Skip to main content

Caffeic acid and chlorogenic acid mediate the ADPN-AMPK-PPARα pathway to improve fatty liver and production performance in laying hens

Abstract

Background

Caffeic acid (CA) and its derivative, chlorogenic acid (CGA), have shown promise in preventing and alleviating fatty liver disease. CA, compared to CGA, has much lower production costs and higher bioavailability, making it a potentially superior feed additive. However, the efficacy, mechanistic differences, and comparative impacts of CA and CGA on fatty liver disease in laying hens remain unclear. This study aimed to evaluate and compare the effects of CA and CGA on production performance, egg quality, and fatty liver disease in laying hens.

Results

A total of 1,440 61-week-old Hyline Brown laying hens were randomly divided into 8 groups and fed diets supplemented with basal diet, 25, 50, 100 and 200 mg/kg of CA, and 100, 200 and 400 mg/kg of CGA (CON, CA25, CA50, CA100, CA200, CGA100, CGA200 and CGA400, respectively) for 12 weeks. Both CA and CGA improved production performance and egg quality, while reducing markers of hepatic damage and lipid accumulation. CA and CGA significantly decreased TG, TC, and LDL-C levels and increased T-SOD activity. Transcriptomic and proteomic analyses revealed that CA and CGA reduced hepatic lipid accumulation through downregulation of lipid biosynthesis-related genes (ACLY, ACACA, FASN, and SCD1) and enhanced lipid transport and oxidation genes (FABPs, CD36, CPT1A, ACOX1, and SCP2). Of note, low-dose CA25 exhibited equivalent efficacy to the higher dose CGA100 group in alleviating fatty liver conditions. Mechanistically, CA and CGA alleviated lipid accumulation via activation of the ADPN-AMPK-PPARα signaling pathway.

Conclusions

This study demonstrates that dietary CA and CGA effectively improve laying performance, egg quality, and hepatic lipid metabolism in laying hens, with CA potentially being more economical and efficient. Transcriptomic and proteomic evidence highlight shared mechanisms between CA25 and CGA100. These findings provide a foundation for CA and CGA as therapeutic agents for fatty liver disease and related metabolic diseases in hens, and also offer insights into the targeted modification of CGA (including the isomer of CGA) into CA, thereby providing novel strategies for the efficient utilization of CGA.

Graphical Abstract

Highlights

(1) Dietary CA and CGA improve fatty liver, laying performance and egg quality.

(2) Lower dose of CA25 achieves the equivalent improvement as CGA100 or CGA200.

(3) CA and CGA mediate the ADPN-AMPK-PPARα pathway to alleviate fatty liver.

Introduction

As laying hens gradually enter the later laying stages, their production performance and egg quality begin to decline [1,2,3], accompanied by significant health challenges. Among these, fatty liver disease emerges as a critical concern, characterized by excessive hepatic lipid deposition, coupled with organ aging and poor health. This disease is the major cause of non-infectious death in caged laying hens and has enormous economic ramifications for the poultry industry [4,5,6,7]. Fatty liver is characterized by the pathological accumulation of hepatic and abdominal lipid, manifesting as lipid metabolism disorder, hepatic steatosis, and inflammatory reactions [8,9,10], largely driven by genetics, nutrition, environment, and hormones [11,12,13,14].

Fatty liver in mammals is usually caused by an increase in liver fatty acid uptake and synthesis, coupled with an inability to timely transport or oxidatively decompose these fats resulting in excessive fat accumulation in hepatocytes and eventually leading to hepatic steatosis [15]. This process can further develop into severe fatty liver when uncontrolled [14]. Unlike mammals however, avian species rely heavily on the liver for lipogenesis, with 90%–95% of de novo fatty acid synthesis occurring in this organ [16, 17]. Thus, reducing hepatic lipid storage is an effective intervention strategy to alleviate fatty liver in laying hens.

Caffeic acid (CA) and chlorogenic acid (CGA) are abundant in coffee beans, Eucommia leaves, honeysuckle and tea, and are also one of the major active ingredients of these plants [18, 19]. These compounds are widely studied in the alleviation of metabolic diseases such as obesity and metabolically associated fatty liver disease (MAFLD) due to their excellent lipid-lowering effect [20]. Mechanistically, both CA and CGA exhibit protective effects against liver steatosis [21,22,23,24], oxidative stress [25, 26], inflammation [27], and fibrosis [28] in mammals. Furthermore, studies indicate that both CA and CGA exert anti-lipogenesis effect by activating the AMPK signaling pathway [29,30,31]. Unfortunately, the low absorptivity of CGA is a primary factor limiting the efficient utilization among various kinds of diseases at present [32]. Studies have shown that most of the CA is absorbed into the blood, while only about one third of the CGA is absorbed [33, 34]. Meanwhile, the efficiency of CGA utilization largely depends on its metabolism by gut microflora in rats [32], with CA as the primary metabolite, accounting for 57.4% of CGA intake [32, 35].

At present, CA production has a much lower cost advantage and whether CA has equivalent beneficial effect as CGA is unknown in laying hens. Based on these studies, the pharmacological action of CGA is perhaps largely determined by how much CGA is enzymatically decomposed into CA and enters the liver to exert biological effects. However, these conjectures need to be further verified. Meanwhile, the beneficial effects and pharmacological mechanisms of CA and CGA on fatty liver in laying hens have not yet been documented.

Therefore, we hypothesized that both CA and CGA can improve fatty liver disease and production performance of laying hens, but the utilization efficiency of CA is higher and feed cost is more economical. To explore this hypothesis, the effect of various concentrations of CA and CGA was tested and the mechanisms by which CA and CGA may alleviate fatty liver and improve production performance in the aged laying hens were explored. The findings of this study can provide a theoretical basis and research foundation for the application of CA and CGA to alleviate fatty liver in laying hens.

Materials and methods

Animals and experimental design

A total of 1,440 61-week-old Hyline Brown laying hens were randomly divided into 8 groups, with each group consisting of 10 replicates and 18 hens per replicate. The hens were fed with diets supplemented with 0 (basal diet), 25, 50, 100 and 200 mg/kg of caffeic acid (CA), or 100, 200 and 400 mg/kg of chlorogenic acid (CGA) (CON, CA25, CA50, CA100, CA200, CGA100, CGA200 and CGA400 groups, respectively) for 12 weeks. CA and CGA were purchased from Shanxi Bolin Biotechnology Co., Ltd. (Shanxi, China) (purity > 98%). The CA and CGA supplemental doses were based on references reported in other animals [36,37,38]. Feed and water were available ad libitum throughout the experiment, which followed a lighting schedule of 16 h of light and 8 h of darkness during the whole experimental period. The composition and nutrient contents of the basal diet are presented in Table 1. All diets were formulated to meet or exceed the estimated nutrient requirements for laying hens as recommended by the NRC (1994).  

Table 1 Composition and nutrient contents of basal diet

Sample collection and procedures

At the end of the 12-week feeding period, one hen was randomly selected from each replicate per treatment group after a 12-h feed deprivation. The body weight was recorded, and blood was collected from the brachial vein and centrifuged at 4 °C to obtain plasma, which was then stored at −80 °C for further analysis. Another tube of anticoagulant blood (whole blood) was immediately sent to the laboratory for routine blood tests. The hens were sacrificed by exsanguination following cervical dislocation. Liver tissues were immediately removed and weighed, and then the morphology was photographed and observed. The liver specimens were fixed using 4% paraformaldehyde solution for the production of pathological sections. Parts of the livers were then rapidly frozen in liquid nitrogen and stored at −80 °C for further analysis.

Measurements of laying performance and egg quality

Throughout the study, egg numbers and egg weight were recorded daily, laying rate, average daily feed intake (ADFI) and feed conversion ratio (FCR) were calculated weekly. At the end of 1, 6 and 12 weeks, 3 eggs were taken from each replicate to measure egg quality indicators. Eggshell strength was determined using an egg force reader (Orka Food Technology Ltd., Ramat Hasharon, Israel). Eggshell thickness (air cell, equator, and sharp end) was estimated using electronic digital calipers (ABS LUTE 547–360, Mitutoyo, Japan). Haugh unit and albumen height were evaluated using an egg quality analyzer (Orka Food Technology Ltd., Ramat Hasharon, Israel). Yolk color (International commission on illumination L*a*b*, CIELAB) value was assessed using a colorimeter (CR-400, Konica Minolta, Inc., Tokyo, Japan). L*, a*, and b* indicate relative lightness, relative redness, and relative yellowness, respectively.

Blood and serum biochemical indices

Whole blood samples were immediately transferred to the laboratory of Animal Hospital at China Agricultural University for hematological analysis. Hematology was analyzed using a TEK-II automatic hematology analyzer (Beijing Kangjia Hongyuan Biological Technology Co., Ltd., Beijing, China). Serum lipid metabolism-related indices, including triglyceride (TG), total cholesterol (TC), high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C), very low-density lipoprotein cholesterol (VLDL-C), and liver injury biomarkers (AST and ALT), were analyzed according to the manufacturer's instructions (Nanjing Jiancheng Bioengineering Institute, Nanjing, China) using the Mindray BS-370E fully automatic biochemical detection system (Mindray, Shenzhen, China). The activity levels of glutathione peroxidase (GSH-PX), total superoxide dismutase (T-SOD) and total antioxidant oxidase (T-AOC) and malondialdehyde (MDA) were determined using the corresponding kits (Nanjing Jiancheng Bioengineering Institute, Nanjing, China) according to the manufacturer’s instructions.

