and impede the growth of harmful bacteria.

4.2. The Impact of C. butyricum on the Fecal Microbiota of Weaned Piglets Stimulated by Escherichia coli K88

The equilibrium of the animal intestinal microbiota is crucial for sustaining overall animal health. This delicate balance is vulnerable to interference from external pathogens. Should this balance be disrupted, it can result in the onset of various diseases within the body. Adding probiotics to the diet can enhance the intestinal microbiota [35,36], improve gastrointestinal function [37], and boost systemic immunity [38], consequently enhancing animal production performance [37]. Studies have demonstrated that prolonged antibiotic use can modify the structure and composition of the gut microbiota, leading to a decreased expression of immune response genes [9,39]. With the implementation of widespread antibiotic restrictions, probiotics, prebiotics, synbiotics, and other supplements serve as antibiotic alternatives, proven to regulate the intestinal microbiota and enhance the well-being of piglets. C. butyricum is adept at maintaining this microecological equilibrium by enhancing the population of beneficial bacteria and impeding the colonization and growth of pathogens. It also works in concert with other beneficial bacteria to neutralize harmful bacteria, vie with pathogens for colonization sites, and diminish the probability of pathogen colonization, thereby checking the growth of pathogens. The metabolic activities of C. butyricum yield compounds such as short-chain fatty acids (SCFAs) and antimicrobial peptides that exert inhibitory effects on pathogens. SCFAs can reduce the pH level of the intestinal environment, which in turn suppresses the proliferation of pathogens. Additionally, bacteriocins and antimicrobial peptides can weaken the colonization potential of pathogens or directly assault them, thereby safeguarding intestinal health [40,41].
In the intestinal microbiota of weaned piglets, the main phyla are Firmicutes and Bacteroidetes [42,43]. The majority of Bacteroidetes can decompose starch, hemicellulose, and protein. They hydrolyze indigestible dietary polysaccharides into smaller molecules that are more easily absorbed by the body, providing supplementary energy [44]. Hossain [45] administered a composite probiotic blend of C. butyricum, Bacillus subtilis, and Lactobacillus to broilers. The study revealed that probiotic supplementation significantly increased Lactobacillus and Bifidobacterium counts, while notably inhibiting the growth of Escherichia coli and Clostridium perfringens. Here, the higher 0.2% dosage exhibited superior inhibitory effects compared to the lower 0.1% dose, outperforming the antibiotic group. The dietary supplementation with C. butyricum or Enterococcus faecalis appears to have conferred several advantages to the weaned piglets challenged with lipopolysaccharide (LPS). The supplements enhanced the pigs’ growth performance, bolstered their immune response, alleviated damage to the intestinal villi, reduced inflammation, and improved the balance of the intestinal microbiota [46].
The Lachnospiraceae family is a fundamental component of the gut microbiota, colonizing the intestinal lumen from birth and expanding in terms of species diversity and relative abundance throughout the host’s life span [47]. Lachnospiraceae can produce large amounts of SCFAs, such as acetic acid by metabolizing L-glutamic acid [48]. Megasphaera is a primary lactic acid-utilizing bacterium. It can decompose lactic acid into propionate via two pathways: acrylic acid and succinate. Alternatively, it can produce acetic acid through the acetyl–CoA pathway with the action of acetyl kinase (AK) and phosphotransacetylase (PTA). Furthermore, acetic and propionic acids are converted into butyric and valeric acids [49,50]. In this investigation, the addition of 5 × 108 CFU/kg C. butyricum in the diet of diarrheic piglets markedly enhanced the diversity of the fecal microbiota, substantially raised the relative abundance of unclassified_f_Lachnospiraceae, and significantly decreased the relative abundance of Megasphaera. This may be due to the ability of C. butyricum to improve the stability of the animal’s digestion environment, thereby enhancing the capacity to recover health from a state of diarrhea.

