Skip to main content

Dietary sodium acetate and sodium butyrate improve high-carbohydrate diet utilization by regulating gut microbiota, liver lipid metabolism, oxidative stress, and inflammation in largemouth bass (Micropterus salmoides)

Abstract

Background

Adequate level of carbohydrates in aquafeeds help to conserve protein and reduce cost. However, studies have indicated that high-carbohydrate (HC) diet disrupt the homeostasis of the gut–liver axis in largemouth bass, resulting in decreased intestinal acetate and butyrate level.

Method

Herein, we had concepted a set of feeding experiment to assess the effects of dietary sodium acetate (SA) and sodium butyrate (SB) on liver health and the intestinal microbiota in largemouth bass fed an HC diet. The experimental design comprised 5 isonitrogenous and isolipidic diets, including LC (9% starch), HC (18% starch), HCSA (18% starch; 2 g/kg SA), HCSB (18% starch; 2 g/kg SB), and HCSASB (18% starch; 1 g/kg SA + 1 g/kg SB). Juvenile largemouth bass with an initial body weight of 7.00 ± 0.20 g were fed on these diets for 56 d.

Results

We found that dietary SA and SB reduced hepatic triglyceride accumulation by activating autophagy (ATG101, LC3B and TFEB), promoting lipolysis (CPT1α, HSL and AMPKα), and inhibiting adipogenesis (FAS, ACCA, SCD1 and PPARγ). In addition, SA and SB decreased oxidative stress in the liver (CAT, GPX1α and SOD1) by activating the Keap1-Nrf2 pathway. Meanwhile, SA and SB alleviated HC-induced inflammation by downregulating the expression of pro-inflammatory factors (IL-1β, COX2 and Hepcidin1) through the NF-κB pathway. Importantly, SA and SB increased the abundance of bacteria that produced acetic acid and butyrate (Clostridium_sensu_stricto_1). Combined with the KEGG analysis, the results showed that SA and SB enriched carbohydrate metabolism and amino acid metabolism pathways, thereby improving the utilization of carbohydrates. Pearson correlation analysis indicated that growth performance was closely related to hepatic lipid deposition, autophagy, antioxidant capacity, inflammation, and intestinal microbial composition.

Conclusions

In conclusion, dietary SA and SB can reduce hepatic lipid deposition; and alleviate oxidative stress and inflammation in largemouth bass fed on HC diet. These beneficial effects may be due to the altered composition of the gut microbiota caused by SA and SB. The improvement effects of SB were stronger than those associated with SA.

Introduction

With the rapid development of intensive aquaculture mode, high-energy and low-protein feeds has become important factor to consider in sustainable and cost-effective fish farming [1]. As high-energy and low-cost substance, carbohydrates have been added to aquatic feeds to reduce cost [2]. However, due to physiological limitations, carnivorous fish generally have a low utilization rate of carbohydrates [3]. A high-carbohydrate (HC) diet can result in the disruption of fish intestinal flora and induce excessive lipid deposition in the liver, which can result in fatty liver [4, 5], a syndrome that is accompanied by oxidative stress and inflammation [6, 7]. The use of feed additives was one of the effective measures to mitigate the negative effects of the HC diet. For example, dietary betaine mitigated HC diet-induced hepatic lipid accumulation in blunt-snout bream by improving the expression of trimethylamine formation-associated microbial genes and bacterial taxa [8]. Clostridium butyricum cultures improved carbohydrate utilization, antioxidant capacity, and gut microbiota in largemouth bass fed an HC diet [9]. Similarly, studies in Nile tilapia found that insulin and Bacillus amyloliquefaciens ameliorated HC diet-induced metabolic syndrome by altering gut bacterial composition and enriching acetate-producing bacteria [10, 11]. Thus, utilizing gut microbes as a possible target to mitigate the detrimental effects of HC diets on fish may be a key issue for the continuous development and sustainable aquaculture approach.

The role of gut microbes in the formation of fatty liver has become a research focus on fish nutrition. The regulation of gut microbes and their metabolites is a novel method for the treatment of fatty liver, but the mechanism behind this is not fully understood [12]. Short-chain fatty acids (SCFAs) are generated by intestinal microorganisms through anaerobic fermentation [13]. Many studies have shown that SCFAs play important roles in alleviating liver steatosis, inflammatory damage, and oxidative stress [14,15,16]. For instance, SCFAs can regulate the TLR-mediated NF-κB cascade by promoting the secretion of its associated pro-inflammatory/anti-inflammatory cytokines, thereby modulating the immune response during inflammation [17].

Two important SCFAs, sodium acetate (SA) and sodium butyrate (SB), are noteworthy in this regard [18]. In mammals, both SA and SB can effectively reduce hepatic lipid deposition by inhibiting lipogenesis and promoting fatty acid β-oxidation, and they also participate in the regulation of hepatic lipid metabolism as regulatory factors [19,20,21]. In addition, SA and SB can restore mitochondrial respiratory function, resulting in enhancing antioxidant response of hepatocytes [22]. Thereby, SA and SB may be involved in mitigating the negative effects of HC on fish. Indeed, several relevant studies have examined the roles of SA and SB in nutrient metabolism, immune regulation, and the alteration of the intestinal microbial composition in aquatic animals. For example, studies in Nile tilapia found that the addition of 1.8 g/kg SA alleviated HC-induced metabolic disorders and intestinal inflammation [23], and reduced high-fat diet-induced hepatic lipid deposition and oxidative stress [24]. SA at 2 g/kg ameliorated HC diet-induced liver injury in eels [25]. Similarly, supplementation with 2 g/kg of SB reduced liver lipid deposition and improved the intestinal flora in largemouth bass [26]. Addition of 2 g/kg SB to the diet improved gut microbiota and alleviated inflammation in largemouth bass fed with high soybean meal diet [27]. These studies suggest that SA and SB are essential for metabolic homeostasis and physiological regulation. Therefore, SA and SB may be potential supplements for improving liver health and alleviating metabolic disorders induced by HC diets in fish. Similarly, the concentrations of SA and SB were set at 2 g/kg in this study. Interestingly, previous studies have demonstrated that HC diets can cause an intestinal microbial imbalance in largemouth bass, thereby reduce intestinal SA and SB levels [28]. However, whether the exogenous addition of SA and SB can mitigate the liver damage caused by excess carbohydrates in largemouth bass requires further investigation.

The largemouth bass is a typical carnivorous commercial fish. Numerous studies have identified appropriate nutrient requirements for juvenile largemouth bass. For example, the appropriate protein and lipid requirements for juvenile largemouth bass are 45.3% and 10%, respectively [29, 30]. Our research has shown that the appropriate carbohydrate requirement for juvenile largemouth bass is 9%. HC diets (18% starch) will lead to liver metabolite conversion, lipid deposition, oxidative stress, and disruption of gut flora structure in juvenile largemouth bass [31, 32]. Therefore, the goal of this study was to determine whether the exogenous addition of SA and SB could mitigate the damaging impact of carbohydrates on liver lipid deposition, oxidative stress, inflammatory, and the intestinal flora in largemouth bass. An additional goal was to explore the possible mechanisms behind this.

Materials and methods

Experimental diets

Five isonitrogenous (44% crude proteins) and isolipidic (10% crude lipids) experimental groups were as: group LC (9% starch), group HC (18% starch), group HCSA (18% starch; 2 g/kg SA), group HCSB (18% starch; 2 g/kg SB), and group HCSASB (18% starch; 1 g/kg SA + 1 g/kg SB). Carbohydrate levels (9% and 18%) and feed formulations were derived from our previous studies [33]. The added levels of SA and SB were based on previous studies where 2 g/kg of SA and SB were effective in improving liver and intestinal health in fish (Additional file 1: Table S1) [23,24,25,26,27, 34]. According to the principle of step-by-step enlargement, all feed ingredients were thoroughly mixed by hand, and soybean oil and distilled water were added sequentially. Finally, 3 mm and 4 mm sinking pellets were made by pelletizing machine [33]. The made pellet feed was placed in a cool and ventilated place for natural air drying and stored at −20 ºC. Detailed feed ingredient composition for each group is shown in Table 1. Dietary chemical composition was measured using standard method according to Baur and Ensminger [35].