Determination of hepatic biochemical indexes and antioxidant capacity

A 0.1 g sample of each liver tissue was homogenized in 0.9 mL phosphate-buffered saline (PBS) with a tissue homogenizer. Then, centrifugation at 4 °C for 10 min was performed, and the supernatant was harvested for biochemical parameter determination. The contents of TG, TC, HDL-C, LDL-C, VLDL-C, and the activity levels of GSH-PX, T-SOD, T-AOC and MDA in supernatant were quantified with the commercial kits (Nanjing Jiancheng Bioengineering Institute, Nanjing, China) following the manufacturer’s protocols. Total protein concentration in the supernatant was determined with Nanjing Jiancheng Total Protein Quantitative Assay Kit (with standard: BCA method) to correct for all liver biochemical parameters.

Histological observation of liver

The liver tissues were fixed with 4% paraformaldehyde, dehydrated with gradient alcohol and made transparent with xylene. These transparent samples were embedded in paraffin, cut into 5-μm slices, stained with hematoxylin and eosin (H&E), and sheet sealed. The morphology of the liver was observed under a light microscope with different magnification (CK-40, Olympus, Tokyo, Japan).

Liver Oil Red O staining

For Oil Red O staining, the liver tissues were fixed, embedded in OTC compound and immediately put on the −20 °C quick-freezing machine, sections were cut at 8 μm slide by the CryoStar NX50 HOVPD freezing microtome (Thermo, MA, USA), and stored at −20 °C. The samples were stained with freshly diluted Oil Red O solution for 15 min under dark conditions at room temperature. Following color separation with 60% isopropanol for 20 s and a gentle rinse with water for 3 min, the nuclei were counterstained with hematoxylin for 5 min. After rinsing with distilled water, the slides were sealed with glycerin gelatin. Images of each group were photographed with a light microscope (CK-40, Olympus, Tokyo, Japan).

RNA sequencing and data analysis

A total of 24 liver samples (6 samples per group) were used for RNA-seq. The primary process is as follows: (1) RNA extraction and quality detection: briefly, total RNA was extracted from liquid nitrogen-frozen liver tissue by using TRIzol (Invitrogen). NanoDrop 2000/N50 detected the purity and concentration of RNA samples, and Agilent 4200 assessed the integrity, to ensure the qualified samples for subsequent transcriptomic sequencing. (2) Library construction and sequencing: Transcriptome sequencing libraries were constructed according to the standard Illumina RNA-seq protocol. After the library was qualified, PE150 mode sequencing was performed using Illumina NovaSeq 6000 sequencing platform (Illumina Inc., San Diego, CA, USA; Berry & Kang Biotechnology Co., Ltd., Beijing, China). (3) Bioinformatic analysis: The raw reads obtained by sequencing were filtered to obtain clean reads, bowtie2 (v.2.3.2) was used to compare clean reads to the silva database to remove rRNA [39]. The remaining reads were compared with the reference genome (Gallus gallus: https://www.ncbi.nlm.nih.gov/datasets/genome/GCF_016699485.2/) using Hisat2 (v.2.2.1) [40]. The ratio of sequencing data to the reference genome was calculated based on the gene location information specified in the genome annotation file (GTF format). FeatureCounts (v.1.6.3) is used to recount the gene levels of each sample respectively [41], and the FPKM value is obtained. (4) Principal component analysis (PCA) represented the relationships and differences between samples by means of linear dimensionality reduction. Differentially expressed genes (DEGs) were identified with edgeR as follows: |log2(Fold Change)| > 1.0 and P-value < 0.05. These DEGs were enriched by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) using topGO and KOBAS (v3.0) respectively.

Proteomic analysis

Protein extraction and quality control

Four samples were randomly selected from each group for proteomic analysis. In brief, the steps were as follows: (1) 25 mg of sample was weighed, and transferred into a 2-mL centrifuge tube. Steel ball, lysis solution containing 8 mol/L Urea/50 mmol/L Tris-HCl and Roche cocktail with 1X final concentration were added into the centrifuge tubes, and placed on ice for 5 min. (2) Crush and crack using a grinding instrument (60 Hz, 2 min), centrifuge at 20,000 × g and 4 °C for 15 min, and collect the supernatant. (3) Add dithiothreitol (DTT) with a final concentration of 10 mmol/L and bathe in medium water at 37 °C for 1 h. (4) Add iodoacetamide (IAA) with a final concentration of 20 mmol/L and place away from light. (5) Take 10 μg protein solution for each sample and add appropriate loading buffer, mix well, heat at 95 °C for 5 min, centrifuge at 20,000 × g for 5 min, take supernatant and add it into the sample hole of 4%–12% SDS polyacrylamide gel. 80 V constant pressure electrophoresis for 20 min, then 120 V constant pressure electrophoresis for 60 min. After the end of electrophoresis, the glue is dyed and decolorized, and it was taken out and photographed, and the quality was judged according to the glue map.

Enzymatic digestion

The experimental procedures were as follows: (1) 150 μg protein was taken from each sample. (2) Add 3 μg Trypsin at the ratio of 50:1 (protein:enzyme), enzymolysis at 37 °C for 14–16 h. (3) The enzymolysis peptides were desalted using Waters solid phase extraction column and vacuum-drained. (4) The drained peptides were redissolved in pure water and stored at −20 °C.

Quantitative detection (Nano-LC–MS/MS) and analysis

The extracted peptide sample was redissolved with 0.1% FA, centrifuged at 20,000 × g for 10 min. The supernatant was collected and injected into a self-loading C18 column (100 μm I.D., 1.8 μm column media particle size, approximately 35 cm column length). Separation was performed by Thermo Scientific EASY-nLC™ 1200 system at a flow rate of 300 nL/min. The liquid phase separated peptides were ionized by nanoESI source and then entered the mass spectrometer Orbitrap Exploris™ 480 (Thermo Fisher Scientific, San Jose, CA, USA). A new data-independent acquisition (DIA) model was adopted for mass spectrometry data acquisition [42]. It has greater advantages than the Data-dependent acquisition (DDA) mode previously adopted by TMT, Label-free and SILAC. The DIA can determine protein molecules with very low abundance in samples more efficiently, greatly improving the reliability of quantitative analysis, and has high quantitative accuracy and repeatability.

DIA-NN software (https://hpc.nih.gov/apps/diann.html) (version 1.8) was used to conduct protein search, identification and quantitative analysis of the DIA mass spectrometry data. The main parameters of DIA-NN are as follows: The protein identification mode was DirectDIA (no actual DDA data was needed to construct the spectral library, and the theoretical virtual spectral library was constructed from the protein sequence library of species Gallus gallus by information analysis method). Then, t-test was utilized to identify differentially expressed proteins (DEPs) based on their relative quantitative values (|log2(Fold Change)| ≥ 0.263 and P-value < 0.05 as the threshold standard). Functional enrichment analysis maps all differential proteins to individual entries annotated in the protein GO and KEGG databases. GSEA (Gene Set Enrichment Analysis) was based on information about the expression levels of all proteins. STRING database (version 11.0) was employed for the analysis of protein–protein interaction (PPI) networks [43].

RNA extraction, cDNA synthesis, and qRT-PCR analysis

Total RNA was isolated from liver tissues using TRIzol reagent (Invitrogen). RNA (0.5 µg) was transcribed into cDNA with the Hiscript® III All-in-one RT SuperMix Perfect for qPCR kits (R333-01, Vazyme Biotech Co., Ltd., Nanjing, China). qRT-qPCR was performed using the Applied Biosystems QuantStudio 7 Flex System (Life Technologies, Waltham, USA) following the instructions of Taq Pro Universal SYBR qPCR Master Mix (Q712-02, Vazyme Biotech Co., Ltd., Nanjing, China). mRNA expression levels were normalized to housekeeping genes (β-actin and GAPDH), and the data were analyzed using the comparative 2Ct method. The primers utilized in the experiment are shown in Table 2.

Table 2 Primer sequences for qRT-PCR

Statistical analysis

SPSS 27.0 software (SPSS Inc., Chicago, IL, USA) was utilized to conduct one-way ANOVA and Duncan multiple comparison of all data, encompassing laying hen performance, egg quality, biochemical indexes, antioxidant and anti-inflammatory indices, etc. Results were presented as mean ± SEM (standard error of the mean); n = 10 hens per group. GraphPad Prism 9.5 (GraphPad Software, San Diego, CA, USA) was employed for data visualization. Statistical significance was considered at a P-value threshold less than 0.05.

