Go to Top Go to Bottom
Anim Biosci > Volume 38(5); 2025 > Article
Huang, Li, Yang, Zhang, Huang, Lin, Wang, Xiang, and Zhu: CIDEB promotes lipid deposition in goat intramuscular adipocytes

Abstract

Objective

Cell death-inducing DNA fragmentation factor alpha-like effector B (CIDEB), a family member of Cell death-inducing DFF45-like effectors (CIDEs), is well known as a crucial regulator for lipid metabolic signaling pathways in various metabolic tissues and secretory glands. However, its role in regulating intramuscular fat (IMF) deposition in goat remains unclear.

Methods

The expression vector pcDNA3.1-CIDEB was constructed and transfected into goat intramuscular preadipocytes; the overexpression and interference efficiency and expression of genes related to lipid metabolism were measured by Real-time polymerase chain reaction; the effect of overexpression of CIDEB and interfering with CIDEB on lipid droplet formation was observed by Oil Red O staining and glycerol phosphate oxidase-Trinder enzymatic reaction. Then RNA-Seq was used to investigate the metabolic pathway of CIDEB affecting adipocyte deposition in goat intramuscular preadipocytes.

Results

Overexpression of CIDEB significantly promoted the lipid droplets accumulation and the triglyceride deposition, and significantly upregulated the expression of genes related to lipid metabolism. After overexpression of CIDEB in goat intramuscular preadipocytes, 171 differentially expressed genes (DEGs) were found, including 122 up-regulated and 49 down-regulated DEGs, and the top three significantly changed pathways filtered by Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis were Cocaine addiction, Amphetamine addiction and Malaria pathways. Conversely, the silencing of CIDEB significantly reduced lipid accumulation in goat intramuscular preadipocytes, meanwhile changing the expression of lipid metabolism genes. For CIDEB silencing, a total of 2140 DEGs were found, including 1252 up-regulated and 888 down-regulated DEGs, and the top three significantly changed pathways filtered by KEGG analysis were Ribosome, Thyroid hormone signaling pathway and Alzheimer disease.

Conclusion

The expression of CIDEB can significantly promote lipid deposition of intramuscular adipocytes in goats, and these results provide important data to support further clarifying the mechanism of CIDEB gene on the regulation of intramuscular adipogenesis, and the IMF formation in goats.

INTRODUCTION

Adipose tissue, composed mainly of adipocytes, is a metabolically heterogeneous endocrine organ [1], mainly related to lipid metabolism, energy homeostasis, support structure and meat quality [2]. Anatomically, adipose tissue could be classified as subcutaneous adipose tissue (SAT), visceral adipose tissue (VAT), abdominal adipose tissue (AAT), intramuscular adipose tissue, intermuscular adipose tissue, heart adipose tissue, and kidney adipose tissue [1]. Of these, intramuscular fat is essential for improving the flavor and palatability of meat. Fat deposition traits are genetically and environmentally influenced and can affect meat quality, growth rate and energy metabolism in livestock. However, the molecular mechanism underlying fat deposition in goats is not fully understood [1].
The family of Cell death-inducing DFF45-like effectors (CIDEs) includes CIDEA, CIDEB, and CIDEC/Fsp27, named for their homology to the N-terminal domain of DNA fragmentation factor 45 (DFF45) [3,4]. CIDEs were firstly found to be associated with apoptosis [3,5,6], and were proved to induces caspase-independent cell death in human [7,8]. Besides apoptosis, CIDEs were also found to involve in cellular lipid metabolism regulation [9], including lipid storage, secretion and synthesis [10,11] in metabolic tissues and secretory glands [1214], and also plays an important role in the development of obesity, insulin resistance, and fatty liver [1517]. Several CIDE proteins have also been shown to affect intracellular lipid metabolism, such as lipid metabolism in adipocytes, hepatocytes, and macrophages [18]. Compared with other family members, Cell death-inducing DNA fragmentation factor alpha-like effector B (CIDEB) is relatively stable and unaffected by nutritional conditions [19,20], mainly expressed in liver [14] and lower in intestine [21].
The role of CIDEB in promoting very low-density lipoprotein (VLDL) lipidation, maturation and secretion has been well studied in mice [22,23]. The absence of CIDEB resulted in reduced VLDL lipidation and maturity and altered hepatic cholesterol homeostasis [22,24,25]. The cholesterol and low-density lipoprotein in the blood of CIDEB−/− mice were significantly decreased, and the synthesis pathway of cholesterol in the liver was inhibited by CIDEB deficiency [25]. What’s more, CIDEB also promotes hepatic lipid storage through fusion and growth of hepatic lipid droplets [26]. In mouse liver, CIDEB was found to anchor SREBP/SCAP complex to Coatomer Complex II to regulate the activity of the sterol regulatory element binding protein 1 (SREBP1). CIDEB is also involved in maintaining lipid homeostasis by promoting the loading of the SREBP/SCAP protein complex to the endoplasmic reticulum (ER) exit site and helping its export from the ER [23]. The deficiency of CIDEB significantly reduced the activity of SREBP1, decreased the expression of downstream target genes of SREBP pathway, and alleviated diet-induced hepatic steatosis. Taken together, these studies performed on CIDEB−/− mice suggest that CIDEB plays a multifunctional role in controlling hepatic lipid secretion, lipid storage, and lipid synthesis. In the early stages of fat formation, the expression of CIDEB gene is significantly upregulated, which may contribute to the initiation of fat formation [27].
However, little data was available for the direct association of CIDEB gene with adipocyte deposition in domestic animals. In the present study, CIDEB was found to significantly promote cellular lipid deposition by overexpression of CIDEB in goat intramuscular preadipocytes. And then transcriptional profiles accompanying the alteration of CIDEB was performed to screen the signal pathways induced by CIDEB by RNA-Seq in affecting adipocyte deposition. These data may enhance our understanding about the regulation of formation of intramuscular fat in goat.