Table 1 Formulation and chemical composition of the diets1, % dry matter

Fish preparation and feeding trial

Juvenile largemouth bass were acquired from Qionglai Hongbo Agricultural Co., Ltd. (Chengdu, China). The fish were acclimatized in the laboratory for two weeks prior to the feeding trial, and fed on the LC diet during acclimatization. The fish (initial weight: 7.00 ± 0.20 g) were randomly assigned to 15 tanks (5 groups, 3 replicates per group, 35 fish per replication). The tanks were 1 m in diameter with 300 L tap water after aeration. The fish were fed twice a day by satiation feeding for 8 weeks. Water temperature, DO, pH, and NH4+-N levels were kept at 25.0 ± 2 °C, 7.1 ± 0.5 mg/L, 6.7 ± 0.5, and < 0.15 mg/L, respectively, during feeding. Water temperature, DO, and pH were measured daily with a water quality monitor (AZ86031, Dongguan Hengxin Instrument Co., Ltd., China), and NH4+-N levels were measured weekly using Nesser's reagent spectrophotometry.

Sample collection

The fish were fasted for 24 h and anesthetized with 0.1 g/L MS-222 (Sigma, USA) prior to sample. After the fish were dissected, the liver was removed, and a portion of it was fixed in 4% paraformaldehyde for sectioning (n = 3); the remaining portion was placed in a sterile tube and stored at −80 °C for subsequent Western blot, qPCR, and enzyme analyses. Intestinal contents were collected for microbiome analysis (n = 3).

Tissue sectioning and staining

Liver tissues fixed in paraformaldehyde were dehydrated in ethanol (50%–100%), extracted with xylene, and embedded in paraffin. The samples were then cut into 5 µm-thick slices. The slices were partially stained with hematoxylin and eosin and partially stained with Oil red O. Photographs were taken using a digital slicing scanner and then quantized with Image J.

Liver biochemical index analysis

Liver catalase (CAT, A007-1-1), total superoxide dismutase (T-SOD, A001-1-2), malondialdehyde (MDA, A003-1-2), glutathione peroxidase (GSH-PX, A005-1-2), triglyceride (TG, A110-1-1), and total cholesterol (T-CHO, A111-1-1) levels were measured following the instructions of commercially available kits (Nanjing Jiancheng Bioengineering Institute, China). The detection limits of these kits were 0.2 U/mL, 5.0 U/mL, 0.2 nmol/mL, 20 U/mL, 0.3 mmol/L and 0.1 mmol/L, respectively.

Quantitative real-time PCR

Total RNA extraction and cDNA synthesis were performed following the instructions of an Animal Total RNA Isolation kit (Cat. No. RE-03014, Foregene, Chengdu, China) and RT Easy™ I (Cat. No. RT-01032, Foregene), respectively. The primer sequences were designed based on our previous genome and were listed in Table 2. The 2−ΔΔCT technique was used to evaluate gene expression levels [37], with β-actin as the reference gene.

Table 2 Primer sequences used for real-time PCR analysis

Western blot

The liver tissue was lysed in protein lysis solution and then centrifuged at 12,000 × g for 20 min to obtain the supernatant. The target proteins were transferred to precast gels for electrophoresis for 50 min and then transferred onto PVDF membranes. The PVDF membrane was placed in bovine serum protein and soaked for 2 h. The PVDF membrane was removed, washed with TBST to remove the primary antibody, transferred to the secondary antibody culture, incubated for 1 h, and washed again with TBST. Finally, the chromogenic solution was added to the PVDF membrane and images were captured. Quantification of the target protein was performed using Image J. The antibodies used in this study were listed in Table S2.

Intestinal microbiota analysis

After genomic DNA was obtained from the samples, the V3 + V4 region of 16S rDNA was amplified with specific primers. Sequencing libraries were constructed from the amplicons and sequenced on an Illumina platform. Once the raw reads were acquired, we separated the double-ended reads into tags, filtered the tags to produce clean tag, and then eliminated any low-quality or biologically irrelevant data. After that, clustering was done using the clean tag, and chimeric tags were eliminated to get the effective tag at the end. Following the acquisition of OTUs, OTU abundance data were calculated using the effective tag. The analytical process ordered the following steps: community function prediction, indicator species analysis, α-diversity analysis, β-diversity analysis, and species composition sub-licensing. The data were analyzed via the Omicsmart cloud platform (https://www.omicsmart.com).

Statistical analysis

The results were expressed as Mean ± SEM. Significant difference between the LC and HC groups were analyzed using t-test, and differences among the HC, HCSA, HCSB, and HCSASB groups were analyzed by one-way ANOVA. An asterisk (*) indicates a significant difference between the LC and HC group (P < 0.05). Means without a common superscript differ significantly (P < 0.05). All data were analyzed with GraphPad Prism 9.1.

Results

Growth performance

This study employed the same growth experiment as in our previous work on largemouth bass [38]. After 8 weeks of the feeding trial, FBW, WGR and SGR were significantly lower in the HC group than in the LC group (P < 0.05) (Fig. 1A–C). At 6th week, FBW, WGR and SGR in HCSB and HCSASB groups were significantly higher than those in HC group (P < 0.05) (Fig. 1A–C). At 8th week, FBW in HCSA, HCSB and HCSASB groups were significantly higher than that in HC group, and WGR and SGR in HCSA and HCSASB groups were significantly higher than that in HC group (P < 0.05) (Fig. 1A–C). There was no significant difference in SR among the groups in this study (P > 0.05) (Fig. 1D).

Fig. 1
figure 1

Effects of sodium acetate and sodium butyrate on growth performance of largemouth bass. A Final body weight (FBW). B Weight gain rate (WGR). C Specific growth rate (SGR). D Survival rate (SR). WGR = 100 × (final body weight − initial body weight)/initial body weight; SGR = 100 × [ln(final body weight) − ln(initial body weight)]/breeding days. Values were expressed as mean ± SEM (n = 3). a,bDifferent lowercase letters indicate significant differences (P < 0.05). *P < 0.05

Hepatic lipid deposition

The livers of the LC group appeared reddish or dark red, while those of the HC group were milky white, and those of the HCSA, HCSB, and HCSASB groups were light red with improved apparent liver color (Fig. 2A). Hematoxylin and eosin sections revealed that compared with the LC group, the livers of the HC group demonstrated severe fatty vacuolation, and most of the nuclei were displaced. However, liver fatty vacuolation was attenuated in the HCSA, HCSB and HCSASB groups (Fig. 2A). Both SA and SB attenuated HC-induced hepatic lipid deposition using Oil red O staining. (Fig. 2A and B). The biochemical indicators shown that SA and SB significantly reduced the liver triglyceride (TG) content (P < 0.05) (Fig. 2C), while the total cholesterol (T-CHO) content did not differ among groups (P > 0.05) (Fig. 2D). These results suggested that SA and SB could reduce HC-induced hepatic lipid deposition in largemouth bass.

Fig. 2
figure 2

Effects of sodium acetate and sodium butyrate on the liver lipid metabolism of largemouth bass. A Liver morphology; HE staining of liver (scale bars, 500 μm); Oil- red O staining of liver (scale bars, 500 μm). B Relative Oil- red O area in liver. C Hepatic TG levels. D Hepatic T-CHO levels. E Expression levels of lipogenesis-related genes. F Expression levels of lipolysis-related genes. and H Protein expression of PPARγ and AMPKα. Values were expressed as mean ± SEM (n = 6). a–cDifferent lowercase letters indicate significant differences (P < 0.05). *P < 0.05

To further investigate the mechanism by which SA and SB reduced hepatic lipid deposition, we examined the expression levels of genes involved in lipid metabolism. Compared with the LC group, the expression levels of lipid synthesis-related genes (ACCA, FAS, SCD1, and PPARγ) in the HC group were significantly increased (P < 0.05) (Fig. 2E), whereas the expression levels of SCD1 and PPARγ were significantly depressed in the HCSA, HCSB, and HCSASB groups compared to the HC group (P < 0.05) (Fig. 2E). The expression level of ACCA in the HCSB and HCSASB groups was noticeably decreased compared to the HC group, while the expression level of FAS in the HCSA and HCSB groups was significantly decreased compared to the HC group (P < 0.05) (Fig. 2E). Likewise, the levels of key lipogenesis PPARγ proteins were strongly upregulated in the HC group (P < 0.05), while dietary SA and SB strongly inhibited their expression (P < 0.05) (Fig. 2G and H). Furthermore, the mRNA levels of lipolysis-associated genes (CPTα, HSL, and AMPKα) were significantly decreased in the HC group compared to the LC group (P < 0.05) (Fig. 2F). CPT1α in the HCSA and HCSASB groups was significantly lower than that in the HC group, and AMPKα was considerably reduced in the HCSB group compared to the LC group (P < 0.05) (Fig. 2F). Importantly, dietary SA and SB had markedly elevated levels of AMPKα protein expression (P < 0.05) (Fig. 2G and H).