Results

CA and CGA enhanced laying performance in laying hens

Laying performance was recorded daily during the period of test. As illustrated in Table 3, the supplementation groups of CA and CGA exhibited noticeably higher egg laying rate compared to the CON group from week 1 to 6. Moreover, compared to the CON group, the average egg weight was significantly increased in the CA25, CGA200 and CGA400 groups. The data also showed that one-eighth of the CA additive dose achieved an equivalent improvement as CGA by comparing the CA25 group with the CGA200 group in terms of egg-laying rate and average egg weight. However, ADFI and FCR were not strikingly altered. However, these differences were only observed in the first 6 weeks, and sustained better performance was not observed in all CA and CGA groups from week 7 to 12. A dose dependent effect of CA was also not found. In summary, dietary CA and CGA contributed to enhanced egg-laying performance.

Table 3 Effect of the dietary CA and CGA on the laying performance

CA and CGA improved egg quality in laying hens

The results of egg quality determination are presented in Table 4. At the end of weeks 1, 6, and 12, the albumen height and Haugh unit were enhanced in CA and CGA groups compared with the CON group, but shell strength, shell thickness, and yolk color were not significantly altered. However, yolk lightness (L*), yolk redness (a*), and yolk yellowness (b*) were reduced in CA and CGA groups, compared to the CON group. The results also indicated that a quarter of the added CA had an equivalent effect to CGA (comparing CA25 and CGA100) on albumen height and Haugh unit from week 1 to week 6. Overall, dietary CA and CGA resulted in promoted egg quality compared to CON group.

Table 4 Effect of the dietary CA and CGA on the egg quality

CA and CGA changed hematology indices

Table 5 presents the hematology indices at the end of week 12. Generally speaking, hematological indicators are vital for assessing laying hens’ health, revealing immune function, hematopoiesis, and stress response. Increases in white blood cells (WBC) indicate infections or immune responses, while changes in red blood cells (RBC) suggest anemia from deficiencies or health issues. Here, our results shown that the addition of CA and CGA significantly decreased the WBC compared to the CON group, yet it did not alter the RBC, indicating that CA and CGA benefited the immunity of laying hens with basically no health issues. The other blood parameters were not significantly influenced by CA25 compared to the CON group. Additionally, excluding middle cell absolute value (MID#), hemoglobin (HGB), mean corpuscular hemoglobin (MCH), mean corpuscular hemoglobin (MCHC) and platelet distribution width (PDW), CGA also had no significant effect on blood when its addition did not exceed 200 mg/kg compared to the CON group. In summary, addition of CA and CGA has positive significance in improving the immunity of laying hens.

Table 5 Effect of the dietary CA and CGA on the routine blood test

CA and CGA ameliorated the levels of serum lipid metabolism and antioxidant activity

The serum lipid metabolism and antioxidant activity were assessed at the end of week 12. As presented in Fig. 1A, compared with the CON group, serum TG, TC, LDL-C, and VLDL-C levels were significantly reduced in the CA25 group, but no significant differences were observed at higher concentrations of CA, including serum HDL-C levels. However, compared to the CON group, serum TG, LDL-C, and VLDL-C levels were not significantly affected by CGA groups except for a significant reduction in serum TC and an increase in HDL-C in the CGA200 and CGA400 groups (Fig. 1D). However, compared to the CON group, the serum antioxidant system (GSH-PX, T-SOD and T-AOC) was not altered, but dietary CA and CGA resulted in an increase in the level of the oxidant cue (MDA) in most of CA and CGA groups (Fig. 1B, C, E, and F). The results also established that the CA25 group had the best effect on reducing serum lipid levels compared with other groups.

Fig. 1
figure 1

Effects of CA and CGA on serum lipid metabolism levels and antioxidant activity. A and D Serum TG, TC, HDL-C, LDL-C and VLDL-C levels (n = 10). B and E Serum GSH-PX, T-SOD and T-AOC activity levels (n = 10). C and F Serum MDA content (n = 10). Data are presented as the mean ± SEM. a−cMeans with different letter differ significantly (P < 0.05)

CA and CGA alleviated hepatic lipid accumulation and liver damage

To evaluate the therapeutic effects of CA and CGA on fatty liver, focused analysis was conducted using liver specimens. Liver tissue H&E staining and phenotype images revealed that the CON group exhibited hepatic steatosis with yellowish liver and numerous fat vacuoles. However, this deterioration was significantly attenuated by administration of CA and CGA (Fig. 2A, B, D, and E). Meanwhile, CA and CGA had no significant effect on liver index compared with the CON group (Fig. 2G and J). Oil Red O staining indicated that the liver tissues in the CA and CGA groups had lower lipid accumulation compared to the CON group (Fig. 2C and F). ALT and AST activities, which are serum indicators used to assess liver damage [44], were significantly decreased in the CA25 and CGA100 groups compared to the CON group, suggesting alleviation of liver damage (Fig. 2H, I, K, and L). Additionally, intrahepatic TG, TC, LDL-C contents were decreased, while HDL-C content increased in the CA and CGA groups compared to the CON group. These data suggest that CA and CGA have regulatory effects on hepatic lipid metabolism (Fig. 2M and Q). The effects of CA and CGA on the liver antioxidant system are presented in Fig. 2O, P, R, and S. Compared to the CON group, T-SOD antioxidant activity was significantly enhanced in the CA and CGA groups, but dietary CA and CGA resulted in a remarkable decrease in GSH-PX activity. In summary, as established by the above results, dietary CA and CGA can effectively mitigate liver injury by inhibiting fat deposition and enhancing some antioxidant capacities. Furthermore, comparing the CA25 group with the CGA200 group, particularly regarding TG and TC contents, the results revealed that one-eighth the dosage of CA could achieve significant effects comparable to CGA.

Fig. 2
figure 2

CA and CGA alleviated hepatic lipid accumulation and liver damage. and D liver morphology (n = 6). and E liver H&E staining (n = 6). and F Hepatic Oil Red O staining (n = 6). and J (Liver index = liver weight/body weight) × 100 (n = 10). H and K Serum ALT activity level (n = 10). and L Serum AST activity level (n = 10). and Q Liver TG, TC, HDL-C, LDL-C and VLDL-C levels (n = 10). O and R Hepatic GSH-PX, T-SOD and T-AOC activity levels (n = 10). and S Liver MDA content (n = 10). a−cMeans with different letters differ significantly (P < 0.05)

CA and CGA regulated hepatic gene expression profiles

To elucidate the underlying mechanisms by which CA and CGA mitigate fatty liver, we examined their regulatory impact on hepatic gene transcription. Notably, our integrated analysis revealed that superior therapeutic effects were observed in both the CA25 and CGA100 groups. Additionally, to conduct a more nuanced comparison at the same dosage, the CA100 group was also subjected to transcriptomic analysis. Statistics of transcriptome sequencing were indicated that the data reliable for further analysis (Table S1). As depicted in Fig. 3A, PCA demonstrated that the gene expression profiles of the CON, CA25, CA100, and CGA100 groups were largely analogous. However, in group-wise comparison, CA25, CA100, and CGA100 manifested 445, 1,634, and 412 differentially expressed genes (DEGs), respectively compared to the CON (Fig. 3B–D, Tables S2–S4). The DEGs were utilized for GO terms enrichment analysis, including Biological Process (BP), Molecular Function (MF), and Cellular Component (CC). The top 10–15 GO terms jointly enriched in CA25 and CGA100 groups were collagen-activated signaling pathway, signaling receptor activity, collagen receptor activity, cell periphery and intrinsic component of membrane (Fig. 3E–G, Tables S5–S7). Moreover, KEGG pathway analysis indicated that among the top 20 pathways, there were several pathways collectively enriched in CA25 and CGA100 groups, including biosynthesis of unsaturated fatty acids, citrate cycle (TCA cycle), fatty acid elongation, fatty acid metabolism, cell adhesion molecules (CAMs), ECM-receptor interaction, neuroactive ligand-receptor interaction, NOD-like receptor signaling pathway, and TGF-beta signaling pathway, predominantly focusing on fatty acid metabolism and immune responses (Fig. 3H, J, Tables S8, S10). Similarly, some of the same enriched pathways also existed in the CA100 group (Fig. 3I, Table S9). These results suggested that CA and CGA may operate under similar underlying mechanisms to alleviate fatty liver in laying hens.