MATERIALS AND METHODS

Cell isolation, culture and treatment

All animal procedures used in this study were approved by the Institutional Animal Care and Use Committee, Southwest Minzu University (permit number: S2020-013, revised on 1 June 2004). Goat primary intramuscular preadipocytes were isolated using the previously described method with minor modification [28]. Briefly, the longissimus dorsi muscle tissues were collected from two-day-old Jianzhou goats from Jianyang County (Sichuan, China) after slaughter. The tissues were then washed 3 times with phosphate-buffered saline (PBS), separated and trimmed on a super clean bench, following which 2 volumes of type II Collagenase (Sigma, St. Louis, MO, USA) was supplemented for digestion at 37°C for 90 min (shaking every 5 min). The digestion was stopped by adding an equal volume of complete culture medium containing 10% fetal bovine serum (DMEM/F12 containing 10% FBS, 1% antibiotic, and 50 μmol/L oleic acids [Sigma, Tokyo, Japan]). The samples were then filtered using sterile gauze and a 75-μm cell strainer, and the cells were then centrifuged at 2,000 r/min for 5 min. After discarding the supernatant, the red blood cell lysate was used for resuspended the cells, followed by standing for 5 min and centrifuging at 2,000 r/min for 5 min. After resuspending in clear complete culture medium, the cells were seeded into 25 cm2 culture flasks for the following cultivation under 5% CO2 at 37°C with new complete medium after 2 h. The cell culture medium was changed every two days until the cell density reached 80%. For cell treatment, the cells were passed to the third generation at a ratio of 1 to 3 and inoculated into 6-well plate (10-cm2). After treatment was completed, the cell culture medium was replaced with an adipocyte-inducing medium.

Construction of goat CIDEB gene overexpression vector and siRNA synthesis

The plasmid of pMD19-T-CIDEB was stored in our laboratory. A subcloning system was preformed to amplify the target sequences after added the protective base, Hind III and myc-tagged and BamHI to the primers. The primers as follows: S: CCCAAGCTTATGGAGCAGAAACTCATCTCTGAAGAGGATCTGGAGTACCTCTCTAACCTGGAC, A: CGCGGATCCTCAGTAGGGTTTAAGGCGACCCTG.
Subcloning conditions as follows: 98°C for 3 min; 98°C for 15 s, 57°C for 30 s, 72°C for 2 min, 30 cycles; 72°C for 10 min. The polymerase chain reaction (PCR) product was then recycled and digested and linked to digested pcDNA3.1 to build pcDNA3.1-CIDEB recombination plasmid. After being transformed into competent cells DH5α (Tsingke, Beijing, China) and identified by double digestion, the plasmid was validated by DNA sequencing with the help of Chengdu Tsingke biotechnology company.
Negative control (NC) siRNA and two siRNAs of goat CIDEB gene were synthesized by Shanghai GenePharma company. siRNA-NC: S: UUCUCCGAACGUGUCACGUTT, A: ACGUGACACGUUCGGAGAATT. siRNA-CIDEB165: S: GUCAGGAACUGCUAGACAATT, A: UUGUCUAGCAGUUCCUGACTT. siRNA-CIDEB396: S: GCAUCACCUUCGACGUAUATT, A: UAUACGUCGAAGGUGAUGCTT.

Cell transfection

Cell transfection was performed as previously described [29]. After cell passage, transfection was performed with Lipofectamine 3000 (Thermo Fisher Scientific, Shanghai, China) when the cells filled 70% to 80% of the culture vial. They were transfected with pcDNA3.1-CIDEB, pcDNA3.1, siRNA-CIDEB165 and NC siRNA. In this experiment, pcDNA3.1-CIDEB plasmid group and siRNA-CIDEB (si-CIDEB) group were the experimental group, and pcDNA3.1 plasmid (NC) group and negative control siRNA (si-NC) group were the control group. First, the cells are starved; After the medium was discarded and washed three times with PBS, 900 μL Opti medium was added to each well of the six-well plate and placed in a constant temperature incubator at 37°C for 4 hours. After starvation treatment, the transfection premix was prepared: In the overexpression experiment, the premix was 50 μL Opti medium and 3 μL lip3000. Premixed solution B consists of 50 μL Opti medium mixed with 2.5 μL P3000 and 1μg plasmid DNA (pcDNA3.1-CIDEB or pcDNA3.1). Mix the premix well and let it sit at room temperature for 20 minutes before using. Interference assay: 100 μL Opti medium, 6 μL lip3000, and 4 μL siRNA were mixed, and the premix was also placed at room temperature for 10 minutes, then gently suspended and instillated to complete cell transfection for overexpression and interference studies. Six hours after transfection, 50 μmol/L Sigma oleic acid induction solution was added, and cells were collected after 48 hours of culture.

Oil red O staining

Oil red O staining was performed as the previously reported methods [30]. Briefly, the cells were slowly washed 3 times with PBS and fixed with 10% formaldehyde for 30 min. After discarding the formaldehyde and washing the cells 3 times with PBS, Oil Red O working solution (the mixture of 3 mL Oil Red 5 g/L dissolved in isopropanol and 2 mL of ddH2O) was supplemented for 20 min incubation at room temperature. Finally, the cells were cleaned with PBS and photographed with inverted fluorescence microscope (Zeiss, Tokyo, Japan). For the quantification of Oil Red O, 1 mL of isopropanol was added to each well of the 6-well plate and the absorbance value at 510 nm was measured using a spectrophotometer (Thermo Fisher Scientific, Shanghai, China).

Triglyceride determination

Cellular triglyceride content in goat intramuscular adipocytes were used to determine Tissue Triglyceride Content Assay Kit (Applygen, Beijing, China). Briefly, the cells were washed three times with PBS, and treated by 200 μL of triglyceride lysate on ice for 10 min. Then the lysate was divided into two portions for optical density value assay, one for triglyceride detection at 562 nm while the other one for cellular total protein detection by bicinchoninic acid assay at 550 nm according to the manual of the kit (Vazyme, Nanjing, China), which was then used for triglyceride content correction.

Transcriptome analysis of CIDEB goat intramuscular adipocytes

After the treatment of overexpression and siRNA silencing of CIDEB for 48h, the cells were harvested and treated by 1 mL Trizol (Takara, Dalian, China) for total RNA extraction. The RNA transcriptome sequencing was performed by Shanghai OE Biotechnology Co, Ltd. Gene Ontology (GO) and Kyoto encyclopedia of genes and genomes (KEGG) analysis were analyzed using oebiotech (https://www.oebiotech.com/index.php?c=show&id=414). All the experiments were replicated three times.

Quantitative real-time polymerase chain reaction

To further explore the mechanism of CIDEB in lipid metabolism, its overexpression and interference were used to detect the effect on the expression of genes related to lipid metabolism. The primers of quantitative real-time PCR (RT-qPCR) are shown in Table 1. Real-time quantitative PCR was performed using three biological replicates and technical triplicates of each cDNA sample, according to the manufacturer’s protocol. Relative expression levels of target genes for gene correction using ubiquitously expressed transcript (UXT) as an internal reference. PCR was performed using the same volume of cDNA sample for each gene to record the cycle threshold (Ct) value. PCR experiments were performed with a Bio-Rad CFX96 PCR System using Taq Pro Universal SYBR qPCR Master Mix (Vazyme, Q712-02). PCR reaction system 10 μL, including 5 μL SYBR Green Real-time PCR Master Mix (Vazyme), 0.2 L (10 μM) primers, 1 μL sample and 3.6 μL ddH2O. The following cycling conditions were used: 95°C for 3 min to activate the polymerase followed by 40 cycles of denaturation at 95°C for 10 s, annealing at 60°C for 30 s and extension at 72°C for 30 s. Fluorescence signal was obtained at a temperature of 72°C extension.