4.3. The Influence of C. butyricum on Fecal Metabolites in Weaned Piglets Affected by Escherichia coli K88

The metabolome serves as a comprehensive reflection of the genome, transcriptome, and proteome and is instrumental in exploring the metabolic pathways within biological systems. Aspartic acid, an α-amino acid, acts as a precursor for synthesizing amino acids like lysine, threonine, isoleucine, and methionine, as well as purine and pyrimidine bases. Additionally, it can function as a crucial excitatory neurotransmitter receptor in the central nervous system of mammals [51]. Stearic acid, a naturally abundant fatty acid, is found in various fats, particularly in animal fats. Adipic acid, a valuable dicarboxylic acid within the fatty acid group, holds wide-ranging applications in medicine, pesticides, and fuels [52]. The results of fecal metabolite analysis from this study reveal that the supplementation of C. butyricum to the diet of diarrheic piglets leads to the presence of four distinct fecal metabolites, namely, L-aspartic acid, 5-hydroxyindole-3-acetic acid, stearic acid, and adipic acid, which serve as potential biomarkers. The major metabolic pathways involved encompass cell apoptosis, necrotic apoptosis, sphingolipid signaling pathways, and bacterial chemotaxis. The correlation analysis demonstrates a negative association between diarrhea incidence and adipic acid levels, as well as a positive relationship with L-aspartic acid and 5-hydroxyindole-3-acetic acid. This suggests that C. butyricum in weaned piglets may offer protection against E. coli K88 infection by decreasing diarrhea occurrence through elevating adipic acid levels and reducing L-aspartic acid and 5-hydroxyindole-3-acetic acid levels.

5. Conclusions

The supplementation of the diet with 5 × 108 CFU/kg of C. butyricum significantly decreases the occurrence of diarrhea induced by Escherichia coli K88 in weaned piglets. This supplementation also enhances fecal microbiota diversity and richness in piglets affected by E. coli K88. This is mainly related to the metabolism of L-aspartic acid, 5-hydroxyindole-3-acetic acid, stearic acid, and adipic acid. C. butyricum could potentially serve as a feasible alternative to antibiotics for preventing E. coli K88 infection. The outcomes of this study provide empirical evidence and a theoretical basis for the application of C. butyricum in preventing diarrhea in piglet production.

Author Contributions

Conceptualization, J.L., S.W. and W.Z.; implementation, J.L. and S.K.; formal analysis and methodology, C.C., S.W. and C.N.; writing—original draft preparation, J.L.; writing—review and editing, S.W. and C.N.; visualization, J.L. and S.W.; supervision, C.N.; project administration, W.Z. and C.N.; funding acquisition, C.N. and J.L. All authors have read and agreed to the published version of the manuscript.

Funding

This study was financially supported by the “high-level talent program” project from Yulin University (grant number: 22GK21).

Institutional Review Board Statement

This study was approved by the Animal Care and Use Committee of Shihezi University (approval code: A2019-154-01).

Informed Consent Statement

Not applicable.

Data Availability Statement

The original contributions presented in the study are included in the article; further inquiries can be directed to the corresponding authors.

Acknowledgments

We acknowledge Zhejiang Huijia Biological Technology Co., Ltd. (Zhejiang, China), for providing Clostridium butyricum. The commercial product contained a viable count of 5.0 × 108 CFU/g.

Conflicts of Interest

There are no conflicts of interest to disclose for this study.