Fig. 3
figure 3

CA and CGA regulated hepatic gene expression profiles. A PCA analysis. B–D Volcano plot of DGEs (n = 6). E–G GO term enrichment analysis of DEGs. BP: Biological Process, MF: Molecular Function, CC: Cellular Component (n = 6). H–J KEGG pathway enrichment analysis of DEGs. n = 6 hens per group

CA and CGA modulated hepatic protein expression profiles

To further elucidate the mechanisms underlying the alleviation of fatty liver by CA and CGA, proteomic analyses were conducted on CON, CA25, CA100, and CGA100 groups. As illustrated in Fig. 4A, PCA analysis revealed similar protein expression profiles across these groups. However, in group-wise analysis, CA25, CA100, and CGA100 exhibited 191, 208, and 250 differentially expressed proteins (DEPs), respectively, compared to the CON group (Fig. 4B–D, Tables S11–S13). GO term enrichment analysis mainly belonging to Biological Process (BP), Molecular Function (MF), and Cellular Component (CC). Among them in the CA25 group, the significant top 10–15 terms including phosphorylation, mitochondrial respiratory chain complex I assembly, electron transport chain, CDP-diacylglycerol biosynthetic process, bile acid secretion, regulation of insulin secretion involved in cellular response to glucose stimulus, ATP binding, phosphatidylinositol-3,5-bisphosphate phosphatase activity, propionate-CoA ligase activity, glycerol-1-phosphatase activity, fatty acid synthase activity, acetoacetate-CoA ligase activity, mitochondrion, and mitochondrial matrix, which are closely related to lipid synthesis and lipid oxidation (Fig. 4E, Table S14). Moreover, the significant top 10–15 terms of relating to lipid synthesis and lipid oxidation, including metabolic process, protein autophosphorylation, oxidoreductase activity, enzyme activator activity, and respiratory chain complex I, were enriched in the CGA100 group (Fig. 4G, Table S16). These related significant terms, including sphingomyelin catabolic process, lipid storage, maintenance of mitochondrion location, sphingomyelin phosphodiesterase, steroid receptor RNA activator RNA binding activity, sterol esterase activity, and peroxisomal membrane also illustrated in the CA100 group (Fig. 4F, Table S15). KEGG analysis indicated that DEPs in the CA25, CA100, and CGA100 groups were enriched in 51, 44, and 45 pathways (Tables S17–S19), respectively. Furthermore, these top 5 pathways were highly similar, among which ferroptosis, cell adhesion molecules, apelin signaling pathway, MAPK signaling pathway, and metabolic pathways were closely related to lipid metabolism and immunity (Fig. 4H–J, Tables S17–S19).

Fig. 4
figure 4

CA and CGA regulated hepatic protein expression profiles. A PCA analysis. BD Volcano plot of DGPs (n = 6). E–G GO term enrichment analysis of DEPs. BP: Biological Process, MF: Molecular Function, CC: Cellular Component (n = 6). HJ KEGG pathway enrichment analysis of DEPs. n = 4 hens per group

For comprehensive analysis, all expressed proteins, beyond merely DEPs, underwent GSEA enrichment analysis. Figure 5A revealed that GO terms enriched in these proteins showed downregulation of lipid synthesis pathways (including lipid droplet, carbohydrate metabolic process, fatty acid metabolic process, fatty acid biosynthetic process, cholesterol biosynthetic process, and lipid binding) and upregulation of lipid oxidation metabolism pathways (including mitochondrion, mitochondrial intermembrane space, mitochondrial matrix, tricarboxylic acid cycle, lipid metabolic process) (Table S20). This trend was consistently mirrored in KEGG pathways through GSEA enrichment analysis (Fig. 5B, Table S21). Specifically, some of the pathways, such as fatty acid biosynthetic process, lipid droplet formation, tricarboxylic acid cycle, and mitochondrial function, were highlighted in Fig. 5C–F (Tables S20 and S21).

Fig. 5
figure 5

GO term and KEGG pathway of GSEA (Gene Set Enrichment Analysis) enrichment analysis using all proteins. A GO term of GSEA enrichment analysis related to lipid metabolism. B KEGG pathway of GSEA enrichment analysis related to lipid metabolism. C–E GO term of GSEA enrichment analysis in fatty acid biosynthetic, lipid droplet, tricarboxylic acid cycle and mitochondrion. F KEGG pathway of GSEA enrichment analysis in pyruvate metabolism. A positive ES (enrichment score) value means that the term or pathway is up-regulated, whereas a negative ES value means that term or pathway is down-regulated. n = 4 hens per group

CA and CGA reduced hepatic lipid deposition by activating the ADPN-AMPK-PPARα signal pathway

As illustrated in Fig. 6A, joint transcriptomic and proteomic analysis revealed that genes involved in fatty acid biosynthesis (ACLY, ACACA, FASN, HMGCR, SCD1, and ME1) were downregulated following CA25 and CGA100 intervention. Conversely, genes associated with fatty acid oxidation and transport (PPARA, PPARG, CPT1, CD36, LPL, FABP3, and FABP4) were upregulated. These findings were corroborated by qRT-PCR analysis in CA and CGA treatment groups (Fig. 6B and C). Current studies have indicated that the activation of AMPK is crucial for the function of CGA [45, 46]. Additionally, adiponectin receptors AdipoR1 and AdipoR2 serve as upstream activators of AMPK and PPARα, respectively [47]. Interestingly, the genes expression of AMPK, AdipoR1, and AdipoR2 were increased following CA and CGA treatment (Fig. 6D and E). Furthermore, protein interaction analyses showed strong relationships among these genes (Fig. 6F). Collectively, these results suggested that CA and CGA might reduce hepatic lipid accumulation through activation of the ADPN-AMPK-PPARα signaling pathway.

Fig. 6
figure 6

Comprehensive analysis of liver gene transcription and protein. A The relative genes transcription and protein expression levels of lipid synthesis, lipid transport and oxidation. B–E The relative mRNA expression levels of lipid synthesis, lipid transport and oxidation (n = 6). F Protein interaction analysis of target genes by STRING. a−cMeans with different letters differ significantly (P < 0.05)

Discussion

In the current study, we found that dietary CA and CGA can ameliorate hepatic lipid metabolism disorder and steatosis, thereby improving the egg production performance of laying hens.

Our findings suggest that laying hens in the late laying period showed typical symptoms of fatty liver in CON group, characterized by the emergence of yellow color and elevated hepatic lipid storage which are in line with previous literature [48, 49]. Here, we showed that CA and CGA ameliorated the typical symptoms of fatty liver when introduced into the diet. The incidence of fatty liver in laying hens adversely impacts both laying performance and egg quality, posing a threat to their health and productivity [2, 10]. Here, our results indicated that treatment with CA and CGA effectively improved the production performance and egg quality of laying hens. These therapeutic effects largely meet the recommended criteria for the treatment of fatty liver [50,51,52].

The pathological and physiological mechanisms underlying fatty liver in laying hens remain poorly understood. However, studies have shown similarities to mammalian MAFLD, all of which involve in lipid metabolism disorders, hepatic steatosis, inflammatory responses, and oxidative stress processes [53,54,55,56]. Dyslipidemia has been widely postulated to play a primary and fundamental role in the pathogenesis of fatty liver. It is usually characterized by elevated levels of TG, TC, VLDL-C and decreased levels of HDL-C [5, 57]. Notably, addressing dyslipidemia has been demonstrated to be effective in preventing or delaying the progression of fatty liver [58]. A previous report indicated that CA and CGA supplementation inhibits the progression of dyslipidemia in HFD-induced mice [59, 60], which showed similar effects in laying hens in our study that the administration of CA and CGA resulted in a significant reduction in serum levels of TG and TC. Plasma enzyme activities such as ALT and AST, which are released into the bloodstream upon hepatocyte injury, were employed to assess liver damage [44, 61]. As anticipated, we observed significant reductions in AST and ALT activities following the administration of CA25 and CGA100. Lipid metabolism disorder in the liver is a hallmark of fatty liver disease, leading to elevated TG and TC levels, and decreased HDL-C [55, 56]. Interestingly, Jia et al. [62] suggested that lipoprotein density correlates with cholesterol transport; HDL-C and VLDL-C are protective against hepatic lipid metabolism, whereas LDL-C is detrimental. Here, our results demonstrated that both CA25 and CGA100 significantly lowered TG and TC levels in hepatic tissues. Furthermore, our study revealed that LDL-C levels decreased in both the CA and CGA groups, and HDL-C levels increased significantly in the CA treatment group. Interestingly, we found that these effects did not exhibit a linear dose–response relationship; improvements were not significant when CA exceeded 25 mg/kg and remained non-additive when CGA exceeded 200 mg/kg. Interestingly, the egg laying performance of the CA25 group was better than that of the high-dose CA group. The liver is universally acknowledged as the pivotal organ for lipid biosynthesis and metabolism. In the context of oogenesis, yolk lipids are predominantly synthesized within hepatocytes. Impairments in hepatic metabolic function led to lipid accumulation, resulting in steatosis, which subsequently impedes vitellogenesis, thereby reducing both egg yield and quality. Our findings indicated that the transcriptomic analysis of lipid synthesis-related genes revealed a more pronounced down-regulation in the CA25 group, whereas certain lipid synthesis genes within the CA100 group exhibited up-regulation, notably including key enzymes such as ACLY, ACACA, and FASN. These results suggest that the CA100 regimen may be less effective than CA25 in mitigating fatty liver disease, which may explain why the egg production performance of the CA25 group is better than that of the high-dose CA treatment group.