Data analysis

RT-qPCR results were statistically analyzed by 2−ΔΔCt method. All test data were set with at least 3 biological replicates. GraphPad Prism 9.0 was used for statistical analysis and plotting. Two-tailed t-test was used for significance identification.

RESULTS

Overexpression of CIDEB promotes lipid deposition in goat intramuscular preadipocytes

To further elucidate the role of CIDEB in fat deposition, we ligated the CDS region of the CIDEB gene into vector, obtained 5428 bp (pcDNA3.1 vector) and 708 bp (CIDEB gene) by Hind III and Bam HI restriction enzymes digestion and transfected them into goat intramuscular precursor adipocytes. The expression level of CIDEB was increased by about 230-fold compared with the control after overexpression of CIDEB (Figure 1A). The cellular triglyceride was increased significantly by 1.62-fold after overexpression of CIDEB compared with control (Figure 1B). Consistent with the results of triglyceride formation, the accumulation of lipid droplets in cells also increased by 1.39-fold compared with control (Figure 1C). Meanwhile, the overexpression of CIDEB also significantly increased the expression of adipose differentiation marker genes peroxisome proliferator activated receptor gamma (PPARγ) by 1.57-fold and enhancer binding protein alpha (C/EBPα) by 1.67-fold (Figure 2A), lipid droplet accumulation-related genes perilipin 1 (PLIN1) by 33.82-fold and tail-interacting protein 47 (TIP47) by 5.07-fold (Figure 2B), fatty acid synthesis and transport genes acetyl-CoA carboxylase (ACC) by 2.48-fold, acyl-CoA synthetase long-chain family member 1 (ACSL1) by 1.64-fold, fatty acid binding protein 3 (FABP3) by 1.74-fold and fatty acid synthase (FASN) by 1.18-fold (Figure 2D), triglyceride synthesis-related gene diacylglycerol O-acyltransferase 2 (DGAT2) by 1.16-fold (Figure 2C). Correspondingly, the overexpression of CIDEB significantly decreased the expression of lipolysis and fatty acid β oxidation-related genes acyl-CoA oxidase 1 (ACOX1) by 50% and hormone-sensitive lipase (HSL) by 58% (Figure 2E). In conclusion, it can be considered that overexpression of CIDEB promotes adipogenesis in goat intramuscular preadipocytes.

Effect of CIDEB-overexpressing on transcriptome profile in goat intramuscular preadipocytes

To determine the possible mechanism underlying the activated lipid deposition, RNA-Seq analysis was performed after CIDEB overexpression in goat intramuscular preadipocytes. In results, a total of 171 differentially expressed genes (DEGs) were screened (p<0.05) by the overexpression of CIDEB, including 122 up-regulated genes and 49 down-regulated genes (Figures 3A, 3B). GO enrichment analysis showed that the upregulated DEGs were related to cytosol, membrane-enclosed lumen, organelle lumen, intracellular organelle lumen, nuclear lumen and protein maturation (Figure 3C); the downregulated DEGs were related to organelle membrane, ER, mitochondrial membrane, quinone or similar compound as acceptor, mitochondrial inner membrane and organelle inner membrane (Figure 3D). By GO analysis, the DEGs were enriched in 21 terms of biological processes (mainly containing cellular processes, metabolic processes, biological regulation, biological process regulation and stimulus response), 8 terms of molecular functions (mainly containing binding and catalytic activities) and 17 terms of cellular components (mainly containing cells, cellular structural material, organelles, membrane and organellar structural material) (Figure 3E). Three lipid metabolism-related pathways were screened from the functional annotation results of GO, respectively aminophospholipid transport, lipid metabolic process, and glycerolipid metabolic process (p<0.05) (Table 2). To probe the potential signal pathways by CIDEB overexpression, the up- and down-regulated DEGs were analyzed by KEGG analysis respectively. For the up-regulated DEGs, malaria, taurine and hypotaurine metabolism, amphetamine addiction and cocaine addiction were enhanced (Figure 3F). In addition, we verified their expression levels through RT-qPCR analysis. Consistently, overexpression of CIDEB downregulated the expression of FASN and ACSL1 (Figure 4).

Interfering of CIDEB inhibited lipid deposition in goat intramuscular preadipocytes

To elucidate the role of CIDEB in regulating intramuscular lipid accumulation in goats, we silenced the expression of CIDEB by two pairs of siRNAs in goat intramuscular preadipocytes. The expression level of CIDEB was reduced by 82% with treatment of CIDEB-165 and 75% with the treatment of CIDEB-396 (Figure 5A), so CIDEB-165 was used in the subsequent experiment. Contrary to overexpression, the abundance of TAG decreased by 28% after CIDEB knockdown (Figure 5B), and the accumulation of lipid droplets decreased significantly by 27% (Figure 5C). The knockdown of CIDEB significantly decreased the expression of PPARγ by 42%, LPL by 35%, C/EBPα by 69%, SREBP1 by 62% (Figure 6A), PLIN1 by 31%, TIP47 by 84% (Figure 6B), ACC by 62%, FASN by 80%, ACSL1 by 78% and FABP3 by 14% (Figure 6D), and meanwhile up-regulated the expression of ATGL (1.29-fold), CPT1B (1.28-fold), ACOX1 (7.92-fold) and CPT1A (1.29-fold) (Figure 6E) by CIDEB interference. In conclusion, it can be launched to conclude that interfering of CIDEB inhibited lipid deposition in goat intramuscular preadipocytes.