References

  1. Brüssow, H. Microbiota and the human nature: Know thyself. Environ. Microbiol. 2015, 17, 10–15. [Google Scholar] [CrossRef] [PubMed]
  2. Aluthge, N.D.; Van Sambeek, D.M.; Carney-Hinkle, E.E.; Li, Y.S.; Fernando, S.C.; Burkey, T.E. BOARD INVITED REVIEW: The pig microbiota and the potential for harnessing the power of the microbiome to improve growth and health1. J. Anim. Sci. 2019, 97, 3741–3757. [Google Scholar] [CrossRef] [PubMed]
  3. Stokes, C.R.; Bailey, M.; Haverson, K.; Harris, C.; Jones, P.; Inman, C.; Pié, S.; Oswald, I.P.; Williams, B.A.; Akkermans, A.D.; et al. Postnatal development of intestinal immune system in piglets: Implications for the process of weaning. Anim. Res. 2004, 53, 325–334. [Google Scholar] [CrossRef]
  4. Gaskins, H.R.; Croix, J.A.; Nakamura, N.; Nava, G.M. Impact of the Intestinal microbiota on the development of mucosal defense. Clin. Infect. Dis. 2008, 46 (Suppl. S2), S80–S86. [Google Scholar] [CrossRef] [PubMed]
  5. Fanaro, S.; Chierici, R.; Guerrini, P.; Vigi, V. Intestinal microflora in early infancy: Composition and development. Acta Paediatr. 2003, 91, 48–55. [Google Scholar] [CrossRef] [PubMed]
  6. Rauch, M.; Lynch, S. Probiotic manipulation of the gastrointestinal microbiota. Gut Microbes 2010, 1, 335–338. [Google Scholar] [CrossRef] [PubMed]
  7. Rakoff-Nahoum, S.; Medzhitov, R. Innate immune recognition of the indigenous microbial flora. Mucosal Immunol. 2008, 1 (Suppl. S1), S10–S14. [Google Scholar] [CrossRef] [PubMed]
  8. Konstantinov, S.R.; Awati, A.A.; Williams, B.A.; Miller, B.G.; Jones, P.; Stokes, C.R.; Akkermans, A.D.L.; Smidt, H.; de Vos, W.M. Post-natal development of the porcine microbiota composition and activities. Environ. Microbiol. 2006, 8, 1191–1199. [Google Scholar] [CrossRef]
  9. Rhouma, M.; Fairbrother, J.M.; Beaudry, F.; Letellier, A. Post weaning diarrhea in pigs: Risk factors and non-colistin-based control strategies. Acta Vet. Scand. 2017, 59, 31–50. [Google Scholar] [CrossRef]
  10. Fairbrother, J.M.; Nadeau, E.; Gyles, C.L. Escherichia coli in postweaning diarrhea in pigs: An update on bacterial types, pathogenesis, and prevention strategies. Anim. Health Res. Rev. 2005, 6, 17–39. [Google Scholar] [CrossRef]
  11. Sayan, H.; Assavacheep, P.; Angkanaporn, K.; Assavacheep, A. Effect of Lactobacillus salivarius on growth performance, diarrhea incidence, fecal bacterial population and intestinal morphology of suckling pigs challenged with F4+ enterotoxigenic Escherichia coli. Asian-Australas. J. Anim. Sci. 2018, 31, 1308–1314. [Google Scholar] [CrossRef]
  12. Nagy, B.; Fekete, P.Z. Enterotoxigenic Escherichia coli in veterinary medicine. J. Med. Microbiol. 2005, 295, 443–454. [Google Scholar] [CrossRef] [PubMed]
  13. Campbell, J.M.; Crenshaw, J.D.; Polo, J. The biological stress of early weaned piglets. J. Anim. Sci. Biotechnol. 2013, 4, 19–23. [Google Scholar] [CrossRef]
  14. Hu, F.; Gao, X.; She, R.; Chen, J.; Mao, J.; Xiao, P.; Shi, R. Effects of antimicrobial peptides on growth performance and small intestinal function in broilers under chronic heat stress. Poult. Sci. 2017, 96, 798–806. [Google Scholar] [CrossRef]
  15. Sato, S.; Nagai, H.; Igarashi, Y. Effect of probiotics on serum bile acids in patients with ulcerative colitis. Hepatogastroenterology 2012, 59, 1804–1808. [Google Scholar] [PubMed]
  16. Hayashi, A.; Sato, T.; Kamada, N.; Mikami, Y.; Matsuoka, K.; Hisamatsu, T.; Hibi, T.; Roers, A.; Yagita, H.; Ohteki, T.; et al. A single strain of Clostridium butyricum induces intestinal IL-10-producing macrophages to suppress acute experimental colitis in mice. Cell Host Microbe 2013, 13, 711–722. [Google Scholar] [CrossRef] [PubMed]
  17. Zhao, X.; Guo, Y.; Guo, S.; Tan, J. Effects of Clostridium butyricum and Enterococcus faecium on growth performance, lipid metabolism, and cecal microbiota of broiler chickens. Appl. Microbiol. Biotechnol. 2013, 97, 6477–6488. [Google Scholar] [CrossRef]