Persistent lipid metabolism disorders can induce liver lipid peroxidation and subsequently lead to ROS accumulation [63, 64], resulting in diminished antioxidant capacity, culminating in oxidative damage to hepatic cells [61]. Laying hens with fatty liver usually exhibit elevated ROS and MDA contents and significantly lowered activities of GSH-PX, T-SOD, and T-AOC [65, 66]. Our study demonstrated that CA25 and CA50 additives increased the activity of GSH-PX, but notably increased MDA content in serum. Nonetheless, dietary CA and CGA both significantly enhanced antioxidant activity of T-SOD in liver, indicating a general positive effect of CA and CGA administration on liver antioxidant capacity.

CA and CGA (catechin and esters of quinic acid) share similar structures and pharmacological actions [67, 68]. CGA can be hydrolyzed into CA, which is one of the major active metabolites of CGA both in vivo and in vitro [69, 70]. On average, CGA absorption is 33%, while CA absorption is as high as 95% in rats or humans [33, 34]. Our study demonstrated that one-fourth the dosage of CA (25 mg/kg) exerted an equivalent improvement effect on laying hens compared to CGA (100 mg/kg). Based on these findings, we speculated that one-third, or even less, of CGA and almost all of CA were absorbed in the laying hens. This implies that only a small part of CGA enters into the bloodstream, with most being excreted in the stool. Research indicates that the absorption and utilization of CGA largely depend on its metabolism by colonic gut microflora in rats [32]. However, it is unclear whether the same is true for laying hens, especially given the underdeveloped colon in laying hens.

Liver transcriptomics and proteomics analysis revealed that fatty acid biosynthesis pathways were inhibited, whereas fatty acid transport and oxidation pathways were enhanced in our data. This finding aligns with the mechanism for treating fatty liver [66]. Hepatic lipogenesis is primarily regulated by the transcription factors of SREBP1 and ChREBP1, which activate downstream lipogenic genes, including FASN (the key enzyme for catalyzing fatty acid synthesis), ACACA (the rate-limiting enzyme for fatty acid synthesis), and SCD1 (the major enzyme for catalyzing lipogenesis), thereby contributing to hepatic lipid accumulation [71,72,73,74]. Conversely, activation of the transcription factor PPARA (PPARα) promotes the expression of its downstream genes, such as CPT1A, CD36, FABP3, FABP4, and LPL, enhancing lipid transport and oxidative utilization in the liver [75]. Consistent with these findings, our results demonstrated that CA and CGA inhibited SREBP1 and ChREBP1, and downregulated the gene expression of ACLY, ACACA, FASN, and SCD1. Additionally, PPARα, along with its downstream target genes (CPT1A, CD36, FABP3, FABP4, and LPL), were upregulated. Studies have shown that the FXR/LXRα receptor is a crucial mediator in the liver, up-regulation of FXR and down-regulation of LXRα can alleviate high-fat diet-induced MAFLD [76, 77]. In line with literature, our results exhibited that FXR and LXRα were up-regulated simultaneously. These findings collectively suggest that the effect of CA and CGA against fatty liver may be attributed to diminishing lipid synthesis and accelerating fatty acid oxidation.

Recent studies have demonstrated that the AMPK signaling pathways play crucial regulatory roles in CGA-induced benefits [20, 45]. It was also found that activated AMPK suppresses the expression of ACACA and FASN, which may reduce fatty acid synthesis, serum triglycerides, and cholesterol levels, thereby alleviating hepatic steatosis [45]. Peroxisome proliferator-activated receptors (PPARs) are crucial in the pathogenesis of non-alcoholic fatty liver disease (NAFLD), PPARα primarily enhances fatty acid oxidation in the liver, while PPARG (PPARγ) facilitates lipid transport [78]. Here, our results revealed that CA and CGA promoted the mRNA expression of AMPK and PPARα in liver of laying hens. In the current literature on the upstream activation proteins of AMPK and PPARα, it is well-established that adiponectin binds to adiponectin receptors AdipoR1 and AdipoR2 and exerts anti-lipid effects in obesity via activation of AMPK and PPARα pathways, respectively [47]. At present, there is no report on the regulation of adiponectin and its receptor AdipoR1/2 by CA and CGA. Our results observed that CA and CGA up-regulated adiponectin and its receptor AdipoR1/2, along with the up-regulation of AMPK and PPARα. According to these results, we initially inferred a potential role of adiponectin in the activation of the AMPK-PPARα pathway by CA and CGA. However, how CA and CGA regulate the ADPN-AMPK-PPARα pathway, especially the regulation of adiponectin and AdiporR1/2 by CA or CGA, still needs to be further clarified in vitro hepatocyte experiments.

Additionally, this study will offer theoretical insights into the efficient utilization of CGA (including its isomers, neochlorogenic acid and cryptochlorogenic acid). For instance, these compounds can be directionally transformed into CA through methods such as microbial enzymatic hydrolysis, in vitro hydrolysis, and chemical structural modification, thereby enhancing their utilization efficiency.

Conclusions

Our study shows that CA and CGA can be two outstanding feed additives for the prevention and mitigation of fatty liver by regulating lipid synthesis and transport metabolism, and ameliorating hepatic lipid metabolism disorder and steatosis. Moreover, CA may be one of the principal active metabolites of CGA in laying hens. CA also represents a more economical and efficient additive for alleviating fatty liver compared to CGA. Our work provides foundational evidence for the potential therapeutic efficacy of CA and CGA in treating fatty liver and related metabolic disorders in laying hens via the activation of the ADPN-AMPK-PPARα pathway, and offers insights into the efficient utilization of CGA through targeted modification to CA.

Data availability

The raw sequencing data have been deposited in the China National Center for Bioinformation (CNCB) of BIG Submission Portal (BIG Sub) with the accession number CRA020037 (transcriptome) and OMIX007802 (proteome).

Abbreviations

ACACA:

Acetyl-CoA carboxylase

ACOX1:

Acyl-CoA oxidase 1

ADFI:

Average daily feed intake

ADPN:

Adiponectin

ALT:

Alanine aminotransferase

AMPK:

AMP-activated protein kinase

AST:

Aspartate aminotransferase

CA:

Caffeic acid

CGA:

Chlorogenic acid

ChREBP1:

Carbohydrate response element binding protein 1

CoA:

Coenzyme A

CON:

Control

CPT1A:

Carnitine palmitoyl transferase 1A

DEG:

Differentially expressed gene

DEP:

Differentially expressed protein

FABP3:

Fatty acid binding protein 3

FABP4:

Fatty acid binding protein 4

FASN:

Fatty acid synthase

FCR:

Feed conversion rate

GO:

Gene Ontology

GSH-PX:

Glutathione peroxidase

HDL-C:

High-density lipoprotein cholesterol

H&E:

Hematoxylin-eosin

KEGG:

Kyoto Encyclopedia of Genes and Genomes

LDL-C:

Low-density lipoprotein cholesterol

LPL:

Lipoprotein lipase

MAFLD:

Metabolically associated fatty liver disease

MDA:

Malondialdehyde

ME1:

Malic enzyme 1

PCA:

Principal component analysis

PPARA/PPARα:

Peroxisome proliferator-activated receptor α

PPARG /PPARγ:

Peroxisome proliferator-activated receptor γ

ROS:

Reactive oxygen species

SCD1:

Stearoyl-CoA desaturase 1

SREBP-1c:

Sterol regulatory element binding proteins-1c

T-AOC:

Total antioxidant capacity

TC:

Total cholesterol

TG:

Triglyceride

T-SOD:

Total superoxide dismutase

VLDL-C:

Very low-density lipoprotein cholesterol

References

  1. Arulnathan V, Turner I, Bamber N, Ferdous J, Grassauer F, Doyon M, et al. A systematic review of potential productivity, egg quality, and animal welfare implications of extended lay cycles in commercial laying hens in Canada. Poult Sci. 2024;103(4):103475. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.psj.2024.103475.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Butler EJ. Fatty liver diseases in the domestic fowl-a review. Avian Pathol. 1976;5(1):1–14. https://doiorg.publicaciones.saludcastillayleon.es/10.1080/03079457608418164.

    Article  CAS  PubMed  Google Scholar 

  3. Lee K, Flegal CJ, Wolford JH. Factors affecting liver fat accumulation and liver hemorrhages associated with fatty liver-hemorrhagic syndrome in laying chickens. Poult Sci. 1975;54(2):374–80. https://doiorg.publicaciones.saludcastillayleon.es/10.3382/ps.0540374.

    Article  CAS  PubMed  Google Scholar 

  4. Chen W, Shi Y, Li G, Huang C, Zhuang Y, Shu B, et al. Preparation of the peroxisome proliferator-activated receptor alpha polyclonal antibody: its application in fatty liver hemorrhagic syndrome. Int J Biol Macromol. 2021;182:179–86. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ijbiomac.2021.04.018.

    Article  CAS  PubMed  Google Scholar 

  5. Harms RH, Simpson CF. Serum and body characteristics of laying hens with fatty liver syndrome. Poult Sci. 1979;58(6):1644–6. https://doiorg.publicaciones.saludcastillayleon.es/10.3382/ps.0581644.