Hepatic autophagy

Compared with the LC group, the expression levels of autophagosome membrane initiation-related genes (Beclin1, ATG1, ATG101, and ATG13) were significantly decreased in the HC group (P < 0.05), whereas the expression levels of ATG101 and ATG13 were significantly higher in the HCSA, HCSB, and HCSASB groups than in the HC group (P < 0.05) (Fig. 3A). The expression levels of autophagosome membrane expansion-related genes (ATG4b, ATG5, ATG16, and LC3B) in the HC group were significantly decreased compared to the LC group (P < 0.05), while the expression levels of ATG4b, ATG5, ATG16, and LC3B in the HCSA and HCSB groups were markedly increased compared to those of the HC group (P < 0.05) (Fig. 3B). The HC group had significantly lower expression levels of autophagosome-lysosome fusion-related genes (mcoln2, TFEB, and Lamp2) compared to the LC group (P < 0.05); following SA and SB therapy, these gene expression levels were considerably increased (P < 0.05) (Fig. 3C). Furthermore, SA and SB significantly increased the expression levels of key autophagy-related proteins (LC3B, ATG5, and Lamp) (P < 0.05) (Fig. 3D and E). The results demonstrated that HC inhibited autophagy activity, but SA and SB enhanced autophagy activity, thereby improving lipid metabolism.

Fig. 3
figure 3

Effects of dietary sodium acetate and sodium butyrate on liver autophagy in largemouth bass. A Expression levels of autophagosome membrane initiation-related genes. B Expression levels of autophagosome membrane expansion-related genes. C Expression levels of autophagosomal lysosome fusion-related genes. DE Expression levels of autophagy-related proteins. Values were expressed as mean ± SEM (n = 6). a–cDifferent lowercase letters indicate significant differences (P < 0.05). *P < 0.05

Hepatic oxidative stress

The MDA content in the HC group was significantly higher than in the LC group (P < 0.05) (Fig. 4C), although the GSH-Px activity was significantly lower (P < 0.05) (Fig. 4D). However, GSH-Px activity was dramatically boosted, while the MDA content was significantly lowered by SA and SB (P < 0.05). The activities of CAT and T-SOD were not significantly different (P > 0.05) (Fig. 4A and B). Meanwhile, the expression levels of antioxidant genes (CAT, GPX1α, and SOD1) were significantly lower in the HC than in the LC group (P < 0.05) (Fig. 4E). However, SA and SB significantly enhanced the transcription levels of CAT and GPX1α (P < 0.05) (Fig. 4E). Keap1-Nrf2 was the main regulator of antioxidant reactions; SA and SB significantly inhibited the protein expression level of Keap1 (P < 0.05) but increased the protein expression level of Nrf2 (P < 0.05) (Fig. 4F and G). Therefore, these findings revealed that SA and SB could boost antioxidant capacity in largemouth bass fed on an HC diet via activating the Keap1-Nrf2 pathway.

Fig. 4
figure 4

Effects of sodium acetate and sodium butyrate on the liver antioxidant capacity of largemouth bass. A Activities of catalase (CAT) in the liver. B Activities of total superoxide dismutase (T-SOD) in the liver. C Contents of malondialdehyde (MDA) in the liver. D Activities of glutathione peroxidase (GSH-Px) in the liver. E Expression levels of antioxidant-related genes. F and G Expression levels of antioxidant-related proteins. Values were expressed as mean ± SEM (n = 6). a–dDifferent lowercase letters indicate significant differences (P < 0.05). *P < 0.05

Hepatic inflammation response

The HC group had considerably higher expression levels of pro-inflammatory factors (IL-1β, COX2, Hepcidin1, and NF-κB) compared to the LC group (P < 0.05) (Fig. 5A). Compared to the HC group, the expression levels of COX2, Hepcidin1, and NF-κB were significantly lower in the HCSA, HCSB, and HCSASB groups (P < 0.05) (Fig. 5A). However, the expression level of the anti-inflammatory factor (IL-10) in the HCSA, HCSB, and HCSASB groups was considerably higher than in the HC group (P < 0.05) (Fig. 5A). Further studies showed that SA and SB significantly inhibited total NF-κB protein and phosphorylated NF-κB protein expression (P < 0.05) (Fig. 5B), indicating that SA and SB could suppress HC-induced inflammation.

Fig. 5
figure 5

Effects of sodium acetate and sodium butyrate on liver inflammation in largemouth bass. A Expression levels of inflammation-related genes. B Expression levels of inflammation-related proteins. Values were expressed as mean ± SEM (n = 6). a–cDifferent lowercase letters indicate significant differences (P < 0.05). *P < 0.05

Intestinal microbiome analysis

Diversity analysis

The α-diversity analysis of gut microbes was presented in Fig. 6A–D. Compared to the LC group, the Chao1, Sob, and ACE indices of the HC group were significantly decreased (P < 0.05), while those of the HCSA, HCSB, and HCSASB groups were increased, although the difference was not observed (P > 0.05). Only the HCSA group’s Simpson index significantly decreased in comparison to the HC group (P < 0.05).

Fig. 6
figure 6

Effects of sodium acetate and sodium butyrate on the gut microbial diversity of largemouth bass. A Chao1 index. B Sob index. C ACE index. D Simpson index. E UPGMA hierarchical clustering based on Bray distance analysis of intestinal microbial patterns at the genus level. E PCoA analysis of intestinal flora at the genus level. Data were expressed as mean ± SEM (n = 3). a–cDifferent lowercase letters indicate significant differences (P < 0.05)

The β-diversity analysis (Fig. 6E and F) and the UPGMA classification tree (Fig. 6E) showed that the HC group and the LC, HCSB, and HCSASB groups clustered in different branches, indicating that dietary SA and SB markedly changed the intestinal flora. However, the LC and HCSB groups were initially clustered together and then clustered with the HCSASB group, suggesting that the LC, HCSB, and HCSASB groups had high similarity. The PCoA (Fig. 6F) also showed that SA and SB altered the gut microbiome composition of largemouth bass fed on an HC diet.

Intestinal microbial composition

The abundance of intestinal flora at the phylum level (Fig. 7A) indicated that the dominant phyla in the LC group were Proteobacteria (59%) and Firmicutes (32%). Compared to the LC group, the abundance of Proteobacteria decreased (23%) in the HC group, while the abundance of Firmicutes (56%) and Fusobacteria (20%) increased. The dominant bacteria in the HCSA group were Firmicutes (64%) and Proteobacteria (33%). The dominant phyla in the HCSB group were Proteobacteria (56%), Fusobacteria (21%), and Firmicutes (20%). The dominant phyla in the HCSASB group were Firmicutes (68%), Proteobacteria (17%), and Fusobacteria (13%). These results suggested that dietary SA and SB altered the dominant gut microbiota at the phylum level in the largemouth bass fed an HC diet.

Fig. 7
figure 7

Effects of sodium acetate and sodium butyrate on the intestinal microbial composition of largemouth bass. A Gut flora abundance at the phylum level (Top 10). B Gut flora abundance at the genus level (Top 10). C Venn diagram of intestinal flora at the genus level. D Intestinal flora Indicator analysis at the genus level. E Significant changes in the relative abundance of intestinal microbiota observed at the genus level. Data were expressed as mean ± SEM (n = 3). a–cDifferent lowercase letters indicate significant differences (P < 0.05). *P < 0.05

Analysis of the gut flora at the genus level (Fig. 7B and E) indicated that the abundance of Clostridium_sensu_stricto_1 in the HC group was remarkably lower than in the LC group, and the abundances of Lactococcus and Cetobacterium were significantly higher than those in the LC group (P < 0.05). The HCSB and HCSASB groups demonstrated a significant decrease in Lactococcus abundance and an increase in Clostridium_sensu_stricto_1 abundance compared to the HC group (P < 0.05). The HCSA and HCSASB groups showed a significantly reduced abundance of Cetobacterium (P < 0.05). The Venn diagram (Fig. 7C) presented that the total effective intestinal flora comprised 393 genera, of which 131 genera were in the LC group. The HC group comprised 55 genera; there were 61 genera in the HCSA group, the HCSB group comprised 90 genera, and the HCSASB group comprised 56 genera. The five groups had 27 common genera. Indicator analysis was used to screen for marker species in each group (Fig. 7D). The results revealed that the top three biomarkers were Sediminibacterium, Mesorhizobium, and Ralstonia for the LC group. For the HC group, these were Lactococcus, Cetobacterium, and Plesiomonas. For the HCSA group, these were Lactococcus, Aurantimicrobium, and Bacillus. The indicators for the HCSB group were Cetobacterium, Aurantimicrobium, and Bacillus, and for the HCSASB group, these were Clostridium_sensu_stricto_1, Cetobacterium, and Mycoplasma.