Effect of CIDEB-interference on transcriptome profile in goat intramuscular preadipocytes

To determine the possible mechanism underlying the inactivated lipid deposition, RNA-Seq analysis was performed after CIDEB silencing in goat muscle preadipocytes. A total of 2140 DEGs were screened (p<0.05), including 1252 up-regulated genes and 888 down-regulated genes (Figures 7A, 7B). GO enrichment analysis showed that the upregulated DEGs were related to intracellular anatomical structure, organelle, intracellular organelle, membrane-bounded organelle, cytoplasm and intracellular membrane-bounded organelle (Figure 5C); the downregulated DEGs were related to intracellular anatomical structure, intracellular organelle, organelle, cellular protein metabolic process, cytoplasm, non-membrane-bounded organelle, intracellular non-membrane-bounded organelle (Figure 5D). By GO analysis, the DEGs were enriched in 25 terms of biological processes (mainly containing metabolic processes, developmental process, rhythmic process, reproductive process, multi-organism processes, locomotion and growth response), 8 terms of molecular functions (mainly containing binding and catalytic activities) and 17 terms of cellular components (mainly containing organelle part, membrane-enclosed lumen, nucleoid, extracellular region part and extracellular matrix component) (Figure 7E). Four lipid metabolism-related pathways were screened from the functional annotation results of GO, respectively lipid transport involved in lipid storage, lipid storage, aminophospholipid transport and cellular response to lipid (p<0.05) (Table 3). To probe the potential signal pathways by CIDEB interference, the up- and down-regulated DEGs were analyzed by KEGG analysis respectively. For the up-regulated DEGs, apelin signaling pathway, insulin signaling pathway, glycolysis/gluconeogenesis and focal adhesion were enhanced (Figure 7F). In addition, we verified their expression levels through RT-qPCR analysis. Consistently, interference with CIDEB downregulated expression of FASN and ACSL1 (Figure 4).

DISCUSSION

We understand that CIDEB as a lipid droplet fusion-associated protein has been extensively studied, especially in mouse and human diabetes, cancer, etc. However, there is still relatively little research on CIDEB in the field of animal husbandry, especially in goats. Although it has an important role in lipid droplet fusion, it does not follow from this that it influences lipid metabolic networks and lipid deposition in goat intramuscular preadipocytes. The innovation of this paper lies in the fact that we systematically verified the regulatory effects of CIDEB on intramuscular fat deposition and lipid metabolism networks at both overexpression and interference levels, using a domesticated animal (goat) as the study subject. It is of positive significance for our in-depth understanding of the potential function of CIDEB in goat intramuscular fat formation and its application prospects in animal husbandry from the perspective of molecular breeding.
The crucial role of PPARγ and SREBP1 in controlling cellular lipid accumulation and the expression of adipogenesis genes have been well studied. In mice, the up-regulated expression of SREBP1 promoted the lipid fat metabolism and the secretion of VLDL, while the CIDEB deficiency cancelled the diet-induced obesity via the significantly decreased of SREBP1 [14,24]. Even in goats, both of PPARγ and SREBP1 were positively regulated lipogenesis in mammary gland. Recently, the central role of PPARγ and its target genes was confirmed in the control of milk fat synthesis in goat mammary epithelial cells [31]. In the present study, the expression of PPARγ and SREBP1 were upregulated by the overexpression of CIDEB, and then validated by the decreased expression by CIDEB silencing, proposed the hypothesis that CIDEB, as an original function of lipid droplets confusion, may regulate lipid metabolism via the control of PPARγ and SREBP1. Supporting the hypothesis, their downstream genes, including ACC, FASN and SCD et al, were upregulated along with the increased PPARγ and SREBP1.
Differentiation into adipocytes requires the sequential expression of the transcription factors, C/EBPβ, C/EBPδ, PPARγ, and C/EBPα [32]. C/EBPα plays an important role in cell proliferation, differentiation, metabolism, inflammation, and other responses. Numerous studies have shown that many adipose-specific gene promoters have C/EBP effector structural domains that can be activated by C/EBPα [33]. For example, C/EBPα can induce PPARγ expression by recognizing the C/EBP effector structural domain of the PPARγ promoter. PPARγ and C/EBPα cross-regulate each other’s expression as well as governing expression of the entire adipogenicity program, which includes activation of additional transcription factors [34]. Previous studies have shown that C/EBPα as a transcription factor can bind to the ACOX1 promoter region and repress its transcriptional activity [35]. This conclusion was verified in this experiment. Of these, interference with CIDEB significantly repressed C/EBPα expression. Loss of CIDEB also affected accumulation of PPARγ but to a much lesser extent, presumably reflecting its reduced positive feedback regulation by C/EBPα. It has been found that the CIDE family protein Fsp27 interacts with C/EBPβ to regulate the expression of a subset of genes downstream of C/EBPβ in adipocytes, and that CIDEA acts as a previously unknown transcriptional co-activator of C/EBPβ in the mammary gland to control lipid secretion and pup survival [36]. It is likely then that CIDEA causes changes in CEBPβ by regulating PPARγ and thus CIDEA. So, does CIDEB have a similar role? It has been reported in the literature that C/EBPβ inhibits CIDEB expression only under ER stress, which inhibits lipoprotein transport in the liver [37]. However, whether CIDEB interacts with CEBPα is still unclear to us.
In addition, we also found significant changes in the pyruvate metabolism after overexpression of CIDEB by enrichment analysis of the differential gene KEGG pathway. Here, we found that ACOT12 was upregulated in the pyruvate metabolism pathway after overexpression of CIDEB. Among them, Acyl-CoA Thioesterase 12 (ACOT12), is the major cytoplasmic enzyme that preferentially hydrolyzes acetyl-CoA which hydrolyze fatty acyl-CoA to free FA and CoA and promote pyruvate metabolism [3840]. However, acetyl-CoA is a key indicator of the regulation of metabolic status by ACOT12. Acetyl-CoA a is involved in De novo lipogenesis (DNL) metabolism and is used by ACC to catalyze malonyl-CoA synthesis [41]. With this change, we hypothesize that CIDEB may regulate DNL by upregulating the ACOT12 and thus changes in these pathways. Its exact mechanism needs to be verified by subsequent experiments. We also found significant changes in the malaria, taurine and hypotaurine metabolism, amphetamine addiction and cocaine addiction after overexpression of CIDEB by enrichment analysis of the differential gene KEGG pathway. It has been reported that lipid metabolism is significantly elevated during intraerythrocytic development of malaria parasites infected [42], which mainly through the TLR/MyD88 signaling pathway. TLR/MyD88 is a classic signaling inflammatory response that also involves in obesity development and is inhibited by the overexpression of PPARγ via the negative control of the expression of TLR4, and then activates the expression of CD36 for promoting fatty acid uptake and fat accumulation. This may highlight the important role of PPARγ in regulating.
Furthermore, we also found significant changes in the insulin signaling pathway, glycolysis/gluconeogenesis, focal adhesion and apelin signaling pathway after interference of CIDEB by KEGG pathway analysis. Apelin was reported to stimulate glucose uptake, increase insulin sensitivity, and regulate lipolysis and fatty acid oxidation. Apelin-13, possibly by activating the PI3K/AKT pathway, could improve the lipid metabolism [43]. PI3K/AKT is the downstream pathway of focal adhesion pathway [41], and the PI3K-AKT-mTOR signaling pathway can regulate downstream gene SREBP1. We found that the apelin signaling pathway also contain the cAMP-PKA pathway to regulating the expression levels of PPARγ and SREBP1. Moreover, the insulin signaling pathway controls the Glycolysis/glycogenesis pathway and mediates the interaction between proteins involved in lipid metabolism [44,45]. Therefore, we further speculated that CIDEB may regulate lipid deposition in goat intramuscular adipocytes through insulin signaling pathway, glycolysis/gluconeogenesis, focal adhesion and apelin signaling pathway by regulating the expression levels of PPARγ and SREBP1. In addition, after interfering with CIDEB, we also found enhanced function of the thyroid hormone signaling pathway. Studies have shown that the extract of Sanghuangporus vaninii extract can reduce blood lipids in mice, which may promote lipid metabolism and cholesterol excretion through thyroid hormone signaling pathway, and inhibit cholesterol biosynthesis [46]. We speculate that interference with CIDEB promotes the enhancement of thyroid hormone signaling function, promotes lipid metabolism and inhibits lipid deposition.
Moreover, two pathways are known for triglyceride metabolism within lipid droplets, cytoplasmic lipolysis (lipolysis) and lysosome-mediated autophagy (lipophagy), the latter of which involves the hydrolysis of lipid droplets by lysosomal acid lipase after engulfment by autophagic vesicles [47]. In this study we found that the lysosome pathway was altered by KEGG analysis during both overexpression and interference with CIDEB. Then whether CIDEB is involved in lipophagy is not clear to us, but it also gives us referable data for our next study.
In summary, the present study found that CIDEB plays an important role in intramuscular fat deposition in goats. CIDEB was predicted to regulate cellular lipid deposition by regulating the expression levels of SREBP1 and PPARγ as well as the fat synthesis-related genes ACC and FASN. Understanding the regulatory role of CIDEB in intramuscular fat deposition in goats may help improve goat meat quality.