  18. Zhang, S.T.; Shi, Y.; Zhang, S.L.; Wang, H.K. Progress on the Studies and Application of Clostridium butyricum. Biotechnol. Bull. 2013, 9, 27–33. [Google Scholar] [CrossRef]
  19. Liu, J.; Fu, Y.; Zhang, H.; Wang, J.; Zhu, J.; Wang, Y.; Guo, Y.; Wang, G.; Xu, T.; Chu, M.; et al. The hepatoprotective effect of the probiotic Clostridium butyricum against carbon tetrachloride-induced acute liver damage in mice. Food Funct. 2017, 8, 4042–4052. [Google Scholar] [CrossRef]
  20. He, Q.H.; Tang, H.R.; Ren, P.P.; Kong, X.F.; Wu, G.Y.; Yin, Y.L.; Wang, Y.L. Dietary supplementation with l-arginine partially counteracts serum metabonome induced by weaning stress in piglets. J. Proteome Res. 2011, 10, 5214–5221. [Google Scholar] [CrossRef]
  21. Yu, M.; Li, Z.; Chen, W.; Rong, T.; Wang, G.; Ma, X. Microbiome-Metabolomics Analysis Investigating the Impacts of Dietary Starch Types on the Composition and Metabolism of Colonic Microbiota in Finishing Pigs. Front. Microbiol. 2019, 10, 1143–1156. [Google Scholar] [CrossRef]
  22. Liu, P.; Piao, X.S.; Thacker, P.A.; Zeng, Z.K.; Li, P.F.; Wang, D.; Kim, S.W. Chito-oligosaccharide reduces diarrhea incidence and attenuates the immune response of weaned pigs challenged with Escherichia coli K88. J. Anim. Sci. 2010, 88, 3871–3879. [Google Scholar] [CrossRef]
  23. Guevarra, R.B.; Lee, J.H.; Lee, S.H.; Seok, M.J.; Kim, D.W.; Kang, B.N.; Johnson, T.J.; Isaacson, R.E.; Kim, H.B. Piglet gut microbial shifts early in life: Causes and effects. J. Anim. Sci. Biotechnol. 2019, 10, 1. [Google Scholar] [CrossRef] [PubMed]
  24. Bhandari, S.K.; Xu, B.; Nyachoti, C.M.; Giesting, D.W.; Krause, D.O. Evaluation of alternatives to antibiotics using an Escherichia coli K88+ model of piglet diarrhea: Effects on gut microbial ecology. J. Anim. Sci. 2008, 86, 836–847. [Google Scholar] [CrossRef] [PubMed]
  25. Li, S.; Zheng, J.; Deng, K.; Chen, L.; Zhao, X.L.; Jiang, X.; Fang, Z.; Che, L.; Xu, S.; Feng, B.; et al. Supplementation with organic acids showing different effects on growth performance, gut morphology, and microbiota of weaned pigs fed with highly or less digestible diets. J. Anim. Sci. 2018, 96, 3302–3318. [Google Scholar] [CrossRef] [PubMed]
  26. Li, Y.; Zhang, H.; Su, W.; Ying, Z.; Chen, Y.; Zhang, L.; Lu, Z.; Wang, T. Effects of dietary supplementation on growth performance, intestinal morphology, inflammatory response, and microbiota of intra-uterine growth retarded weanling piglets. J. Anim. Sci. Biotechnol. 2018, 9, 22–38. [Google Scholar] [CrossRef] [PubMed]
  27. Chen, L.; Li, S.; Zheng, J.; Li, W.; Jiang, X.; Zhao, X.; Li, J.; Che, L.; Lin, Y.; Xu, S.; et al. Effects of dietary Clostridium butyricum supplementation on growth performance, intestinal development, and immune response of weaned piglets challenged with lipopolysaccharide. J. Anim. Sci. Biotechnol. 2018, 9, 62–76. [Google Scholar] [CrossRef]
  28. Han, Y.S.; Tang, C.H.; Li, Y.; Yu, Y.N.; Zhan, T.F.; Zhao, Q.Y.; Zhang, J.M. Effects of Dietary Supplementation with on Growth Performance, Serum Immunity, Intestinal Morphology, and Microbiota as an Antibiotic Alternative in Weaned Piglets. Animals 2020, 10, 2287. [Google Scholar] [CrossRef]
  29. Zhang, L.; Cao, G.T.; Zeng, X.F.; Zhou, L.; Ferket, P.R.; Xiao, Y.P.; Chen, A.G.; Yang, C.M. Effects of Clostridium butyricum on growth performance, immune function, and cecal microflora in broiler chickens challenged with Escherichia coli K88. Poult. Sci. 2014, 93, 46–53. [Google Scholar] [CrossRef]
  30. Zhang, L.; Zhan, X.; Zeng, X.; Zhou, L.; Cao, G.; Chen, A.; Yang, C. Effects of dietary supplementation of probiotic, Clostridium butyricum, on growth performance, immune response, intestinal barrier function, and digestive enzyme activity in broiler chickens challenged with Escherichia coli K88. J. Anim. Sci. Biotechnol. 2016, 7, 3–12. [Google Scholar] [CrossRef]