Prediction of intestinal flora function

Using PICRUSt to predict intestinal flora functions, the KEGG pathway analysis showed six significantly different categories, namely, metabolism, genetic information processing, cellular processes, environmental information processing, organic systems, and human diseases, as shown in the results of predictive analysis of intestinal flora functions (Fig. 8A and B). Among these categories, carbohydrate metabolism was the most abundant, followed by cofactor and vitamin metabolism, amino acid metabolism, and lipid metabolism. Therefore, a specific analysis of the four metabolic pathways was performed, and the results (Fig. 8C) indicated that compared to the LC group, lipid metabolism was significantly increased in the HC group (P < 0.05), while carbohydrate metabolism, cofactor and vitamin metabolism, and amino acid metabolism had no significant differences (P > 0.05). Carbohydrate metabolism, cofactor and vitamin metabolism, and amino acid metabolism were higher in the HCSA and HCSB groups than in the HC group (P < 0.05).

Fig. 8
figure 8

Effects of sodium acetate and sodium butyrate on the intestinal microbial KEGG functions of largemouth bass. A Functional stack of intestinal flora. B Functional clustering heat map of intestinal flora. C Significantly different functions of intestinal flora. Data were expressed as mean ± SEM (n = 3). a–cDifferent lowercase letters indicate significant differences (P < 0.05). *P < 0.05

Correlation between growth performance, hepatic lipid deposition, autophagy, antioxidant capacity, inflammation, and gut microbial composition

The Pearson correlation analysis (Fig. 9) presented that growth performance was closely correlated with hepatic lipid deposition, autophagy, antioxidant capacity, inflammatory response, and intestinal microbial composition. WGR and SGR were significantly positively correlated with ATG101, ATG16, CAT, GPX, and IL-10 mRNA expression (P < 0.01), and were negatively correlated with TG contents, T-CHO contents, PPARγ, FAS, and Lactococcus abundance (P < 0.01).

Fig. 9
figure 9

Pearson correlations between growth performance, lipid deposition, autophagy, antioxidant capacity, inflammation, and intestinal flora composition. The correlation coefficients were expressed by color intensity, ranging from green to red. *P < 0.05, **P < 0.01

Discussion

SA and SB reduced hepatic lipid deposition by enhancing autophagy in largemouth bass fed an HC diet

Many reports have demonstrated that long-term consumption of an HC diet can cause fatty liver formation in farmed fish [8, 39, 40]. Often the formation of fatty liver can be explained by the “multiple blow model” [41]. The “first blow” is the excessive accumulation of TGs in the liver; as a result, reducing hepatic lipid deposition is the key to alleviating fatty liver. In this study, the appearance color of the liver in the HC group was milky white, implying that the HC diet caused excessive lipid deposition in the liver of largemouth bass. This was further confirmed by Oil red O staining, histological observation, and the measurement of TG content; the results were agreed with prior research [5, 42]. However, dietary sodium acetate (SA) and sodium butyrate (SB) alleviated hepatic lipid deposition in largemouth bass caused by HC feeding by inhibiting lipogenesis (FAS, ACCA, SCD1, and PPARγ) and activating the lipolysis pathway (CPT1α, HSL, and AMPKα). In mammals, SCFAs significantly reduce the production of fatty acid synthase in the liver by activating the UCP2-AMPK-ACCA pathway, thereby reducing lipid synthesis [43]. Moreover, SCFAs can also promote the expression of liver CPT1 promoter by inhibiting HDAC1 and enhancing promoter activity, thereby increasing CPT1 and allowing more fatty acids to enter the liver mitochondria for β-oxidation [44]. Interestingly, previous studies have shown that SA can ameliorate liver lipid deposition induced by a high-fat diet in Nile tilapia to a certain extent by activating lipid catabolism [24]. In addition, coated SB may accelerate lipid metabolism in laying hens by inhibiting liver lipogenesis enzyme activity (ACCA and FAS) and promoting liver lipolysis (AMPKα and CPT1) [45]. Therefore, SA and SB can alleviate hepatic lipid deposition in largemouth bass fed on an HC diet.

Previous research has demonstrated that autophagy is impaired in largemouth bass fed on an HC diet [33], in turn disturbing lipid metabolism [46, 47]; thus, effects of SA and SB on autophagy was investigated. Current study results have shown that SA and SB could activate autophagy, including the expression levels of autophagosome membrane initiation genes (Beclin1, ATG1, ATG101, and ATG13), autophagosome membrane expansion genes (ATG4b, ATG5, ATG16, and LC3B), and autophagosome lysosomal fusion genes (mcoln2, TFEB, and Lamp2). To date, there have been relatively few studies on the effects of SA and SB on liver autophagy in fish. However, a study in piglets has shown that SB induces autophagy by activating AMPK, thereby reducing H2O2-induced oxidative stress, intestinal epithelial barrier damage, and mitochondrial dysfunction [48]. Studies in mice have also shown that SB can slow the progression of osteoarthritis by restoring impaired autophagy and autophagic flux [49]. Therefore, we propose that this autophagy-promoting effect of SA and SB is maintained in the liver of HC-induced largemouth bass.

SA and SB alleviated HC-induced hepatic oxidative stress and inflammation through Keap1-Nrf2 and the NF-κB pathway

The “second hit” in the “multi-hit model” refers to increases in oxidative stress and inflammation and is usually caused by the “first hit,” the accumulation of TGs in the liver [50]. In this study, the HC diet decreased the activities of CAT and GSH-Px in the liver, while it increased the content of MDA. This suggested that largemouth bass fed on HC diet were more susceptible to sustained oxidative stress, consistent with previous studies in golden pomfret and eels [4, 6]. The Keap1-Nrf2 pathway can regulate oxidative stress in the body. When the body is subjected to oxidative stress, the Keap1-Nrf2 signaling pathway is triggered, allowing the nuclear transcription factor Nrf2 to enter the nucleus and activate the transcription of antioxidant genes to maintain oxidation-antioxidant physiological balance [51]. In this study, both SA and SB could reduce oxidative stress by inhibiting the expression of the Keap1 protein and activating the expression of the Nrf2 protein. In addition, SA and SB increased the activities of CAT, GSH-Px, and T-SOD and decreased the content of MDA, indicating that the antioxidant activity was enhanced and that oxidative stress was reduced. It has been reported that SA and SB can restore mitochondrial respiratory function and promote mitochondrial repair by inducing mitochondrial transition to the fusion process, thus enhancing the antioxidant response of hepatocytes [16, 22]. SB is the main energy source for the intestinal epithelium; its nutritional effects can reduce oxidative stress and liver damage by increasing the concentration of glutathione [52]. Similar studies have found that the addition of SA/SB improved oxidative stress in mice through the NLRP3/Caspase-1 signaling pathway [53]. In fish, SA and SB have also been found to alleviate liver oxidative stress in Nile tilapia and largemouth bass induced by a high-fat diet [24, 26]. In summary, SA and SB can alleviate HC-induced oxidative stress in largemouth bass through the Keap1-Nrf2 signaling pathway.

Significant accumulation of TGs in the liver can induce the secretion of pro-inflammatory factors and trigger inflammation in the body [54]. In this study, we found that the HC diet activated the expression of pro-inflammatory factors (IL-1β, COX2, and Hepcidin1) and inhibited the expression of anti-inflammatory factors (IL-10 and IKBα). Further research revealed that the HC diet increased the expression levels of total NF-κB protein and phosphorylated NF-κB protein. These results suggested that the HC diet induced an inflammatory response in largemouth bass. However, after treatment with SA and SB, the inflammatory response was significantly ameliorated. Recent studies have suggested that SA and SB may not only reduce the hepatitis response by improving the entero–liver axis, but also directly regulate hepatic immune cells, reduce the release of pro-inflammatory factors (IL-1β, TNF-α, and IL-1) in the liver, and increase the expression of anti-inflammatory factors (IL-4 and IL-10) [55, 56]. Similarly, dietary SA can upregulate immune-related genes (TNF-α, TGF-β, and IL-8) and improve disease resistance in common carp [57]. Dietary SB enhanced the expression of IL-1β, IL-8, and TNF-α in Dicentrarchus labrax, thereby improving anti-inflammatory function [58]. Therefore, SA and SB mitigated HC-induced inflammation through the NF-κB signaling pathway.