Notes

CONFLICT OF INTEREST

No potential conflict of interest relevant to this article was reported.

AUTHORS’ CONTRIBUTION

Conceptualization: Xiang H, Zhu J.

Data curation: Li Q.

Formal analysis: Huang Z.

Software: Zhang C.

Validation: Yang C.

Investigation: Li Q.

Writing - original draft: Huang Z, Li Q.

Writing - review & editing: Huang Z, Li Q, Yang C, Zhang C, Huang L, Lin Y, Wang Y, Xiang H, Zhu J.

FUNDING

This work was supported by Sichuan Science and Technology Program (Chengdu, China, 2021YFYZ0003, 2024NSFSC0029, 2022NSFSC0082), National Natural Science Foundation of China (Beijing, China, 32302702), the Fundamental Research Funds for the Central Universities, Southwest Minzu University (Beijing, China, ZYN2023101), the Scientific and Technological Innovation Team for Qinghai-Tibetan Plateau Research in Southwest Minzu University (2024CXTD13).

ACKNOWLEDGMENTS

Not applicable.

DATA AVAILABILITY

The mRNA transcriptional profile data mRNA transcriptional profile PRJNA983224. Further information Contact author or corresponding author.

ETHICS APPROVAL

All animal procedures used in this study were approved by the Institutional Animal Care and Use Committee, Southwest Minzu University (permit number: S2020-013, revised on 1 June 2004).

DECLARATION OF GENERATIVE AI

No AI tools were used in this article.

SUPPLEMENTARY MATERIAL

Not applicable.