    Article  CAS  PubMed  Google Scholar 

  6. Shini A, Shini S, Bryden WL. Fatty liver haemorrhagic syndrome occurrence in laying hens: impact of production system. Avian Pathol. 2019;48(1):25–34. https://doiorg.publicaciones.saludcastillayleon.es/10.1080/03079457.2018.1538550.

    Article  CAS  PubMed  Google Scholar 

  7. Tan X, Liu R, Zhang Y, Wang X, Wang J, Wang H, et al. Integrated analysis of the methylome and transcriptome of chickens with fatty liver hemorrhagic syndrome. BMC Genomics. 2021;22:8. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12864-020-07305-3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Li L, Wang Y, Wang H, Yang Y, Ma H. Protective effects of genistein on the production performance and lipid metabolism disorders in laying hens with fatty liver hemorrhagic syndrome by activation of the GPER-AMPK signaling pathways. J Anim Sci. 2023;101:skad197. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/jas/skad197.

  9. Miao YF, Gao XN, Xu DN, Li MC, Gao ZS, Tang ZH, et al. Protective effect of the new prepared Atractylodes macrocephala Koidz polysaccharide on fatty liver hemorrhagic syndrome in laying hens. Poult Sci. 2021;100(2):938–48. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.psj.2020.11.036.

    Article  CAS  PubMed  Google Scholar 

  10. Wolford JH, Polin D. Lipid accumulation and hemorrhage in livers of laying chickens. A study on fatty liver-hemorrhagic syndrome (FLHS). Poult Sci. 1972;51(5):1707–13. https://doiorg.publicaciones.saludcastillayleon.es/10.3382/ps.0511707.

  11. Choi YI, Ahn HJ, Lee BK, Oh ST, An BK, Kang CW. Nutritional and hormonal induction of fatty liver syndrome and effects of dietary lipotropic factors in egg-type male chicks. Asian-Australas J Anim Sci. 2012;25(8):1145–52. https://doiorg.publicaciones.saludcastillayleon.es/10.5713/ajas.2011.11418.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Mete A, Giannitti F, Barr B, Woods L, Anderson M. Causes of mortality in backyard chickens in northern California:2007–2011. Avian Dis. 2013;57(2):311–5. https://doiorg.publicaciones.saludcastillayleon.es/10.1637/10382-092312-Case.1.

    Article  PubMed  Google Scholar 

  13. Rozenboim I, Mahato J, Cohen NA, Tirosh O. Low protein and high-energy diet: a possible natural cause of fatty liver hemorrhagic syndrome in caged White Leghorn laying hens. Poult Sci. 2016;95(3):612–21. https://doiorg.publicaciones.saludcastillayleon.es/10.3382/ps/pev367.

    Article  CAS  PubMed  Google Scholar 

  14. Trott KA, Giannitti F, Rimoldi G, Hill A, Woods L, Barr B, et al. Fatty liver hemorrhagic syndrome in the backyard chicken: a retrospective histopathologic case series. Vet Pathol. 2014;51(4):787–95. https://doiorg.publicaciones.saludcastillayleon.es/10.1177/0300985813503569.

    Article  CAS  PubMed  Google Scholar 

  15. Peng G, Huang E, Ruan J, Huang L, Liang H, Wei Q, et al. Effects of a high energy and low protein diet on hepatic and plasma characteristics and Cidea and Cidec mRNA expression in liver and adipose tissue of laying hens with fatty liver hemorrhagic syndrome. Anim Sci J. 2019;90(2):247–54. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/asj.13140.

    Article  CAS  PubMed  Google Scholar 

  16. Huang J, Zhang Y, Zhou Y, Zhang Z, Xie Z, Zhang J, et al. Green tea polyphenols alleviate obesity in broiler chickens through the regulation of lipid-metabolism-related genes and transcription factor expression. J Agric Food Chem. 2013;61(36):8565–72. https://doiorg.publicaciones.saludcastillayleon.es/10.1021/jf402004x.

    Article  CAS  PubMed  Google Scholar 

  17. Nafikov RA, Beitz DC. Carbohydrate and lipid metabolism in farm animals. J Nutr. 2007;137(3):702–5. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/jn/137.3.702.

    Article  CAS  PubMed  Google Scholar 

  18. Nabavi SF, Tejada S, Setzer WN, Gortzi O, Sureda A, Braidy N, et al. Chlorogenic acid and mental diseases: from chemistry to medicine. Curr Neuropharmacol. 2017;15(4):471–9. https://doiorg.publicaciones.saludcastillayleon.es/10.2174/1570159X14666160325120625.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Plazas M, Prohens J, Cunat AN, Vilanova S, Gramazio P, Herraiz FJ, et al. Reducing capacity, chlorogenic acid content and biological activity in a collection of scarlet (Solanum aethiopicum) and gboma (S. macrocarpon) eggplants. Int J Mol Sci. 2014;15(10):17221–41. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms151017221.

  20. Lu H, Tian Z, Cui Y, Liu Z, Ma X. Chlorogenic acid: a comprehensive review of the dietary sources, processing effects, bioavailability, beneficial properties, mechanisms of action, and future directions. Compr Rev Food Sci Food Saf. 2020;19(6):3130–58. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/1541-4337.12620.

    Article  CAS  PubMed  Google Scholar 

  21. Mansour A, Mohajeri-Tehrani MR, Samadi M, Qorbani M, Merat S, Adibi H, et al. Effects of supplementation with main coffee components including caffeine and/or chlorogenic acid on hepatic, metabolic, and inflammatory indices in patients with non-alcoholic fatty liver disease and type 2 diabetes: a randomized, double-blind, placebo-controlled, clinical trial. Nutr J. 2021;20(1):35. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12937-021-00694-5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Rafiei H, Omidian K, Bandy B. Comparison of dietary polyphenols for protection against molecular mechanisms underlying nonalcoholic fatty liver disease in a cell model of steatosis. Mol Nutr Food Res. 2017;61(9):1600781. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/mnfr.201600781.

  23. Velazquez AM, Roglans N, Bentanachs R, Gene M, Sala-Vila A, Lazaro I, et al. Effects of a low dose of caffeine alone or as part of a green coffee extract, in a rat dietary model of lean Non-Alcoholic fatty liver disease without inflammation. Nutrients. 2020;12(11):3240. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/nu12113240.

  24. Veronese N, Notarnicola M, Cisternino AM, Reddavide R, Inguaggiato R, Guerra V, et al. Coffee intake and liver steatosis: a population study in a Mediterranean Area. Nutrients. 2018;10(1):89. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/nu10010089.

  25. Kim M, Yoo G, Randy A, Kim HS, Nho CW. Chicoric acid attenuate a nonalcoholic steatohepatitis by inhibiting key regulators of lipid metabolism, fibrosis, oxidation, and inflammation in mice with methionine and choline deficiency. Mol Nutr Food Res. 2017;61(5):1600632. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/mnfr.201600632.

  26. Vergani L, Vecchione G, Baldini F, Grasselli E, Voci A, Portincasa P, et al. Polyphenolic extract attenuates fatty acid-induced steatosis and oxidative stress in hepatic and endothelial cells. Eur J Nutr. 2018;57(5):1793–805. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00394-017-1464-5.

    Article  CAS  PubMed  Google Scholar 

  27. Mu HN, Zhou Q, Yang RY, Tang WQ, Li HX, Wang SM, et al. Caffeic acid prevents non-alcoholic fatty liver disease induced by a high-fat diet through gut microbiota modulation in mice. Food Res Int. 2021;143:110240. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.foodres.2021.110240.

  28. Watanabe S, Takahashi T, Ogawa H, Uehara H, Tsunematsu T, Baba H, et al. Daily coffee intake inhibits pancreatic beta cell damage and nonalcoholic steatohepatitis in a mouse model of spontaneous metabolic syndrome, Tsumura-Suzuki obese diabetic mice. Metab Syndr Relat Disord. 2017;15(4):170–7. https://doiorg.publicaciones.saludcastillayleon.es/10.1089/met.2016.0114.

    Article  CAS  PubMed  Google Scholar 

  29. Liao CC, Ou TT, Huang HP, Wang CJ. The inhibition of oleic acid induced hepatic lipogenesis and the promotion of lipolysis by caffeic acid via up-regulation of AMP-activated kinase. J Sci Food Agric. 2014;94(6):1154–62. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/jsfa.6386.

    Article  CAS  PubMed  Google Scholar 

  30. Namvarjah F, Shokri-Afra H, Moradi-Sardareh H, Khorzoughi RB, Pasalar P, Panahi G, et al. Chlorogenic acid improves anti-lipogenic activity of metformin by positive regulating of AMPK signaling in HepG2 cells. Cell Biochem Biophys. 2022;80(3):537–45. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s12013-022-01077-1.

    Article  CAS  PubMed  Google Scholar 

  31. Xu M, Yang L, Zhu Y, Liao M, Chu L, Li X, et al. Collaborative effects of chlorogenic acid and caffeine on lipid metabolism via the AMPKα-LXRα/SREBP-1c pathway in high-fat diet-induced obese mice. Food Funct. 2019;10(11):7489–97. https://doiorg.publicaciones.saludcastillayleon.es/10.1039/c9fo00502a.