SA and SB improved the intestinal microbial composition of largemouth bass fed an HC diet

Gut microbes are involved in the regulation of host nutritional metabolism, immunity, and development [59]. SCFAs produced by gut microbes can ameliorate a fatty liver condition by maintaining intestinal flora homeostasis, protecting the intestinal barrier, reducing fat deposition, and inhibiting inflammation and oxidative stress [60, 61]. The α-diversity indices (Sob, Chao1, ACE, and Shannon) can be used to assess the diversity of gut microbes. In this study, an HC diet significantly reduced Chao1, Sob, and ACE indices, indicating that gut microbial diversity was severely impaired; which was consistent with previous studies on eels and largemouth bass [62, 63]. However, SA and SB increased the α-diversity of the gut microbiome and mitigated the decrease in gut microbiome diversity caused by the HC diet. A similar study found that SA could increase the α-diversity of the gut flora and promote gut health in Trachinotus ovatus [64]. SB also increased the intestinal microbial diversity of Pacific white shrimp [65]. In addition, β-diversity is a comparison of diversity between different ecosystems and is measured as the rate of change of species composition between communities. A UPGMA classification tree and principal coordinate analysis (PCoA) can be used to study the similarity between samples, where the more similar samples have shorter common branches; these closer distances are also reflected in the PCoA diagram. The UPGMA classification tree and the PCoA results showed that SA and SB partially restored the intestinal microbial β-diversity altered by the HC diet.

In addition, SA and SB affected the composition of the gut microbiota. The HC diet significantly reduced the abundance of Clostridium_sensu_stricto_1 and increased the abundance of Cetobacterium, which was consistent with studies for tilapia and gar [4, 10]. However, dietary SA and SB increased the abundance of Clostridium_sensu_stricto_1, and the abundance of Cetobacterium was significantly higher than that of the LC group. Studies have demonstrated that Clostridium_sensu_stricto_1 and Cetobacterium can increase intestinal SA and SB concentrations [66, 67]. SA and SB play important roles in reducing hepatic lipid deposition, enhancing antioxidant capacity, and alleviating the inflammatory response [68,69,70]. Moreover, dietary SA and SB reduced the increase in Lactococcus abundance caused by the HC diet. Lactococcus has negative effects on host health, including inflammatory responses and the slowing of growth [71, 72]. Previous studies have shown that SA improved the gut microbiota and promoted growth performance and health in zebrafish [73]. In addition, SB can ameliorate steatohepatitis in mice fed a high-fat diet by improving intestinal flora and the intestinal barrier [74]. In conclusion, hepatic steatosis, oxidative stress, and inflammation induced by HC diet were strongly associated with gut microbial dysbiosis in largemouth bass. Fortunately, SA and SB can improve the overall health of largemouth bass fed on HC diet by restoring intestinal microbiota homeostasis.

SA and SB enhanced metabolic pathways and improved carbohydrate tolerance in largemouth bass

In this study, the KEGG function prediction of intestinal flora showed that carbohydrate metabolism, cofactor and vitamin metabolism, and amino acid metabolism pathways were improved by SA and SB treatments, but the effect of SB was more pronounced. Studies have shown that SA and SB can be used as energy sources, providing carbon-containing precursor molecules for carbohydrate metabolism, amino acid metabolism, and cofactor metabolism and vitamin metabolism, so that they can also regulate cell metabolism by triggering signaling pathways [75]. Acetyl CoA is produced during the metabolic processing of SA and SB, and this can promote the entry of acetyl CoA into the tricarboxylic acid cycle and fat synthesis, or else promote carcass synthesis through the hydroxymethyl glutarate monoacyl CoA pathway, which promotes fat absorption and utilization [76]. In addition, SB can also accelerate the catabolic rate of sugars in animals by promoting the expression of transporter genes [77]. Recent studies have shown that dietary SA inhibited the catabolism of proteins, fats, and carbohydrates, but increased the energy supply for acetyl-CoA catabolism from dietary SA sources, ultimately increasing macronutrient deposition and promoting fish growth [78]. SB can improve the utilization of nucleotide derivatives and several common amino acids and thus increase the rate of weight gain [79]. Therefore, both SA and SB can promote the growth of largemouth bass, ameliorate the liver lipid deposition induced by excess carbohydrates, and increase the tolerance of largemouth bass to carbohydrates, possibly by improving the homeostasis and composition of the intestinal flora. In this regard, SB has an effect superior to that of SA.

Conclusions

In this study, dietary SA and SB reduced excessive lipid deposition in the liver of largemouth bass fed on HC diet by activating autophagy, thereby alleviating oxidative stress and inflammation. Additionally, it was found that SA and SB altered the composition of the gut microbiota and increased the bacterial abundance that produce acetic acid and butyrate; consequently, which may be responsible for mitigating HC-induced liver damage and improving carbohydrate utilization. In summary, SA and SB are effective feed additives that can protect the liver health of cultured species in aquaculture practices, and the effect of SB is better than SA.

Abbreviations

ACCA:

Acetyl-CoA carboxylase

ATG:

Autophagy-related gene

Beclin1:

Bcl2-associated X protein

CAT:

Catalase

COX2:

Cyclooxygenase 2

CPT1α:

Carnitine palmitoyl transterase-1 alpha

FAS:

Fatty acid synthase

GPX1α:

Glutathione peroxidase-1 alpha

HSL:

Hormone-sensitive lipase

IKBα:

Inhibitor kappa B alpha

IL-1β:

Interleukin-1β

IL-10:

Interleukin-10

Keap1:

Kelch like ECH-associated protein 1

LC3B:

Microtubule-associated protein 1 light-chain3B

Lamp2:

Lysosome-associated membrane protein 2

mcoln2:

Mucolipin TRP cation channel 2

NF-κB:

Nuclear factor kappa-B

Nrf2:

Nuclear factor erythroid 2-related factor 2

PPAR:

Peroxisome proliferator-activated receptor

SCD1:

Stearoyl-CoA desaturase-1

SCFAs:

Short-chain fatty acids

SOD1:

Superoxide dismutase 1

TFEB:

Transcription factor EB

References

  1. Zhang C, Rahimnejad S, Wang Y, Lu K, Song K, Wang L, et al. Substituting fish meal with soybean meal in diets for Japanese seabass (Lateolabrax japonicus): effects on growth, digestive enzymes activity, gut histology, and expression of gut inflammatory and transporter genes. Aquaculture. 2018;483:173–82. https://doi.org/10.1016/j.aquaculture.2017.10.029.

    Article  CAS  Google Scholar 

  2. Pauly D, Zeller D. Comments on FAO state of world fisheries and aquaculture (SOFIA 2016). Mar Policy. 2017;77:176–81. https://doi.org/10.1016/j.marpol.2017.01.006.

    Article  Google Scholar 

  3. Kamalam BS, Medale F, Panserat S. Utilisation of dietary carbohydrates in farmed fishes: new insights on influencing factors, biological limitations and future strategies. Aquaculture. 2017;467:3–27. https://doi.org/10.1016/j.aquaculture.2016.02.007.

    Article  CAS  Google Scholar 

  4. Shi Y, Zhong L, Zhong H, Zhang J, Liu X, Peng M, et al. Taurine supplements in high-carbohydrate diets increase growth performance of Monopterus albus by improving carbohydrate and lipid metabolism, reducing liver damage, and regulating intestinal microbiota. Aquaculture. 2022;554:738150. https://doi.org/10.1016/j.aquaculture.2022.738150.

    Article  CAS  Google Scholar 

  5. Liang J, Zhao L, Gong C, Liu Q, Hu Y, Liu H, et al. Yinchenhao decoction alleviates high-carbohydrate diet-induced hepatic lipids deposition by strengthening lipids metabolism and transport in largemouth bass. Aquac Res. 2022;53(18):6500–12. https://doi.org/10.1111/are.16119.

    Article  CAS  Google Scholar 

  6. Zhou C, Huang Z, Lin H, Ma Z, Wang J, Wang Y, et al. Rhizoma curcumae longae ameliorates high dietary carbohydrate-induced hepatic oxidative stress, inflammation in golden pompano Trachinotus ovatus. Fish Shellfish Immunol. 2022;130:31–42. https://doi.org/10.1016/j.fsi.2022.08.058.

    Article  CAS  PubMed  Google Scholar 

  7. Pan M, Liu D, Liu J, Li X, Huang D, Luo K, et al. Biotin alleviates hepatic and intestinal inflammation and apoptosis induced by high dietary carbohydrate in juvenile turbot (Scophthalmus maximus L.). Fish Shellfish Immunol. 2022;130:560–71. https://doi.org/10.1016/j.fsi.2022.07.063.

    Article  CAS  PubMed  Google Scholar 

  8. Wang F, Xu J, Jakovlić I, Wang W, Zhao Y. Dietary betaine reduces liver lipid accumulation via improvement of bile acid and trimethylamine-N-oxide metabolism in blunt-snout bream. Food Funct. 2019;10(10):6675–89. https://doi.org/10.1039/C9FO01853K.