Figure 1
Detection of CIDEB overexpression efficiency. (A) Detection of CIDEB overexpression efficiency, CIDEB-OVER is pcDNA3.1-CIDEB treatment group, pcDNA3.1 is control group. (B) Determination of cellular TAG content of CIDEB overexpression group and control group. (C) Oil red O staining of intramuscular preadipocytes in control group (pcDNA3.1) and CIDEB overexpression group (CIDEB-OVER) and Oil red O staining OD value detection (490 nm). ** p<0.01, **** p<0.0001. TAG, triglyceride; OD, optical density.
ab-24-0584f1.jpg
Figure 2
Overexpression of CIDEB altered the expression level of related genes. (A) Adipose differentiation related genes (B) Lipid droplet accumulation related genes. (C) Triglycerides (TAG) synthesis related genes. (D) Fatty acid synthesis and transport related genes. (E) Lipolysis and β oxidation related genes. Data are shown as mean±SEM, * p<0.05, ** p<0.01. SEM, standard error of the mean.
ab-24-0584f2.jpg
Figure 3
Overexpression of CIDEB affected the mRNA transcript profiles. (A) RNA-seq volcano plot of significantly differential expression genes (DEGs) in pcDNA3.1-CIDEB (n = 3) vs NC (n = 3) goat intramuscular adipocytes, red and green dots denote upregulated and downregulated genes, respectively. (B) The heat map shows the relative levels of DEGs. (C) GO enrichment analysis of upregulated differential genes. (D) GO enrichment analysis of downregulated differential genes. (E) The GO pathway analysis of related DEGs, up represents up-regulated DEGs, and down represents down-regulated DEGs (F) The KEGG pathway analysis of related DEGs. NC, negative control; GO, Gene Ontology; KEGG, Kyoto Encyclopedia of Genes and Genomes.
ab-24-0584f3.jpg
Figure 4
Verification of representative DEGs by qPCR (n = 3), including fatty acid synthase (FASN), acyl-CoA synthetase long-chain family member 1 (ACSL1). ** p<0.01. DEGs, differential expression genes; NC, negative control; qPCR, quantitative polymerase chain reaction.
ab-24-0584f4.jpg
Figure 5
Detection of CIDEB interference efficiency. (A) Detection of interference efficiency of CIDEB-396. (B) Detection of interference efficiency of CIDEB-165. (C)Determination of cellular TAG content of CIDEB interference group and control group. (D) Oil red O staining of cells in control group (si-NC) and interference group (CIDEB-165) and Oil red O staining OD value detection (490 nm), ** p<0.01, **** p<0.0001. TAG, triglyceride; OD, optical density.
ab-24-0584f5.jpg
Figure 6
Interference with CIEDB altered the expression level of related genes. (A) Adipose differentiation related genes. (B) Lipid droplet accumulation related genes. (C) Triglycerides (TAG) synthesis related genes. (D) Fatty acid synthesis and transport related genes. (E) Lipolysis and β oxidation related genes. Data are shown as mean±SEM, * p<0.05, ** p<0.01. SEM, standard error of the mean.
ab-24-0584f6.jpg
Figure 7
Interference with CIDEB affected the mRNA transcript profiles. (A) RNA-seq volcano plot of significantly differential expression genes (DEGs) in SI-CIDEB (n = 3) vs NC (n = 3) goat intramuscular adipocytes, red and green dots denote upregulated and downregulated genes, respectively. (B) The heat map shows the relative levels of DEGs. (C) GO enrichment analysis of upregulated differential genes. (D) GO enrichment analysis of downregulated differential genes. (E) The GO pathway analysis of related DEG, up represents up-regulated DEGs, and down represents down-regulated DEGs (F) The KEGG pathway analysis of related DEGs. GO, Gene Ontology; KEGG, Kyoto Encyclopedia of Genes and Genomes; DEGs, differential expression genes; NC, negative control.
ab-24-0584f7.jpg
Table 1
Primers information for RT-qPCR
Gene Full name Sequence (5′-3′) Tm/°C Length/bp Genbank ID
PPARγ Peroxisome proliferator activated receptor gamma F: AAGCGTCAGGGTTCCACTATG 60 197 NM_001285658.1
R: GAACCTGATGGCGTTATGAGAC
C/EBPα Enhancer binding protein alpha F: CCGTGGACAAGAACAGCAAC 60 142 XM_018062278.1
R: AGGCGGTCATTGTCACTGGT
SREBP1 Sterol regulatory element binding transcription factor 1 F: AAGTGGTGGGCCTCTCTGA 60 127 NM_001285755.1
R: GCAGGGGTTTCTCGGACT
ACSL1 Acyl-CoA synthetase long-chain family member 1 F: TGACTGTTGCTGGAGACTGG 60 199 XM_005698718
R: CAGCCGTCTTTATCCAGAGC
DGAT1 Diacylglycerol O-acyltransferase 1 F: CCACTGGGACCTGAGGTGTC 60 101 XM_018058728
R: GCATCACCACACACCAATTCA
DGAT2 Diacylglycerol O acyltransferase 2 F: CATGTACACATTCTGCACCGATT 60 100 XM_018058853.1
R: TGACCTCCTGCCACCTTTCT
AGPAT6 1-acylglycerol-3-phosphate O-acyltransferase 6 F: AAGCAAGTTGCCCATCCTCA 60 101 JI861797.1
R: AAACTGTGGCTCCAATTTCGA
ATGL Adipose triglyceride lipase F: GGAGCTTATCCAGGCCAATG 60 180 NM_001285739
R: TGCGGGCAGATGTCACTCT
HSL Hormone-sensitive lipase F: GGGAGCACTACAAACGCAACG 60 118 EU273879
R: TGAATGATCCGCTCAAACTCG
LPL Lipoprotein lipase F: AGGACACTTGCCACCTCATTC 60 169 XM_013966067.2
R: TTGGAGTCTGGTTCCCTCTTGT
ACOX1 Acyl-CoA oxidase 1 F: CGAGTTCATTCTCAACAGTCCT 60 211 NM_00103528
R: GCATCTTCAAGTAGCCATTATCC
CPT1A Carnitine palmitoyltransferase 1A F: TGACGGCTCTGGCACAAGAT 60 164 XM_018043311.1
R: CGCGAAGTAGTTGCTATTCAC
CPTIB Carnitine Palmitoyltransferase 1B F: ACGAGGAGTCTCACCACTACG 60 111 NM_001009259
R: GTGTGAAGGACTTGTCGAACCA
PLIN1 Perilipin 1 F: CCCATTGCCAGCACTTCAGA 60 95 XM_018066567.1
R: GCAGCGTACTCGGCAGTATCTC
TIP47 Tail-interacting protein, 47 F: GTCCGCTGACGAGACCGAA 60 319 NM_001285595.1
R: CAGATTCTCCTCCAGTTTGTCC
ADFP Adipose differentiation-related protein/Perilipin 2 F: TTGCTGTTGCCAATACCT 60 284 NM_001285596.1
R: CTGCATCATCCGACTTCC
ACSS2 Acyl-CoA synthetase short chain family member 2 F: GGCGAATGCCTCTACTGCTT 60 100 XM_018057751
R: GGCCAATCTTTTCTCTAATCTGCTT
FABP3 Fatty acid binding protein 3 F: GATGAGACCACGGCAGATG 60 120 NM_00128570
R: GTCAACTATTTCCCGCACAAG
UXT Ubiquitously expressed transcript S: GCAAGTGGATTTGGGCTGTAAC 60 180 XM_005700842
R: ATGGAGTCCTTGGTGAGGTTGT
CIDEB Cell death-inducing DFF45- like effector B F: CAGCCGCTACCCGTCAGGAACT 60 233 Sequence obtained from the experiment
R: CTTGCTGTGCTTGGGCTTCTCC
FASN Fatty acid synthase F: GGGCTCCACCACCGTGTTCCA 60 226 NM_001285629.1
R: GCTCTGCTGGGCCTGCAGCTG
ACC Acetyl-CoA carboxylase alpha F: CTCCAACCTCAACCACTACGG 60 171 NM_174224.2
R: GGGGAATCACAGAAGCAGCC

RT-qPCR, quantitative real-time polymerase chain reaction; Tm, melting temperature.

Table 2
Screening results of CIDEB overexpression GO enrichment
Function classification Pathway Function p-value
Biological process GO:0015917 Aminophospholipid transport 0.015374882
Biological process GO:0006629 Lipid metabolic process 0.040561346
Biological process GO:0046486 Glycerolipid metabolic process 0.042506493

GO, Gene Ontology.

Table 3
Screening results of CIDEB interference GO enrichment
Function classification Pathway Function p-value
Biological process GO:0010888 Lipid transport involved in lipid storage 0.010139619
Biological process GO:0019915 Lipid storage 0.010343653
Biological process GO:0015917 Aminophospholipid transport 0.019999842
Biological process GO:0071396 Cellular response to lipid 0.031200491

GO, Gene Ontology.

REFERENCES

1. Du L, Chang T, An B, et al. Transcriptomics and lipid metabolomics analysis of subcutaneous, visceral, and abdominal adipose tissues of beef cattle. Genes 2023;14:37. https://doi.org/10.3390/genes14010037
crossref pmid pmc
2. Frigolet ME, Gutiérrez-Aguilar R. The colors of adipose tissue. Gac Med Mex 2020;156:142–9. https://doi.org/10.24875/GMM.M20000356
crossref pmid
3. Inohara N, Koseki T, Chen S, Wu X, Núñez G. CIDE, a novel family of cell death activators with homology to the 45 kDa subunit of the DNA fragmentation factor. EMBO J 1998;17:2526–33. https://doi.org/10.1093/emboj/17.9.2526
crossref pmid pmc
4. Lugovskoy AA, Zhou P, Chou JJ, McCarty JS, Li P, Wagner G. Solution structure of the CIDE-N domain of CIDE-B and a model for CIDE-N/CIDE-N interactions in the DNA fragmentation pathway of apoptosis. Cell 1999;99:747–55. https://doi.org/10.1016/s0092-8674(00)81672-4
crossref pmid
5. Liang L, Zhao M, Xu Z, Yokoyama KK, Li T. Molecular cloning and characterization of CIDE-3, a novel member of the cell-death-inducing DNA-fragmentation-factor (DFF45)-like effector family. Biochem J 2003;370:195–203. https://doi.org/10.1042/BJ20020656
crossref pmid pmc
6. Wu C, Zhang Y, Sun Z, Li P. Molecular evolution of Cide family proteins: novel domain formation in early vertebrates and the subsequent divergence. BMC Evol Biol 2008;8:159. https://doi.org/10.1186/1471-2148-8-159
crossref pmid pmc
7. Min J, Zhang W, Gu Y, et al. CIDE-3 interacts with lipopolysaccharide-induced tumor necrosis factor, and overexpression increases apoptosis in hepatocellular carcinoma. Med Oncol 2011;28:219–27. https://doi.org/10.1007/s12032-010-9702-1
crossref pmid
8. Liu K, Zhou S, Kim JY, et al. Functional analysis of FSP27 protein regions for lipid droplet localization, caspase-dependent apoptosis, and dimerization with CIDEA. Am J Physiol Endocrinol Metab 2009;297:E1395–413. https://doi.org/10.1152/ajpendo.00188.2009
crossref pmid
9. Chen FJ, Yin Y, Chua BT, Li P. CIDE family proteins control lipid homeostasis and the development of metabolic diseases. Traffic 2020;21:94–105. https://doi.org/10.1111/tra.12717
crossref pmid
10. Gao G, Chen FJ, Zhou L, et al. Control of lipid droplet fusion and growth by CIDE family proteins. Biochim Biophys Acta Mol Cell Biol Lipids 2017;1862:1197–204. https://doi.org/10.1016/j.bbalip.2017.06.009
crossref pmid
11. Yu J, Li P. The size matters: regulation of lipid storage by lipid droplet dynamics. Sci China Life Sci 2017;60:46–56. https://doi.org/10.1007/s11427-016-0322-x
crossref pmid
12. Zhou Z, Yon Toh S, Chen Z, et al. Cidea-deficient mice have lean phenotype and are resistant to obesity. Nat Genet 2003;35:49–56. https://doi.org/10.1038/ng1225
crossref
13. Zhang S, Shui G, Wang G, et al. Cidea control of lipid storage and secretion in mouse and human sebaceous glands. Mol Cell Biol 2014;34:1827–38. https://doi.org/10.1128/MCB.01723-13
crossref pmid pmc
14. Li JZ, Ye J, Xue B, et al. Cideb regulates diet-induced obesity, liver steatosis, and insulin sensitivity by controlling lipogenesis and fatty acid oxidation. Diabetes 2007;56:2523–32. https://doi.org/10.2337/db07-0040
crossref pmid
15. Gong J, Sun Z, Li P. CIDE proteins and metabolic disorders. Curr Opin Lipidol 2009;20:121–6. https://doi.org/10.1097/MOL.0b013e328328d0bb
crossref pmid
16. Slayton M, Gupta A, Balakrishnan B, Puri V. CIDE proteins in human health and disease. Cells 2019;8:238. https://doi.org/10.3390/cells8030238
crossref pmid pmc
17. Sans A, Bonnafous S, Rousseau D, et al. The differential expression of cide family members is associated with nafld progression from steatosis to steatohepatitis. Sci Rep 2019;9:7501. https://doi.org/10.1038/s41598-019-43928-7
crossref pmid pmc
18. Song J, Kang H, Cheng Y. Relationship between the cell death-inducing DNA fragmentation factor 45-like effector protein family and the risk of dyslipidemia. Front Biosci 2024;29:258. https://doi.org/10.31083/j.fbl2907258
crossref
19. Zhou L, Xu L, Ye J, et al. Cidea promotes hepatic steatosis by sensing dietary fatty acids. Hepatology 2012;56:95–107. https://doi.org/10.1002/hep.25611
crossref pmid
20. Zhou L, Park SY, Xu L, et al. Insulin resistance and white adipose tissue inflammation are uncoupled in energetically challenged Fsp27-deficient mice. Nat Commun 2015;6:5949. https://doi.org/10.1038/ncomms6949
crossref pmid pmc
21. Zhang LJ, Wang C, Yuan Y, et al. Cideb facilitates the lipidation of chylomicrons in the small intestine. J Lipid Res 2014;55:1279–87. https://doi.org/10.1194/jlr.M046482
crossref pmid pmc
22. Li X, Ye J, Zhou L, Gu W, Fisher EA, Li P. Opposing roles of cell death-inducing DFF45-like effector B and perilipin 2 in controlling hepatic VLDL lipidation. J Lipid Res 2012;53:1877–89. https://doi.org/10.1194/jlr.M026591
crossref pmid pmc
23. Su L, Zhou L, Chen FJ, et al. Cideb controls sterol-regulated ER export of SREBP/SCAP by promoting cargo loading at ER exit sites. EMBO J 2019;38:e100156. https://doi.org/10.15252/embj.2018100156
crossref pmid pmc
24. Ye J, Li JZ, Liu Y, et al. Cideb, an ER- and lipid droplet-associated protein, mediates VLDL lipidation and maturation by interacting with apolipoprotein B. Cell Metab 2009;9:177–90. https://doi.org/10.1016/j.cmet.2008.12.013
crossref pmid
25. Li JZ, Lei Y, Wang Y, et al. Control of cholesterol biosynthesis, uptake and storage in hepatocytes by Cideb. Biochim Biophys Acta Mol Cell Biol Lipids 2010;1801:577–86. https://doi.org/10.1016/j.bbalip.2010.01.012