    Article  CAS  PubMed  Google Scholar 

  32. Gonthier MP, Verny MA, Besson C, Remesy C, Scalbert A. Chlorogenic acid bioavailability largely depends on its metabolism by the gut microflora in rats. J Nutr. 2003;133(6):1853–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/jn/133.6.1853.

    Article  CAS  PubMed  Google Scholar 

  33. Lafay S, Morand C, Manach C, Besson C, Scalbert A. Absorption and metabolism of caffeic acid and chlorogenic acid in the small intestine of rats. Br J Nutr. 2006;96(1):39–46. https://doiorg.publicaciones.saludcastillayleon.es/10.1079/bjn20061714.

    Article  CAS  PubMed  Google Scholar 

  34. Stalmach A, Steiling H, Williamson G, Crozier A. Bioavailability of chlorogenic acids following acute ingestion of coffee by humans with an ileostomy. Arch Biochem Biophys. 2010;501(1):98–105. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.abb.2010.03.005.

    Article  CAS  PubMed  Google Scholar 

  35. Lafay S, Gil-Izquierdo A, Manach C, Morand C, Besson C, Scalbert A. Chlorogenic acid is absorbed in its intact form in the stomach of rats. J Nutr. 2006;136(5):1192–7. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/jn/136.5.1192.

    Article  CAS  PubMed  Google Scholar 

  36. Kim HM, Kim Y, Lee ES, Huh JH, Chung CH. Caffeic acid ameliorates hepatic steatosis and reduces ER stress in high fat diet-induced obese mice by regulating autophagy. Nutrition. 2018;55–56:63–70. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.nut.2018.03.010.

    Article  CAS  PubMed  Google Scholar 

  37. Zhang Y, Wang Y, Chen D, Yu B, Zheng P, Mao X, et al. Dietary chlorogenic acid supplementation affects gut morphology, antioxidant capacity and intestinal selected bacterial populations in weaned piglets. Food Funct. 2018;9(9):4968–78. https://doiorg.publicaciones.saludcastillayleon.es/10.1039/c8fo01126e.

    Article  CAS  PubMed  Google Scholar 

  38. Zhang K, Li X, Zhao J, Wang Y, Hao X, Liu K, et al. Protective effects of chlorogenic acid on the meat quality of oxidatively stressed broilers revealed by integrated metabolomics and antioxidant analysis. Food Funct. 2022;13(4):2238–52. https://doiorg.publicaciones.saludcastillayleon.es/10.1039/d1fo03622j.

    Article  CAS  PubMed  Google Scholar 

  39. Chen S, Zhou Y, Chen Y, Gu J. Fastp: an ultra-fast all-in-one FASTQ preprocessor. Bioinformatics. 2018;34(17):i884–90. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/bioinformatics/bty560.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Kim D, Langmead B, Salzberg SL. HISAT: a fast spliced aligner with low memory requirements. Nat Methods. 2015;12(4):357–60. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nmeth.3317.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Liao Y, Smyth GK, Shi W. FeatureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics. 2014;30(7):923–30. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/bioinformatics/btt656.

    Article  CAS  PubMed  Google Scholar 

  42. Guzman UH, Martinez-Val A, Ye Z, Damoc E, Arrey TN, Pashkova A, et al. Ultra-fast label-free quantification and comprehensive proteome coverage with narrow-window data-independent acquisition. Nat Biotechnol. 2024. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41587-023-02099-7.

    Article  PubMed  PubMed Central  Google Scholar 

  43. von Mering C, Huynen M, Jaeggi D, Schmidt S, Bork P, Snel B. STRING: a database of predicted functional associations between proteins. Nucleic Acids Res. 2003;31(1):258–61. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/nar/gkg034.

    Article  CAS  Google Scholar 

  44. Dong Y, Zhang J, Gao Z, Zhao H, Sun G, Wang X, et al. Characterization and anti-hyperlipidemia effects of enzymatic residue polysaccharides from Pleurotus ostreatus. Int J Biol Macromol. 2019;129:316–25. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ijbiomac.2019.01.164.

    Article  CAS  PubMed  Google Scholar 

  45. Ong KW, Hsu A, Tan BK. Anti-diabetic and anti-lipidemic effects of chlorogenic acid are mediated by ampk activation. Biochem Pharmacol. 2013;85(9):1341–51. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.bcp.2013.02.008.

    Article  CAS  PubMed  Google Scholar 

  46. Ong KW, Hsu A, Tan BK. Chlorogenic acid stimulates glucose transport in skeletal muscle via AMPK activation: a contributor to the beneficial effects of coffee on diabetes. PLoS ONE. 2012;7(3):e32718. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pone.0032718.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Okada-Iwabu M, Yamauchi T, Iwabu M, Honma T, Hamagami K, Matsuda K, et al. A small-molecule AdipoR agonist for type 2 diabetes and short life in obesity. Nature. 2013;503(7477):493–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nature12656.

    Article  CAS  PubMed  Google Scholar 

  48. Lv Z, Xing K, Li G, Liu D, Guo Y. Dietary genistein alleviates lipid metabolism disorder and inflammatory response in laying hens with fatty liver syndrome. Front Physiol. 2018;9:1493. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fphys.2018.01493.

  49. Meng J, Ma N, Liu H, Liu J, Liu J, Wang J, et al. Untargeted and targeted metabolomics profiling reveals the underlying pathogenesis and abnormal arachidonic acid metabolism in laying hens with fatty liver hemorrhagic syndrome. Poult Sci. 2021;100(9):101320. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.psj.2021.101320.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Diehl AM, Day C. Cause, pathogenesis, and treatment of nonalcoholic steatohepatitis. N Engl J Med. 2017;377(21):2063–72. https://doiorg.publicaciones.saludcastillayleon.es/10.1056/NEJMra1503519.

    Article  CAS  PubMed  Google Scholar 

  51. Llovet JM, Willoughby CE, Singal AG, Greten TF, Heikenwalder M, El-Serag HB, et al. Nonalcoholic steatohepatitis-related hepatocellular carcinoma: pathogenesis and treatment. Nat Rev Gastroenterol Hepatol. 2023;20(8):487–503. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41575-023-00754-7.

    Article  CAS  PubMed  Google Scholar 

  52. Yang X, Li D, Zhang M, Feng Y, Jin X, Liu D, et al. Ginkgo biloba extract alleviates fatty liver hemorrhagic syndrome in laying hens via reshaping gut microbiota. J Anim Sci Biotechnol. 2023;14:97. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s40104-023-00900-w.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Cheng X, Liu J, Zhu Y, Guo X, Liu P, Zhang C, et al. Molecular cloning, characterization, and expression analysis of TIPE1 in chicken (Gallus gallus): its applications in fatty liver hemorrhagic syndrome. Int J Biol Macromol. 2022;207:905–16. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ijbiomac.2022.03.177.

    Article  CAS  PubMed  Google Scholar 

  54. Eslam M, Sanyal AJ, George J. MAFLD: a consensus-driven proposed nomenclature for metabolic associated fatty liver disease. Gastroenterology. 2020;158(7):1999–2014. https://doiorg.publicaciones.saludcastillayleon.es/10.1053/j.gastro.2019.11.312.

    Article  CAS  PubMed  Google Scholar 

  55. Kim H, Lee DS, An TH, Park HJ, Kim WK, Bae KH, et al. Metabolic spectrum of liver failure in type 2 diabetes and obesity: from NAFLD to NASH to HCC. Int J Mol Sci. 2021;22(9):4495. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms22094495.

  56. Vluggens A, Reddy JK. Nuclear receptors and transcription factors in the development of fatty liver disease. Curr Drug Metab. 2012;13(10):1422–35. https://doiorg.publicaciones.saludcastillayleon.es/10.2174/138920012803762710.

    Article  CAS  PubMed  Google Scholar 

  57. Katsiki N, Mikhailidis DP, Mantzoros CS. Non-alcoholic fatty liver disease and dyslipidemia: an update. Metabolism. 2016;65(8):1109–23. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.metabol.2016.05.003.

    Article  CAS  PubMed  Google Scholar 

  58. Adeyanju OA, Badejogbin OC, Areola DE, Olaniyi KS, Dibia C, Soetan OA, et al. Sodium butyrate arrests pancreato-hepatic synchronous uric acid and lipid dysmetabolism in high fat diet fed Wistar rats. Biomed Pharmacother. 2021;133:10994. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.biopha.2020.110994.

  59. Cho AS, Jeon SM, Kim MJ, Yeo J, Seo KI, Choi MS, et al. Chlorogenic acid exhibits anti-obesity property and improves lipid metabolism in high-fat diet-induced-obese mice. Food Chem Toxicol. 2010;48(3):937–43. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.fct.2010.01.003.