  31. Huang, J.; Cui, L.; Lin, H.; Song, M.; Sun, S. Effects of Clostridium butyricum on Production Performance and Bone Development of Laying Hens. Vet. Sci. 2024, 11, 160. [Google Scholar] [CrossRef] [PubMed]
  32. Zhang, C.; Hou, T.; Wang, J.; Yu, Q.; Zhang, Y.; Sun, Y. Clostridium butyricum alleviates LPS-induced acute immune stress in goats by regulating bacterial communities and blood metabolites. Front. Immunol. 2023, 14, 1099186. [Google Scholar] [CrossRef] [PubMed]
  33. Zhang, C.; Yu, Q.; Wang, J.; Yu, Y.; Zhang, Y.; Sun, Y. Effects of Dietary Supplementation With Clostridium butyricum on Growth Performance, Apparent Digestibility, Blood Metabolites, Ruminal Fermentation and Bacterial Communities of Fattening Goats. Front. Nutr. 2022, 9, 888191. [Google Scholar] [CrossRef] [PubMed]
  34. Li, H.H.; Li, Y.P.; Zhu, Q.; Qiao, J.Y.; Wang, W.J. Dietary supplementation with Clostridium butyricum helps to improve the intestinal barrier function of weaned piglets challenged with enterotoxigenic Escherichia coli K88. J. Appl. Microbiol. 2018, 125, 964–975. [Google Scholar] [CrossRef] [PubMed]
  35. Foysal, M.J.; Fotedar, R.; Siddik, M.A.B.; Tay, A. Lactobacillus acidophilus and L. plantarum improve health status, modulate gut microbiota and innate immune response of marron (Cherax cainii). Sci. Rep. 2020, 10, 5916–5929. [Google Scholar] [CrossRef] [PubMed]
  36. Chen, Y.C.; Yu, Y.H. Bacillus licheniformis-fermented products improve growth performance and the fecal microbiota community in broilers. Poult. Sci. 2020, 99, 1432–1443. [Google Scholar] [CrossRef] [PubMed]
  37. Wang, K.; Cao, G.; Zhang, H.; Li, Q.; Yang, C. Effects of Clostridium butyricum and Enterococcus faecalis on growth performance, immune function, intestinal morphology, volatile fatty acids, and intestinal flora in a piglet model. Food Funct. 2019, 10, 7844–7854. [Google Scholar] [CrossRef] [PubMed]
  38. Abdo, Z.; LeCureux, J.; LaVoy, A.; Eklund, B.; Ryan, E.P.; Dean, G.A. Impact of oral probiotic Lactobacillus acidophilus vaccine strains on the immune response and gut microbiome of mice. PLoS ONE 2019, 14, e0225842. [Google Scholar] [CrossRef] [PubMed]
  39. Dethlefsen, L.; Huse, S.; Sogin, M.L.; Relman, D.A. The pervasive effects of an antibiotic on the human gut microbiota, as revealed by deep 16S rRNA sequencing. PLoS Biol. 2008, 6, e280. [Google Scholar] [CrossRef]
  40. Liu, M.; Uyanga, V.A.; Cao, X.; Liu, X.; Lin, H. Regulatory Effects of the Probiotic Clostridium butyricum on Gut Microbes, Intestinal Health, and Growth Performance of Chickens. J. Poult. Sci. 2023, 60, 2023011. [Google Scholar] [CrossRef]
  41. Yamamoto, M.; Ohmori, H.; Takei, D.; Matsumoto, T.; Takemoto, M.; Ikeda, M.; Sumimoto, R.; Kobayashi, T.; Ohdan, H. Clostridium butyricum affects nutrition and immunology by modulating gut microbiota. Biosci. Microbiota Food Health 2022, 41, 30–36. [Google Scholar] [CrossRef] [PubMed]
  42. Liang, J.; Kou, S.; Chen, C.; Raza, S.H.A.; Wang, S.; Ma, X.; Zhang, W.-J.; Nie, C. Effects of Clostridium butyricum on growth performance, metabonomics and intestinal microbial differences of weaned piglets. BMC Microbiol. 2021, 21, 85–101. [Google Scholar] [CrossRef]
  43. Zhang, J.; Chen, X.; Liu, P.; Zhao, J.; Sun, J.; Guan, W.; Johnston, L.J.; Levesque, C.L.; Fan, P.; He, T.; et al. Dietary Clostridium butyricum Induces a Phased Shift in Fecal Microbiota Structure and Increases the Acetic Acid-Producing Bacteria in a Weaned Piglet Model. J. Agric. Food Chem. 2018, 66, 5157–5166. [Google Scholar] [CrossRef] [PubMed]
  44. Yang, Y.; Liu, Y.; Liu, J.; Wang, H.; Guo, Y.; Du, M.; Cai, C.; Zhao, Y.; Lu, C.; Guo, X.; et al. Composition of the Fecal Microbiota of Piglets at Various Growth Stages. Front. Vet. Sci. 2021, 8, 661671. [Google Scholar] [CrossRef] [PubMed]