crossref
26. Xu W, Wu L, Yu M, et al. Differential roles of cell death-inducing DNA fragmentation factor-α-like effector (cide) proteins in promoting lipid droplet fusion and growth in subpopulations of hepatocytes. J Biol Chem 2016;291:4282–93. https://doi.org/10.1074/jbc.M115.701094
crossref pmid pmc
27. Yu GE, Kwon S, Hwang JH, An SM, Park DH, Kang DG, et al. Effects of cell death-inducing DFF45-like effector B on meat quality traits in Berkshire pigs. Genet Mol Res 2017;16:gmr16029408. https://doi.org/10.4238/gmr16029408
crossref
28. Xu Q, Wang Y, Li X, et al. miR-10a-5p inhibits the differentiation of goat intramuscular preadipocytes by targeting KLF8 in goats. Front Mol Biosci 2021;8:700078. https://doi.org/10.3389/fmolb.2021.700078
crossref pmid pmc
29. Yang C, Li Q, Lin Y, et al. Diacylglycerol acyltransferase 2 promotes the adipogenesis of intramuscular preadipocytes in goat. Anim Biotechnol 2023;34:2376–83. https://doi.org/10.1080/10495398.2022.2091586
crossref pmid
30. Huang L, Luo J, Song N, Gao W, Zhu L, Yao W. CRISPR/Cas9-Mediated knockout of miR-130b affects mono- and polyunsaturated fatty acid content via PPARG-PGC1α axis in goat mammary epithelial cells. Int J Mol Sci 2022;23:3640. https://doi.org/10.3390/ijms23073640
crossref pmid pmc
31. Shi HB, Zhao WS, Luo J, et al. Peroxisome proliferator-activated receptor γ1 and γ2 isoforms alter lipogenic gene networks in goat mammary epithelial cells to different extents. J Dairy Sci 2014;97:5437–47. https://doi.org/10.3168/jds.2013-7863
crossref pmid
32. Kawagishi H, Wakoh T, Uno H, et al. Hzf regulates adipogenesis through translational control of C/EBPalpha. EMBO J 2008;27:1481–90. https://doi.org/10.1038/emboj.2008.76
crossref pmid pmc
33. MacDougald OA, Lane MD. Transcriptional regulation of gene expression during adipocyte differentiation. Annu Rev Biochem 1995;64:345–73. https://doi.org/10.1146/annurev.bi.64.070195.002021
crossref pmid
34. Mota de Sá P, Richard AJ, Hang H, Stephens JM. Transcriptional Regulation of Adipogenesis. Compr Physiol 2017;7:635–74. https://doi.org/10.1002/cphy.c160022
crossref pmid
35. Zhang F, Xiong Q, Tao H, et al. ACOX1, regulated by C/EBPα and miR-25-3p, promotes bovine preadipocyte adipogenesis. J Mol Endocrinol 2021;66:195–205. https://doi.org/10.1530/JME-20-0250
crossref pmid pmc
36. Wang W, Lv N, Zhang S, et al. Cidea is an essential transcriptional coactivator regulating mammary gland secretion of milk lipids. Nat Med 2012;18:235–43. https://doi.org/10.1038/nm.2614
crossref pmid
37. Chen Q, Fang W, Shen Y, et al. Suppression of cideb under endoplasmic reticulum stress exacerbated hepatic inflammation by inducing hepatic steatosis and oxidative stress. Free Radic Biol Med 2022;185:67–75. https://doi.org/10.1016/j.freeradbiomed.2022.04.009
crossref pmid
38. Park S, Baek IJ, Ryu JH, Chun CH, Jin EJ. PPARα-ACOT12 axis is responsible for maintaining cartilage homeostasis through modulating de novo lipogenesis. Nat Commun 2022;13:3. https://doi.org/10.1038/s41467-021-27738-y
crossref pmid pmc
39. Paglialunga S, Dehn CA. Clinical assessment of hepatic de novo lipogenesis in non-alcoholic fatty liver disease. Lipids Health Dis 2016;15:159. https://doi.org/10.1186/s12944-016-0321-5
crossref pmid pmc
40. Horibata Y, Ando H, Itoh M, Sugimoto H. Enzymatic and transcriptional regulation of the cytoplasmic acetyl-CoA hydrolase ACOT12. J Lipid Res 2013;54:2049–59. https://doi.org/10.1194/jlr.M030163
crossref pmid pmc
41. Sanders FW, Griffin JL. De novo lipogenesis in the liver in health and disease: more than just a shunting yard for glucose. Biol Rev 2016;91:452–68. https://doi.org/10.1111/brv.12178
crossref pmid pmc
42. Mitamura T, Palacpac NM. Lipid metabolism in Plasmodium falciparum-infected erythrocytes: possible new targets for malaria chemotherapy. Microbes Infect 2003;5:545–52. https://doi.org/10.1016/s1286-4579(03)00070-4
crossref pmid
43. Zheng XD, Huang Y, Li H. Regulatory role of Apelin-13-mediated PI3K/AKT signaling pathway in the glucose and lipid metabolism of mouse with gestational diabetes mellitus. Immunobiology 2021;226:152135. https://doi.org/10.1016/j.imbio.2021.152135
crossref pmid
44. Lee S, Dong HH. FoxO integration of insulin signaling with glucose and lipid metabolism. J Endocrinol 2017;233:R67–79. https://doi.org/10.1530/JOE-17-0002
crossref pmid pmc
45. Guasch-Ferré M, Santos JL, Martínez-González MA, et al. Glycolysis/gluconeogenesis- and tricarboxylic acid cycle-related metabolites, Mediterranean diet, and type 2 diabetes. Am J Clin Nutr 2020;111:835–44. https://doi.org/10.1093/ajcn/nqaa016
crossref pmid pmc
46. Gao N, Liu Y, Liu G, Liu B, Cheng Y. Sanghuangporus vaninii extract ameliorates hyperlipidemia in rats by mechanisms identified with transcriptome analysis. Food Sci Nutr 2024;12:3360–76. https://doi.org/10.1002/fsn3.4002
crossref pmid pmc
47. Wang L, Zhu H, Shi Z, et al. MK8722 initiates early-stage autophagy while inhibiting late-stage autophagy via FASN-dependent reprogramming of lipid metabolism. Theranostics 2024;14:75–95. https://doi.org/10.7150/thno.83051
crossref pmid pmc


Editorial Office
Asian-Australasian Association of Animal Production Societies(AAAP)
Room 708 Sammo Sporex, 23, Sillim-ro 59-gil, Gwanak-gu, Seoul 08776, Korea   
TEL : +82-2-888-6558    FAX : +82-2-888-6559   
E-mail : editor@animbiosci.org               

Copyright © 2025 by Asian-Australasian Association of Animal Production Societies.

Developed in M2PI

Close layer
prev next