    Article  CAS  PubMed  Google Scholar 

  60. Vitaglione P, Mazzone G, Lembo V, D'Argenio G, Rossi A, Guido M, et al. Coffee prevents fatty liver disease induced by a high-fat diet by modulating pathways of the gut-liver axis. J Nutr Sci. 2019;8:e15. https://doiorg.publicaciones.saludcastillayleon.es/10.1017/jns.2019.10.

  61. Yao Y, Wang H, Yang Y, Jiang Z, Ma H. Dehydroepiandrosterone activates the GPER-mediated AMPK signaling pathway to alleviate the oxidative stress and inflammatory response in laying hens fed with high-energy and low-protein diets. Life Sci. 2022;308:120926. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.lfs.2022.120926.

  62. Jia B, Zou Y, Han X, Bae JW, Jeon CO. Gut microbiome-mediated mechanisms for reducing cholesterol levels: implications for ameliorating cardiovascular disease. Trends Microbiol. 2023;31(1):76–91. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.tim.2022.08.003.

    Article  CAS  PubMed  Google Scholar 

  63. Chen Z, Tian R, She Z, Cai J, Li H. Role of oxidative stress in the pathogenesis of nonalcoholic fatty liver disease. Free Radic Biol Med. 2020;152:116–41. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.freeradbiomed.2020.02.025.

    Article  CAS  PubMed  Google Scholar 

  64. Shin GC, Lee HM, Kim N, Yoo SK, Park HS, Choi LS, et al. Paraoxonase-2 contributes to promoting lipid metabolism and mitochondrial function via autophagy activation. Sci Rep. 2022;12:21483. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41598-022-25802-1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Liu M, Kang Z, Cao X, Jiao H, Wang X, Zhao J, et al. Prevotella and succinate treatments altered gut microbiota, increased laying performance, and suppressed hepatic lipid accumulation in laying hens. J Anim Sci Biotechnol. 2024;15:26. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s40104-023-00975-5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Miao S, Mu T, Li R, Li Y, Zhao W, Li J, et al. Coated sodium butyrate ameliorates high-energy and low-protein diet induced hepatic dysfunction via modulating mitochondrial dynamics, autophagy and apoptosis in laying hens. J Anim Sci Biotechnol. 2024;15:15. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s40104-023-00980-8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Agunloye OM, Oboh G, Bello GT, Oyagbemi AA. Caffeic and chlorogenic acids modulate altered activity of key enzymes linked to hypertension in cyclosporine-induced hypertensive rats. J Basic Clin Physiol Pharmacol. 2020;32(3):169–77. https://doiorg.publicaciones.saludcastillayleon.es/10.1515/jbcpp-2019-0360.

    Article  CAS  PubMed  Google Scholar 

  68. Naveed M, Hejazi V, Abbas M, Kamboh AA, Khan GJ, Shumzaid M, et al. Chlorogenic acid (CGA): a pharmacological review and call for further research. Biomed Pharmacother. 2018;97:67–74. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.biopha.2017.10.064.

  69. Chang Y, Huang K, Yang F, Gao Y, Zhang Y, Li S, et al. Metabolites of chlorogenic acid and its isomers: Metabolic pathways and activities for ameliorating myocardial hypertrophy. J Funct Foods. 2022;96:105216. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jff.2022.105216.

  70. Sato Y, Itagaki S, Kurokawa T, Ogura J, Kobayashi M, Hirano T, et al. In vitro and in vivo antioxidant properties of chlorogenic acid and caffeic acid. Int J Pharm. 2011;403(1–2):136–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ijpharm.2010.09.035.

    Article  CAS  PubMed  Google Scholar 

  71. Han J, Li E, Chen L, Zhang Y, Wei F, Liu J, et al. The CREB coactivator CRTC2 controls hepatic lipid metabolism by regulating SREBP1. Nature. 2015;524(7564):243–6. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nature14557.

    Article  CAS  PubMed  Google Scholar 

  72. Regnier M, Carbinatti T, Parlati L, Benhamed F, Postic C. The role of ChREBP in carbohydrate sensing and NAFLD development. Nat Rev Endocrinol. 2023;19(6):336–49. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41574-023-00809-4.

    Article  CAS  PubMed  Google Scholar 

  73. Shimomura I, Bashmakov Y, Ikemoto S, Horton JD, Brown MS, Goldstein JL. Insulin selectively increases SREBP-1c mRNA in the livers of rats with streptozotocin-induced diabetes. Proc Natl Acad Sci U S A. 1999;96(24):13656–61. https://doiorg.publicaciones.saludcastillayleon.es/10.1073/pnas.96.24.13656.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Yang Q, Mao Y, Wang J, Yu H, Zhang X, Pei X, et al. Gestational bisphenol a exposure impairs hepatic lipid metabolism by altering mTOR/CRTC2/SREBP1 in male rat offspring. Hum Exp Toxicol. 2022;41:774819204. https://doiorg.publicaciones.saludcastillayleon.es/10.1177/09603271221129852.

  75. Sanchez-Macedo N, Feng J, Faubert B, Chang N, Elia A, Rushing EJ, et al. Depletion of the novel p53-target gene carnitine palmitoyltransferase 1C delays tumor growth in the neurofibromatosis type I tumor model. Cell Death Differ. 2013;20(4):659–68. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/cdd.2012.168.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Ma S, Pang X, Tian S, Sun J, Hu Q, Li X, et al. The protective effects of sulforaphane on high-fat diet-induced metabolic associated fatty liver disease in mice via mediating the FXR/LXRα pathway. Food Funct. 2022;13(24):12966–82. https://doiorg.publicaciones.saludcastillayleon.es/10.1039/d2fo02341e.

    Article  CAS  PubMed  Google Scholar 

  77. Schmitt J, Kong B, Stieger B, Tschopp O, Schultze SM, Rau M, et al. Protective effects of farnesoid X receptor (FXR) on hepatic lipid accumulation are mediated by hepatic FXR and independent of intestinal FGF15 signal. Liver Int. 2015;35(4):1133–44. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/liv.12456.

    Article  CAS  PubMed  Google Scholar 

  78. Chen H, Tan H, Wan J, Zeng Y, Wang J, Wang H, et al. PPAR-γ signaling in nonalcoholic fatty liver disease: Pathogenesis and therapeutic targets. Pharmacol Ther. 2023;245:108391. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.pharmthera.2023.108391.

Download references

Acknowledgements

We would like to express our gratitude to members of Animal Products Quality Research Term for sample collection.

Funding

This work was supported by National Key R&D Program of China (2023YFD1301200), China Agriculture Research Systems (CARS-40-K11), Beijing Agriculture Innovation Consortium (BAIC06-2024-G05), Strategic Priority Research Program of the National Center of Technology Innovation for Pigs (NCTIP-XD/C08) and The Chinese Academy of Agricultural Science and Technology Innovation Project (ASTIP-IAS-12).

Author information

Authors and Affiliations

Authors

Contributions

Conception and design: YCQ, HYZ, JMZ, QYZ and WJT. Collection and assembly of data: WJT, HW, YYY and CHT. Data analysis and interpretation: WJT, HYZ, CHT and JMZ. Manuscript writing: WJT and HYZ. Manuscript revising: HYZ, JMZ, GBG and YCQ. Final approval of manuscript: All authors.

Corresponding authors

Correspondence to Huiyan Zhang or Yuchang Qin.

Ethics declarations

Ethics approval and consent to participate

All experimental procedures were reviewed and approved by the Experimental Animal Welfare and Ethical Board of the Institute of Animal Science (IAS), Chinese Academy of Agricultural Sciences (CAAS), Beijing, China (Approval number: IAS2023-114).

Consent for publication

All of the authors have approved the final version of the manuscript and agreed with this submission to the Journal of Animal Science and Biotechnology.

Competing interests

The authors declare that they have no competing interests.

Supplementary Information

40104_2025_1175_MOESM1_ESM.xlsx

Additional file 1: Table S1 Statistics of transcriptome sequencing. Table S2–S4 Differentially expressed genes between CA25 group, CA100 group, CGA1000 group and CON group. Table S5–S7 Top 10–15 GO terms of DEGs between CA25 group, CA100 group, CGA1000 group and CON group. Table S8–S10 Top 20 KEGG pathways of DEGs between CA25 group, CA100 group, CGA1000 group and CON group. Table S11–S13 Differentially expressed proteins between CA25 group, CA100 group, CGA1000 group and CON group. Table S14–S16 Top 10–15 GO terms of differentially expressed proteins between CA25 group, CA100 group, CGA1000 group and CON group. Table S17–S19 KEGG enrichment analysis of DEPs between CA25 group, CA100 group, CGA1000 group and CON group. Table S20 GO terms related to lipid metabolism by Gene Set Enrichment Analysis. Table S21 KEGG pathways related to lipid metabolism by Gene Set Enrichment 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

Tian, W., Gonzales, G.B., Wang, H. et al. Caffeic acid and chlorogenic acid mediate the ADPN-AMPK-PPARα pathway to improve fatty liver and production performance in laying hens. J Animal Sci Biotechnol 16, 49 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s40104-025-01175-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s40104-025-01175-z

Keywords