    Article  CAS  PubMed  Google Scholar 

  9. Tao J, Gong Y, Chen S, Li W, Xie R, Zhang H, et al. Dietary inclusion of Clostridium butyricum cultures alleviated impacts of high-carbohydrate diets in largemouth bass (Micropterus salmoides). Br J Nutr. 2023;11:1–18. https://doi.org/10.1017/S0007114523002842.

  10. Wang T, Zhang N, Yu X, Qiao F, Chen L, Du Z, et al. Inulin alleviates adverse metabolic syndrome and regulates intestinal microbiota composition in Nile tilapia (Oreochromis niloticus) fed with high-carbohydrate diet. Br J Nutr. 2021;126(2):161–71. https://doi.org/10.1017/s000711452000402x.

    Article  CAS  PubMed  Google Scholar 

  11. Xu R, Li M, Wang T, Zhao Y, Shan C, Qiao F, et al. Bacillus amyloliquefaciens ameliorates high-carbohydrate diet-induced metabolic phenotypes by restoration of intestinal acetate-producing bacteria in Nile tilapia. Br J Nutr. 2022;127(5):653–65. https://doi.org/10.1017/s0007114521001318.

    Article  CAS  PubMed  Google Scholar 

  12. Park G, Jung S, Wellen KE, Jang C. The interaction between the gut microbiota and dietary carbohydrates in nonalcoholic fatty liver disease. Exp Mol Med. 2021;53(5):809–22. https://doi.org/10.1038/s12276-021-00614-x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Chen X, Chen X, Tang X. Short-chain fatty acid, acylation and cardiovascular diseases. Clin Sci (Lond). 2020;134(6):657–76. https://doi.org/10.1042/cs20200128.

    Article  CAS  PubMed  Google Scholar 

  14. Yang X, Mo W, Zheng C, Li W, Tang J, Wu X. Alleviating effects of noni fruit polysaccharide on hepatic oxidative stress and inflammation in rats under a high-fat diet and its possible mechanisms. Food Funct. 2020;11(4):2953–68. https://doi.org/10.1039/d0fo00178c.

    Article  CAS  PubMed  Google Scholar 

  15. Xu J, Chen X, Yu S, Su Y, Zhu W. Effects of early intervention with sodium butyrate on gut microbiota and the expression of inflammatory cytokines in neonatal piglets. PLoS ONE. 2016;11(9):e162461. https://doi.org/10.1371/journal.pone.0162461.

    Article  CAS  Google Scholar 

  16. de Oliveira AF, de Castro Moreira ME, Lima CF, Toledo RCL, de Sousa AR, Veloso MP, et al. Bacupari (Garcinia brasiliensis) extract modulates intestinal microbiota and reduces oxidative stress and inflammation in obese rats. Food Res Int. 2019;122:199–208. https://doi.org/10.1016/j.foodres.2019.04.012.

    Article  CAS  Google Scholar 

  17. Akhtar M, Chen Y, Ma Z, Zhang X, Shi D, Khan JA, et al. Gut microbiota-derived short chain fatty acids are potential mediators in gut inflammation. Anim Nutr. 2022;8:350–60. https://doi.org/10.1016/j.aninu.2021.11.005.

    Article  CAS  PubMed  Google Scholar 

  18. O’Riordan KJ, Collins MK, Moloney GM, Knox EG, Aburto MR, Fülling C, et al. Short chain fatty acids: microbial metabolites for gut-brain axis signalling. Mol Cell Endocrinol. 2022;546:111572. https://doi.org/10.1016/j.mce.2022.111572.

    Article  CAS  PubMed  Google Scholar 

  19. Zhou H, Yu B, Sun J, Liu Z, Chen H, Ge L, et al. Short-chain fatty acids can improve lipid and glucose metabolism independently of the pig gut microbiota. J Anim Sci Biotechnol. 2021;12:61. https://doi.org/10.1186/s40104-021-00581-3.

    Article  CAS  Google Scholar 

  20. Deng M, Qu F, Chen L, Liu C, Zhang M, Ren F, et al. SCFAs alleviated steatosis and inflammation in mice with NASH induced by MCD. J Endocrinol. 2020;245(3):425–37. https://doi.org/10.1530/joe-20-0018.

    Article  CAS  PubMed  Google Scholar 

  21. Jiao A, Diao H, Yu B, He J, Yu J, Zheng P, et al. Infusion of short chain fatty acids in the ileum improves the carcass traits, meat quality and lipid metabolism of growing pigs. Anim Nutr. 2021;7(1):94–100. https://doi.org/10.1016/j.aninu.2020.05.009.

    Article  CAS  PubMed  Google Scholar 

  22. Hu S, Kuwabara R, de Haan BJ, Smink AM, de Vos P. Acetate and butyrate improve β-cell metabolism and mitochondrial respiration under oxidative stress. Int J Mol Sci. 2020;21(4):1542. https://doi.org/10.3390/ijms21041542.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Li M, Hu F, Qiao F, Du Z, Zhang M. Sodium acetate alleviated high-carbohydrate induced intestinal inflammation by suppressing MAPK and NF-κB signaling pathways in Nile tilapia (Oreochromis niloticus). Fish Shellfish Immunol. 2020;98:758–65. https://doi.org/10.1016/j.fsi.2019.11.024.

    Article  CAS  PubMed  Google Scholar 

  24. Zhou W, Limbu SM, Li R, Luo Y, Ren J, Qiao F, et al. Dietary sodium acetate improves high-fat diet utilization through promoting differential nutrients metabolism between liver and muscle in Nile tilapia (Oreochromis niloticus). Aquaculture. 2023;565:739142. https://doi.org/10.1016/j.aquaculture.2022.739142.

    Article  CAS  Google Scholar 

  25. Zhu W, Cai W, Zeng H, Qiu L, Niu Y, Zhang H, et al. Sodium acetate alleviates the liver injury induced by high carbohydrate diets in swamp eel Monopterus albus through inhibiting inflammation formation. Aquac Rep. 2023;33:101817. https://doi.org/10.1016/j.aqrep.2023.101817.

    Article  Google Scholar 

  26. Chen W, Gao S, Chang K, Zhao X, Niu B. Dietary sodium butyrate supplementation improves fish growth, intestinal microbiota composition, and liver health in largemouth bass (Micropterus salmoides) fed high-fat diets. Aquaculture. 2023;564:739040. https://doi.org/10.1016/j.aquaculture.2022.739040.

    Article  CAS  Google Scholar 

  27. Chen W, Chang K, Chen J, Zhao X, Gao S. Dietary sodium butyrate supplementation attenuates intestinal inflammatory response and improves gut microbiota composition in largemouth bass (Micropterus salmoides) fed with a high soybean meal diet. Fish Physiol Biochem. 2021;47:1805–19. https://doi.org/10.1007/s10695-021-01004-w.

    Article  CAS  PubMed  Google Scholar 

  28. Zhou Y, He G, Jin T, Chen Y, Dai F, Luo L, et al. High dietary starch impairs intestinal health and microbiota of largemouth bass, Micropterus salmoides. Aquaculture. 2020;534:736261. https://doi.org/10.1016/j.aquaculture.2020.736261.

    Article  CAS  Google Scholar 

  29. Shah Alam M, Watanabe WO, Carroll PM. Dietary protein requirements of juvenile black sea bass, Centropristis striata. J World Aquac Soc. 2008;39(5):656–63. https://doi.org/10.1111/j.1749-7345.2008.00204.x.

    Article  Google Scholar 

  30. Huang D, Wu Y, Lin Y, Chen J, Karrow N, Ren X, et al. Dietary protein and lipid requirements for juvenile largemouth bass, Micropterus salmoides. J World Aquac Soc. 2017;48(5):782–90. https://doi.org/10.1111/jwas.12417.

    Article  CAS  Google Scholar 

  31. Zhao L, Liao L, Tang X, Liang J, Liu Q, Luo W, et al. High-carbohydrate diet altered conversion of metabolites, and deteriorated health in juvenile largemouth bass. Aquaculture. 2022;549:737816. https://doi.org/10.1016/j.aquaculture.2021.737816.

    Article  CAS  Google Scholar 

  32. Zhao L, Liang J, Chen F, Tang X, Liao L, Liu Q, et al. High carbohydrate diet induced endoplasmic reticulum stress and oxidative stress, promoted inflammation and apoptosis, impaired intestinal barrier of juvenile largemouth bass (Micropterus salmoides). Fish Shellfish Immunol. 2021;119:308–17. https://doi.org/10.1016/j.fsi.2021.10.019.