  45. Hossain, M.; Begum, M.; Kim, I. Effect of Bacillus subtilis, Clostridium butyricum and Lactobacillus acidophilus endospores on growth performance, nutrient digestibility, meat quality, relative organ weight, microbial shedding and excreta noxious gas emission in broilers. Vet. Med. 2015, 60, 77–86. [Google Scholar] [CrossRef]
  46. Wang, K.; Chen, G.; Cao, G.; Xu, Y.; Wang, Y.; Yang, C. Effects of Clostridium butyricum and Enterococcus faecalis on growth performance, intestinal structure, and inflammation in lipopolysaccharide-challenged weaned piglets. J. Anim. Sci. 2019, 97, 4140–4151. [Google Scholar] [CrossRef]
  47. Vacca, M.; Celano, G.; Calabrese, F.M.; Portincasa, P.; Gobbetti, M.; De Angelis, M. The Controversial Role of Human Gut Lachnospiraceae. Microorganisms 2020, 8, 573. [Google Scholar] [CrossRef]
  48. Chen, L.; Wilson, J.E.; Koenigsknecht, M.J.; Chou, W.-C.; Montgomery, S.A.; Truax, A.D.; Brickey, W.J.; Packey, C.D.; Maharshak, N.; Matsushima, G.K.; et al. NLRP12 attenuates colon inflammation by maintaining colonic microbial diversity and promoting protective commensal bacterial growth. Nat. Immunol. 2017, 18, 541–551. [Google Scholar] [CrossRef]
  49. Counotte, G.H.; Prins, R.A.; Janssen, R.H.; Debie, M.J. Role of Megasphaera elsdenii in the Fermentation of dl-[2-C]lactate in the Rumen of Dairy Cattle. Appl. Environ. Microbiol. 1981, 42, 649–655. [Google Scholar] [CrossRef]
  50. Yohe, T.T.; Enger, B.D.; Wang, L.; Tucker, H.L.M.; Ceh, C.A.; Parsons, C.L.M.; Yu, Z.; Daniels, K.M. Short communication: Does early-life administration of a Megasphaera elsdenii probiotic affect long-term establishment of the organism in the rumen and alter rumen metabolism in the dairy calf? J. Dairy Sci. 2018, 101, 1747–1751. [Google Scholar] [CrossRef]
  51. Yu, G.Q.; Xu, G.H. The influence of N-methyl-D, L-aspartic acid on the growth performance of fattening pigs. Pig Breed. 1999, 1, 7–9. [Google Scholar]
  52. Liu, S.Q.; Geng, N.; Sun, X.X.; Yuan, Q.P. Research Progress on Metabolic Engineering of Biosynthesis of Adipic Acid. J. Food Saf. Qual. Test. 2016, 7, 3872–3878. [Google Scholar]
Figure 1. Venn diagram, rank–abundance and rarefaction curves of fecal microorganism in weaned piglets in different treatment groups. (A) Venn diagram; (B) rank–abundance curve; (C) rarefaction curve.
Figure 1. Venn diagram, rank–abundance and rarefaction curves of fecal microorganism in weaned piglets in different treatment groups. (A) Venn diagram; (B) rank–abundance curve; (C) rarefaction curve.
Animals 14 02287 g001
Figure 2. Non-metric multidimensional scaling analysis (A) and partial least-squares discriminant analysis (B) at the OTU level for three treatment groups.
Figure 2. Non-metric multidimensional scaling analysis (A) and partial least-squares discriminant analysis (B) at the OTU level for three treatment groups.
Animals 14 02287 g002
Figure 3. Classification of the bacterial community composition across the three different treatment groups. (A) Relative abundance of bacterial phylum level and (B) relative abundance of bacterial genus level.
Figure 3. Classification of the bacterial community composition across the three different treatment groups. (A) Relative abundance of bacterial phylum level and (B) relative abundance of bacterial genus level.
Animals 14 02287 g003
Figure 4. Heatmap showing the most relative abundance of dominant bacterial OTUs. Note: the relative values are indicated by color intensity, with the legend indicated at the right corner.
Figure 4. Heatmap showing the most relative abundance of dominant bacterial OTUs. Note: the relative values are indicated by color intensity, with the legend indicated at the right corner.
Animals 14 02287 g004
Figure 5. Two-dimensional score plots of fecal metabolites in three different treatment groups. (A,B) partial least-squares discriminant analysis (PLS-DA) in positive/negative ion modes, (C,D) PLS-DA replacement test in positive/negative ion mode, and (E,F) orthogonal partial least-squares discriminant analysis (OPLS-DA) in positive/negative ion mode (n = 6).
Figure 5. Two-dimensional score plots of fecal metabolites in three different treatment groups. (A,B) partial least-squares discriminant analysis (PLS-DA) in positive/negative ion modes, (C,D) PLS-DA replacement test in positive/negative ion mode, and (E,F) orthogonal partial least-squares discriminant analysis (OPLS-DA) in positive/negative ion mode (n = 6).
Animals 14 02287 g005
Figure 6. Pathway enrichment map analysis of differential metabolites in feces between (AC) using MetaboAnalystR 3.0. Note: the color of the circles from white to yellow to red denotes incremental fold change (−log(p)). The size of the circles from small to large indicates an increment of the impact of the pathway.
Figure 6. Pathway enrichment map analysis of differential metabolites in feces between (AC) using MetaboAnalystR 3.0. Note: the color of the circles from white to yellow to red denotes incremental fold change (−log(p)). The size of the circles from small to large indicates an increment of the impact of the pathway.
Animals 14 02287 g006
Figure 7. Correlation between the body weight, diarrhea incidence, differential microbiota (at the genera level), and metabolites. (A) Correlation between body weight, diarrhea incidence, and microbiota. (B) Correlation between body weight, diarrhea incidence, and metabolites. (C) Correlation between microbiota and metabolites. Note: the strength (Spearman’s ρ value) and significance of correlations are shown as color in shades (red, positive correlation; blue, negative correlation). The values above/below zero represent positive/negative correlations. Significant correlations are noted by * p ≤ 0.05, ** p ≤ 0.01, *** p ≤ 0.001.
Figure 7. Correlation between the body weight, diarrhea incidence, differential microbiota (at the genera level), and metabolites. (A) Correlation between body weight, diarrhea incidence, and microbiota. (B) Correlation between body weight, diarrhea incidence, and metabolites. (C) Correlation between microbiota and metabolites. Note: the strength (Spearman’s ρ value) and significance of correlations are shown as color in shades (red, positive correlation; blue, negative correlation). The values above/below zero represent positive/negative correlations. Significant correlations are noted by * p ≤ 0.05, ** p ≤ 0.01, *** p ≤ 0.001.
Animals 14 02287 g007
Table 1. Composition and nutrient levels of control diet (air-dried basis, %).
Table 1. Composition and nutrient levels of control diet (air-dried basis, %).
IngredientsContentNutrient Levels 2Content
Corn58.60Digestible energy (MJ/kg)13.68
Soybean meal17.50
Expanded soybean7.50Crude protein20.40
Milk powder4.00Lys1.30
Fish meal4.00Met + Cys0.78
Whey powder4.30Thr0.87
NaCl0.30Na0.27
Limestone1.22Ca0.95
CaHPO41.16Available P0.39
DL-met0.07
Lys HCL0.35
Premix 11.00
Total100.00
1 The premix provided the per kg of diets as follows: VA 8 000 IU, VB1 4 mg, VB2 3.6 mg, VB5 40 mg, VB6 4 mg, VB12 0.02 mg, VD3 3 000 IU, VE 20 IU, VK3 2 mg, biotin 0.15, folic acid 1 mg, D-pantothenic acid 11 mg, nicotinic acid 10 mg, antioxidant 100 mg, Cu (as copper sulfate) 10 mg, Fe (as ferrous sulfate) 80 mg, Mn (as manganese sulfate) 30 mg, Zn (as zinc sulfate) 75 mg, I (as potassium iodide) 0.4 mg, and Se (as sodium selenite) 0.3 mg. 2 Digestible energy and Available P were calculated values, while the others were measured values.
Table 2. Effects of C. butyricum supplementation on growth performance and diarrhea of weaned piglets challenged with Escherichia coli K88.
Table 2. Effects of C. butyricum supplementation on growth performance and diarrhea of weaned piglets challenged with Escherichia coli K88.
Treatments 2
Item 1Group IGroup IIGroup IIIp-Value
Initial weight (kg)6.91 ± 0.626.90 ± 0.286.96 ± 0.590.968
Final weight (kg)14.65 ± 2.2814.11 ± 1.2513.94 ± 1.750.735
d 0–7
ADG (g)349.11 ± 34.03347.96 ± 46.60366.33 ± 53.570.696