    Article  CAS  PubMed  Google Scholar 

  33. Zhao L, Liang J, Liu H, Gong C, Huang X, Hu Y, et al. Yinchenhao decoction ameliorates the high-carbohydrate diet induced suppression of immune response in largemouth bass (Micropterus salmoides). Fish Shellfish Immunol. 2022;125:141–51. https://doi.org/10.1016/j.fsi.2022.05.015.

    Article  CAS  PubMed  Google Scholar 

  34. Hou D, Li M, Li P, Chen B, Huang W, Guo H, et al. Effects of sodium butyrate on growth performance, antioxidant status, inflammatory response and resistance to hypoxic stress in juvenile largemouth bass (Micropterus salmoides). Front Immunol. 2023;14:1265963. https://doi.org/10.3389/fimmu.2023.1265963.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Baur FJ, Ensminger LG. The association of official analytical chemists (AOAC). J Am Oil Chem Soc. 1977;54(4):171–2. https://doi.org/10.1007/BF02670789.

    Article  Google Scholar 

  36. National Research Council. Nutrient requirements of fish and shrimp. Washington: National Academic Press; 2011.

  37. Schmittgen TD. Real-time quantitative PCR. Methods. 2001;4(25):383–5. https://doi.org/10.1101/gr.6.10.986.

    Article  Google Scholar 

  38. Zhao L, Cheng L, Hu Y, Li X, Yang Y, Mu J, et al. Dietary sodium acetate and sodium butyrate attenuate intestinal damage and improve lipid metabolism in juvenile largemouth bass (Micropterus salmoides) fed a high carbohydrate diet by reducing endoplasmic reticulum stress. Anim Nutr. 2024;16:443–56. https://doi.org/10.1016/j.aninu.2023.12.002.

  39. Xu R, Wang T, Ding F, Zhou N, Qiao F, Chen L, et al. Lactobacillus plantarum ameliorates high-carbohydrate diet-induced hepatic lipid accumulation and oxidative stress by upregulating uridine synthesis. Antioxidants (Basel). 2022;11(7):1238. https://doi.org/10.3390/antiox11071238.

    Article  CAS  PubMed  Google Scholar 

  40. Wang T, Xu R, Qiao F, Du Z, Zhang M. Effects of mannan oligosaccharides (MOS) on glucose and lipid metabolism of largemouth bass (Micropterus salmoides) fed with high carbohydrate diet. Anim Feed Sci Technol. 2022;292:115449. https://doi.org/10.1016/j.anifeedsci.2022.115449.

    Article  CAS  Google Scholar 

  41. Ipsen DH, Lykkesfeldt J, Tveden-Nyborg P. Molecular mechanisms of hepatic lipid accumulation in non-alcoholic fatty liver disease. Cell Mol Life Sci. 2018;75:3313–27. https://doi.org/10.1007/s00018-018-2860-6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Prisingkorn W, Prathomya P, Jakovlić I, Liu H, Zhao Y, Wang W. Transcriptomics, metabolomics and histology indicate that high-carbohydrate diet negatively affects the liver health of blunt snout bream (Megalobrama amblycephala). BMC Genomics. 2017;18:856. https://doi.org/10.1186/s12864-017-4246-9.

    Article  CAS  Google Scholar 

  43. Den Besten G, Bleeker A, Gerding A, van Eunen K, Havinga R, van Dijk TH, et al. Short-chain fatty acids protect against high-fat diet-induced obesity via a PPARγ-dependent switch from lipogenesis to fat oxidation. Diabetes. 2015;64(7):2398–408. https://doi.org/10.2337/db14-1213.

    Article  CAS  Google Scholar 

  44. Li Q, Chen H, Zhang M, Wu T, Liu R. Altered short chain fatty acid profiles induced by dietary fiber intervention regulate ampk levels and intestinal homeostasis. Food Funct. 2019;10(11):7174–87. https://doi.org/10.1039/c9fo01465a.

    Article  CAS  PubMed  Google Scholar 

  45. Miao S, Li Y, Mu T, Wang X, Zhao W, Li R, et al. Dietary coated sodium butyrate ameliorates hepatic lipid accumulation and inflammation via enhancing antioxidative function in post-peaking laying hens. Metabolites. 2023;13(5):650. https://doi.org/10.3390/metabo13050650.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Singh R, Kaushik S, Wang Y, Xiang Y, Novak I, Komatsu M, et al. Autophagy regulates lipid metabolism. Nature. 2009;458(7242):1131–5. https://doi.org/10.1038/s41467-019-08829-3.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  47. Zhao T, Wu K, Hogstrand C, Xu Y, Chen G, Wei C, et al. Lipophagy mediated carbohydrate-induced changes of lipid metabolism via oxidative stress, endoplasmic reticulum (ER) stress and ChREBP/PPARγ pathways. Cell Mol Life Sci. 2020;77:1987–2003. https://doi.org/10.1007/s00018-019-03263-6.

    Article  CAS  PubMed  Google Scholar 

  48. Li X, Wang C, Zhu J, Lin Q, Yu M, Wen J, et al. Sodium butyrate ameliorates oxidative stress-induced intestinal epithelium barrier injury and mitochondrial damage through ampk-mitophagy pathway. Oxid Med Cell Longev. 2022;2022:3745135. https://doi.org/10.1155/2022/3745135.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Zhou H, Li G, Wang Y, Jiang R, Li Y, Wang H, et al. Microbial metabolite sodium butyrate attenuates cartilage degradation by restoring impaired autophagy and autophagic flux in osteoarthritis development. Front Pharmacol. 2021;12:659597. https://doi.org/10.3389/fphar.2021.659597.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Neuschwander-Tetri BA. Non-alcoholic fatty liver disease. BMC Med. 2017;15:45. https://doi.org/10.1186/s12916-017-0806-8.

    Article  CAS  Google Scholar 

  51. Kobayashi M, Li L, Iwamoto N, Nakajima-Takagi Y, Kaneko H, Nakayama Y, et al. The antioxidant defense system Keap1-Nrf2 comprises a multiple sensing mechanism for responding to a wide range of chemical compounds. Mol Cell Biol. 2009;29(2):493–502. https://doi.org/10.1128/mcb.01080-08.

    Article  CAS  PubMed  Google Scholar 

  52. Li D, Bai X, Jiang Y, Cheng Y. Butyrate alleviates ptz-induced mitochondrial dysfunction, oxidative stress and neuron apoptosis in mice via Keap1/Nrf2/HO-1 pathway. Brain Res Bull. 2021;168:25–35. https://doi.org/10.1016/j.brainresbull.2020.12.009.

    Article  CAS  PubMed  Google Scholar 

  53. Chen X, Kong Q, Zhao X, Zhao C, Hao P, Irshad I, et al. Sodium acetate/sodium butyrate alleviates lipopolysaccharide-induced diarrhea in mice via regulating the gut microbiota, inflammatory cytokines, antioxidant levels, and NLRP3/Caspase-1 signaling. Front Microbiol. 2022;13:1036042. https://doi.org/10.3389/fmicb.2022.1036042.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Calder PC. Dietary factors and low-grade inflammation in relation to overweight and obesity revisted. Br J Nutr. 2022;127(10):1455–7. https://doi.org/10.1017/s0007114522000782.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Zhai S, Qin S, Li L, Zhu L, Zou Z, Wang L. Dietary butyrate suppresses inflammation through modulating gut microbiota in high-fat diet-fed mice. FEMS Microbiol Lett. 2019;366(13):z153. https://doi.org/10.1093/femsle/fnz153.

    Article  CAS  Google Scholar 

  56. Cresci GA, Glueck B, McMullen MR, Xin W, Allende D, Nagy LE. Prophylactic tributyrin treatment mitigates chronic-binge ethanol-induced intestinal barrier and liver injury. J Gastroenterol Hepatol. 2017;32(9):1587–97. https://doi.org/10.1111/jgh.13731.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Li S, Heng X, Guo L, Lessing DJ, Chu W. SCFAs improve disease resistance via modulate gut microbiota, enhance immune response and increase antioxidative capacity in the host. Fish Shellfish Immunol. 2022;120:560–8. https://doi.org/10.1016/j.fsi.2021.12.035.

    Article  CAS  PubMed  Google Scholar 

  58. Terova G, Díaz N, Rimoldi S, Ceccotti C, Gliozheni E, Piferrer F. Effects of sodium butyrate treatment on histone modifications and the expression of genes related to epigenetic regulatory mechanisms and immune response in European sea bass (Dicentrarchus labrax) fed a plant-based diet. PLoS ONE. 2016;11(7):e160332. https://doi.org/10.1371/journal.pone.0160332.

    Article  CAS  Google Scholar 

  59. Ho Do M, Seo YS, Park H. Polysaccharides: bowel health and gut microbiota. Crit Rev Food Sci Nutr. 2021;61(7):1212–24. https://doi.org/10.1080/10408398.2020.1755949.

    Article  CAS  PubMed  Google Scholar 

  60. Zou J, Xiang Q, Tan D, Shi L, Liu X, Wu Y, et al. Zuogui-jiangtang-qinggan-fang alleviates high-fat diet-induced type 2 diabetes mellitus with non-alcoholic fatty liver disease by modulating gut microbiome-metabolites-short chain fatty acid composition. Biomed Pharmacother. 2023;157:114002. https://doi.org/10.1016/j.biopha.2022.114002.

    Article  CAS  PubMed  Google Scholar 

  61. Zhao L, Qi Z, Yi L, Li J, Cui Y, Rehman FU, et al. The interaction between gut microbiota and flavonoid extract from Smilax glabra Roxb. and its potent alleviation of fatty liver. Food Funct. 2021;12(17):7836–50. https://doi.org/10.1039/d1fo00727k.

    Article  CAS  PubMed  Google Scholar 

  62. Shi Y, Zhong L, Fan Y, Zhang J, Zhong H, Liu X, et al. The protective effect of mulberry leaf flavonoids on high-carbohydrate-induced liver oxidative stress, inflammatory response and intestinal microbiota disturbance in Monopterus albus. Antioxidants (Basel). 2022;11(5):976. https://doi.org/10.3390/antiox11050976.

    Article  CAS  PubMed  Google Scholar 

  63. Huang X, Zhong L, Kang Q, Liu S, Feng Y, Geng Y, et al. A high starch diet alters the composition of the intestinal microbiota of largemouth bass Micropterus salmoides, which may be associated with the development of enteritis. Front Microbiol. 2021;12:696588. https://doi.org/10.3389/fmicb.2021.696588.

    Article  PubMed  PubMed Central  Google Scholar 

  64. Xun P, Zhou C, Huang X, Huang Z, Yu W, Yang Y, et al. Effects of dietary sodium acetate on intestinal health of juvenile Trachinotus ovatus based on multi-omics approach. Aquaculture. 2023;562:738776. https://doi.org/10.1016/j.aquaculture.2022.738776.

    Article  CAS  Google Scholar 

  65. Liu L, Wang Y, Ren J, Zou H, Wang C. Effect of dietary supplementation with sodium butyrate and tributyrin on the growth performance and intestinal microbiota of pacific white shrimp (Litopenaeus vannamei). Aquac Int. 2022;30(5):2477–89. https://doi.org/10.1007/s10499-022-00909-4.

    Article  CAS  Google Scholar 

  66. Brennan CA, Garrett WS. Fusobacterium nucleatum-symbiont, opportunist and oncobacterium. Nat Rev Microbiol. 2019;17(3):156–66. https://doi.org/10.1038/s41579-018-0129-6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Hamer HM, Jonkers D, Venema K, Vanhoutvin S, Troost FJ, Brummer RJ. The role of butyrate on colonic function. Aliment Pharmacol Ther. 2008;27(2):104–19. https://doi.org/10.1111/j.1365-2036.2007.03562.x.

    Article  CAS  PubMed  Google Scholar 

  68. Hong J, Jia Y, Pan S, Jia L, Li H, Han Z, et al. Butyrate alleviates high fat diet-induced obesity through activation of adiponectin-mediated pathway and stimulation of mitochondrial function in the skeletal muscle of mice. Oncotarget. 2016;7(35):56071–82. https://doi.org/10.18632/oncotarget.11267.

    Article  PubMed  PubMed Central  Google Scholar 

  69. Aalamifar H, Soltanian S, Vazirzadeh A, Akhlaghi M, Morshedi V, Gholamhosseini A, et al. Dietary butyric acid improved growth, digestive enzyme activities and humoral immune parameters in barramundi (Lates calcarifer). Aquac Nutr. 2020;26(1):156–64. https://doi.org/10.1111/anu.12977.

    Article  CAS  Google Scholar 

  70. Chow EPY, Liong KH, Schoeters E. Dietary encapsulated butyric acid (Butipearl™) and microemulsified carotenoids (Quantum GLO™ Y) on the growth, immune parameters and their synergistic effect on pigmentation of hybrid catfish (Clarias macrocephalus × Clarias gariepinus). J Fish Aquac. 2017;8(2):195. https://doi.org/10.4172/2150-3508.1000195.

  71. Holzapfel W, Wood BJ. The genera of lactic acid bacteria. Springer Science & Business Media; 2012.

  72. Vendrell D, Balcázar JL, Ruiz-Zarzuela I, De Blas I, Gironés O, Múzquiz JL. Lactococcus garvieae in fish: a review. Comp Immunol Microbiol Infect Dis. 2006;29(4):177–98. https://doi.org/10.1016/j.cimid.2006.06.003.

    Article  PubMed  Google Scholar 

  73. Zhang H, Ding Q, Wang A, Liu Y, Teame T, Ran C, et al. Effects of dietary sodium acetate on food intake, weight gain, intestinal digestive enzyme activities, energy metabolism and gut microbiota in cultured fish: zebrafish as a model. Aquaculture. 2020;523:735188. https://doi.org/10.1016/j.aquaculture.2020.735188.

    Article  CAS  Google Scholar 

  74. Zhou DA, Pan Q, Xin F, Zhang R, He C, Chen G, et al. Sodium butyrate attenuates high-fat diet-induced steatohepatitis in mice by improving gut microbiota and gastrointestinal barrier. World J Gastroenterol. 2017;23(1):60–75. https://doi.org/10.3748/wjg.v23.i1.60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Ley RE, Turnbaugh PJ, Klein S, Gordon JI. Human gut microbes associated with obesity. Nature. 2006;444(7122):1022–3. https://doi.org/10.1038/4441022a.

    Article  ADS  CAS  PubMed  Google Scholar 

  76. Roediger WE. Famine, fiber, fatty acids, and failed colonic absorption: does fiber fermentation ameliorate diarrhea? J Parenter Enteral Nutr. 1994;18(1):4–08. https://doi.org/10.1177/014860719401800104.

    Article  CAS  Google Scholar 

  77. Takano M, Rhoads DB, Isselbacher KJ. Sodium butyrate increases glucose transporter expression in LLC-PK1 cells. Proc Natl Acad Sci USA. 1988;85(21):8072–5. https://doi.org/10.1073/pnas.85.21.8072.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  78. Zhou W, Limbu SM, Luo Y, Li R, Ren J, Qiao F, et al. Dietary acetate promotes growth and nutrients deposition in Nile tilapia (Oreochromis niloticus) through increasing acetyl-CoA-triggered energy production. Aquaculture. 2023;575:739750. https://doi.org/10.1016/j.aquaculture.2023.739750.

  79. De Schryver P, Sinha AK, Kunwar PS, Baruah K, Verstraete W, Boon N, et al. Poly-β-hydroxybutyrate (PHB) increases growth performance and intestinal bacterial range-weighted richness in juvenile European sea bass, Dicentrarchus labrax. Appl Microbiol Biotechnol. 2010;86:1535–41. https://doi.org/10.1007/s00253-009-2414-9.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank LetPub for its linguistic assistance in preparing this manuscript.

Funding

This study was supported by the Double Support Project (035–2221993229).

Author information

Authors and Affiliations

Authors

Contributions

QL: Funding acquisition, Project administration, Data curation, Conceptualization, Methodology. LSC: Formal analysis, Manuscript writing. MZW: Investigation, Management. LFS, CXZ and JM: Resource, Investigation. YFH: Data curation. YHY: Writing—review and editing. KH: Date analysis. HXY: Writing—review. LLZ: Writing—review and editing. SY: Conceptualization, Supervision. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Liulan Zhao or Song Yang.

Ethics declarations

Ethics approval and consent to participate

This research was approved by the Animal Care Advisory Committee of Sichuan Agricultural University (No. 2021202068).

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Supplementary Information

Additional file 1:

 Table S1. SA and SB requirements of different fish species. Table S2. Antibodies used for western blot analysis.

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

Liu, Q., Cheng, L., Wang, M. et al. Dietary sodium acetate and sodium butyrate improve high-carbohydrate diet utilization by regulating gut microbiota, liver lipid metabolism, oxidative stress, and inflammation in largemouth bass (Micropterus salmoides). J Animal Sci Biotechnol 15, 50 (2024). https://doi.org/10.1186/s40104-024-01009-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40104-024-01009-4

Keywords