ADFI (g)505.88 ± 39.58461.86 ± 66.17477.43 ± 21.420.194
F/G1.46 ± 0.141.33 ± 0.121.33 ± 0.190.185
Diarrhea incidence (%)1.791.791.791.000
d 7–14
ADG (g)386.61 ± 60.66 a321.43 ± 43.45 b337.76 ± 34.43 ab0.042
ADFI (g)497.38 ± 30.97 a420.14 ± 47.57 b439.14 ± 58.22 b0.011
F/G1.32 ± 0.221.32 ± 0.191.31 ± 0.190.992
Diarrhea incidence (%)000-
d 14–21
ADG (g)370.54 ± 86.09360.20 ± 63.33291.84 ± 59.250.101
ADFI (g)584.63 ± 94.54 ab599.29 ± 83.80 a494.57 ± 93.23 b0.088
F/G1.60 ± 0.141.68 ± 0.131.70 ± 0.090.312
Diarrhea incidence (%)3.57 ab10.71 a1.79 b0.065
d 0–21
ADG (g)368.75 ± 41.58343.20 ± 21.57331.97 ± 32.380.116
ADFI (g)529.29 ± 45.32 a493.76 ± 37.58 ab470.38 ± 31.56 b0.027
F/G1.46 ± 0.111.44 ± 0.101.45 ± 0.120.955
Diarrhea incidence (%)1.79 ab4.17 a1.19 b0.075
1 ADG, average daily gain; ADFI, average daily feed intake; F/G, feed-to-gain ratio. 2 Piglets were fed different diets. Group I, piglets fed the control diet + normal saline; group II, piglets fed the control diet + Escherichia coli K88; group III, piglets fed the control diet supplemented with C. butyricum + Escherichia coli K88. a, b Means in the same row with different superscript letters differ significantly (p < 0.05) (n = 8).
Table 3. Alpha diversity indices of rectal bacterial communities in piglets.
Table 3. Alpha diversity indices of rectal bacterial communities in piglets.
Treatments
ItemGroup IGroup IIGroup IIIp-Value
Sobs378.33 ± 8.08 a310.00 ± 26.87 b382.00 ± 27.22 a0.028
Coverage, %99.88 ± 0.0499.85 ± 0.0399.90 ± 0.010.320
Shannon4.08 ± 0.34 a3.39 ± 0.22 b3.83 ± 0.20 ab0.090
ACE404.56 ± 16.93355.59 ± 43.60407.24 ± 24.150.167
Chao407.27 ± 11.47366.23 ± 42.23410.28 ± 21.170.194
a, b Means in the same row with different superscript letters differ significantly (p < 0.05) (n = 3).
Table 4. Different endogenous metabolites in feces of weaned piglets in group I, group II, and group III.
Table 4. Different endogenous metabolites in feces of weaned piglets in group I, group II, and group III.
Metabolite Namesm/z 1Retention Time 2 (min)Group I vs. Group IIGroup I vs. Group IIIGroup II vs. Group IIIMain Functions
log2(FC) 3p-Value 4Change 5log2(FC)p-ValueChangelog2(FC)p-ValueChange
L-Aspartic acid134.0493.54−1.238<0.0011.364<0.001ABC transporters, biosynthesis of alkaloids derived from ornithine, lysine and nicotinic acid
(5-hydroxyindol-3-YL) acetic acid192.06421.81−1.416<0.0011.310<0.001Phenylalanine metabolism
L-Phenylalanyl-L-Proline263.14375.671.1190.001ABC transporters
2-Keto-3-Deoxy-D-Gluconic acid177.04100.441.4660.003Glucose metabolism
Octadecanoic acid282.94402.91−1.2760.004−0.9680.006Lipid metabolism
CMP324.2167.701.1970.005Pyrimidine metabolism
Adipic acid145.05322.59−0.6220.003Lysine biosynthesis and degradation
1 m/z, mass-to-charge ratio; 2 RT, retention time; 3 FC, fold change; 4 p-value, a significant difference p value, FDR < 0.05; 5 “↑/↓” indicate the increase/decrease of the metabolite level; “—” means it does not exist (n = 6).
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Liang, J.; Wang, S.; Kou, S.; Chen, C.; Zhang, W.; Nie, C. Clostridium butyricum Prevents Diarrhea Incidence in Weaned Piglets Induced by Escherichia coli K88 through Rectal Bacteria–Host Metabolic Cross-Talk. Animals 2024, 14, 2287. https://doi.org/10.3390/ani14162287

AMA Style

Liang J, Wang S, Kou S, Chen C, Zhang W, Nie C. Clostridium butyricum Prevents Diarrhea Incidence in Weaned Piglets Induced by Escherichia coli K88 through Rectal Bacteria–Host Metabolic Cross-Talk. Animals. 2024; 14(16):2287. https://doi.org/10.3390/ani14162287

Chicago/Turabian Style

Liang, Jing, Sihu Wang, Shasha Kou, Cheng Chen, Wenju Zhang, and Cunxi Nie. 2024. "Clostridium butyricum Prevents Diarrhea Incidence in Weaned Piglets Induced by Escherichia coli K88 through Rectal Bacteria–Host Metabolic Cross-Talk" Animals 14, no. 16: 2287. https://doi.org/10.3390/ani14162287

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop