Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
J Genet Genomics ; 2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38582298

ABSTRACT

The phenotypic diversity resulting from artificial or natural selection of sheep has made a significant contribution to human civilization. Hu sheep are a local sheep breed unique to China with high reproductive rates and rapid growth. Genome selection signatures have been widely used to investigate the genetic mechanisms underlying phenotypic variation in livestock. Here, we conduct whole-genome sequencing of 207 Hu sheep and compare them with the wild ancestors of domestic sheep (Asiatic mouflon) to investigate the genetic characteristics and selection signatures of Hu sheep. Based on six signatures of selection approaches, we detect genomic regions containing genes related to reproduction (BMPR1B, BMP2, PGFS, CYP19, CAMK4, GGT5, and GNAQ), vision (ALDH1A2, SAG, and PDE6B), nervous system (NAV1), and immune response (GPR35, SH2B2, PIK3R3, and HRAS). Association analysis with a population of 1299 Hu sheep reveal those missense mutations in the GPR35 (GPR35 g.952651 A>G; GPR35 g.952496 C>T) and NAV1 (NAV1 g.84216190 C>T; NAV1 g.84227412 G>A) genes are significantly associated (P < 0.05) with immune and growth traits in Hu sheep, respectively. This research offers unique insights into the selection characteristics of Hu sheep and facilitates further genetic improvement and molecular investigations.

2.
Front Microbiol ; 15: 1348873, 2024.
Article in English | MEDLINE | ID: mdl-38419634

ABSTRACT

Fecal scores are crucial for assessing the digestive and gastrointestinal status of animals. The Bristol fecal scoring system is a commonly used method for the subjective evaluation of host feces, there is limited research on fecal scoring standards for fattening Hu sheep. In this study, Hu sheep were collected for rumen, rectum, and colon contents for 16S rDNA sequencing. 514 Hu sheep feces were scored based on the Bristol fecal scoring system, and production performance at each stage was measured. Finally, we developed the scoring standard of the manure of Hu sheep in the fattening period (a total of five grades). The result shows that moisture content significantly increased with higher grades (p < 0.05). We analyzed the relationship between fecal scores and production traits, blood indices, muscle nutrients, and digestive tract microorganisms. The growth traits (body weight, body height, body length, average daily gain (ADG), and average daily feed intake (ADFI) during 80-180 days), body composition traits of the F3 group, and the carcass traits were found to be significantly higher (p < 0.05) than those of the F1 and F2 groups. There was no significant difference in gastrointestinal microflora diversity among all groups (p > 0.05). Significant differences were observed in Aspartate aminotransferase, Glucose, Total bilirubin, and Red Blood Cell Count between groups (p < 0.05). The mutton moisture content in group F4 was significantly higher than in the other groups, and the protein content was also the lowest (p < 0.05). The results of the correlation analysis demonstrated that Actinobacteria, Peptostreptococcaceae, Acidaminococcales, Gammaproteobacteria, and Proteobacteria were the significant bacteria affecting fecal scores. In addition, Muribaculaceae and Oscillospiraceae were identified as the noteworthy flora affecting growth performance and immunity. This study highlights the differences in production traits and blood indicators between fecal assessment groups and the complex relationship between intestinal microbiota and fecal characteristics in Hu sheep, suggesting potential impacts on animal performance and health, which suggest strategies for improved management.

3.
Mol Nutr Food Res ; 68(1): e2200842, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37990402

ABSTRACT

SCOPE: Consuming goat milk is known to benefit high-fat diet-fed and streptozocin (STZ)-induced diabetic rats, but the underlying mechanisms are unknown. This study is conducted to investigate the metabolic effects of a goat milk diet (a form of goat milk powder) on glucose homeostasis and pancreatic conditions in a mouse model of Type 2 diabetes mellitus (T2DM) induced by STZ. METHODS AND RESULTS: T2DM mice are fed with a goat-milk-based diet containing 10.3% w/w goat milk powder for 10 weeks for investigating the in vivo effects; a ß-cell line MIN6 cells are used to test the in vitro effects of digested goat milk (DGM). Goat milk diet improves the deleterious effects of STZ on fasting glucose levels and glucose tolerance, accelerates pancreatic structure recovery, and alters blood metabolites in mice. Based on the significant differences observed in metabolites, the key pathways, metabolite regulatory enzymes, metabolite molecular modules, and biochemical reactions are identified as critical integrated pathways. DGM promotes the cell activity, glucose transportation, and AKT activation in cultured STZ-treated MIN6 cells in vitro. CONCLUSIONS: Goat milk diet improves glucose homeostasis and pancreatic conditions of T2DM mice, in association with improved blood metabolite profiles and activation of pancreatic AKT pathway.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Mice , Rats , Animals , Diabetes Mellitus, Type 2/metabolism , Milk/chemistry , Diabetes Mellitus, Experimental/metabolism , Proto-Oncogene Proteins c-akt , Powders , Glucose/metabolism , Diet, High-Fat/adverse effects , Goats/metabolism , Blood Glucose/metabolism , Streptozocin , Insulin
4.
J Anim Sci ; 1012023 Jan 03.
Article in English | MEDLINE | ID: mdl-37742310

ABSTRACT

Feed efficiency is an important indicator in the sheep production process, which plays an important role in improving economic benefits and strengthening energy conservation and emission reduction. Compared with the rumen, the fermentation of the hindgut microorganisms can also provide part of the energy for the host, and the composition of the hindgut microorganisms will affect the feed efficiency. Therefore, we hope to find new ways to regulate sheep feed efficiency by studying the sheep gut microbes. In this study, male Hu sheep with the same birth date were raised under the same conditions until 180 d old. The sheep were divided into high and low groups according to the feed conversion ratio (FCR) at 80 to 180 d old, and the differences in rectal microorganisms between the two groups were compared. The permutational multivariate analysis (PERMANOVA) test showed that there were differences in microorganisms between the two groups (P < 0.05). Combined with linear fitting analysis, a total of six biomarkers were identified, including Ruminobacter, Eubacterium_xylanophilum_group, Romboutsia, etc. Functional enrichment analysis showed that microorganisms may affect FCR through volatile fatty acids synthesis and inflammatory response. At the same time, we conducted a longitudinal analysis of the hindgut microbes, sampling nine-time points throughout the sheep birth to market stages. The microbiota is clearly divided into two parts: before weaning and after weaning, and after weaning microbes are less affected by before weaning microbial composition.


The level of feed efficiency determines the input of sheep production costs and the income of economic benefits. Improving sheep feed efficiency can effectively save energy and reduce emissions. Gut microbes play an important role in the process of feed fermentation. In this study, biomarkers associated with feed efficiency were identified by exploring the relationship between microbes and feed conversion ratio. At the same time, the longitudinal development of microorganisms was explored. It provides a basis for the regulation of intestinal microbes in sheep.


Subject(s)
Microbiota , Animals , Male , Sheep , Fatty Acids, Volatile/metabolism , Weaning , Fermentation , Animal Feed/analysis , Rumen/metabolism
5.
Arch Anim Nutr ; 77(2): 155-169, 2023 Apr.
Article in English | MEDLINE | ID: mdl-37133408

ABSTRACT

Leucine improves the exocrine capacity of the cow pancreas, but the mechanism was not revealed clearly. Mitogen-activated protein kinase-interacting kinase 1 (MNK1) is a pancreatic acinar cell-specific stress response kinase that regulates digestive enzyme abundance. We aimed to investigate the MNK1 gene and protein expression profiles among various organs or tissues of dairy cows and to demonstrate the mechanism by which leucine-stimulated MNK1 regulates pancreatic exocrine function. Firstly, the expression profiles of MNK1 protein and gene in the tissues and organs of dairy cows were measured using immunohistochemistry and RT-qPCR methods. Next, an in vitro model of cultured Holstein dairy calf pancreatic acinar cells was used to detect the role of MNK1 during pancreatic enzymes release which is stimulated by leucine. Cells were incubated in culture medium containing L-leucine (0.45 mM) for 180 min, and samples were collected hourly, with the control not containing L-leucine (0 mM). MNK1 was expressed at very high levels in the pancreatic tissue of dairy cows. Leucine supplementation increased the α-amylase level but not lipase level at three time-points (60, 120, and 180 min), and the interaction between treatments and times was significant only for α-amylase. Leucine treatment enhanced (P < 0.05) the phosphorylation of MNK1 and eIF4E. In addition, inhibition of MNK1 decreased leucine-mediated α-amylase and lipase release (P < 0.05) and the phosphorylation of Mnk1 and eIF4E but did not affect (P > 0.05) the phosphorylation of the mTOR signalling pathway factors 4EBP1 and S6K1. In summary, MNK1 is a key regulator of pancreatic exocrine function, which is regulated by leucine in the pancreas of dairy cows.


Subject(s)
Acinar Cells , Eukaryotic Initiation Factor-4E , Female , Cattle , Animals , Acinar Cells/metabolism , Leucine/pharmacology , Eukaryotic Initiation Factor-4E/metabolism , Animal Feed/analysis , Diet/veterinary , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , alpha-Amylases/metabolism , Mitogen-Activated Protein Kinases/metabolism
6.
J Agric Food Chem ; 71(22): 8527-8539, 2023 Jun 07.
Article in English | MEDLINE | ID: mdl-37224334

ABSTRACT

Goat milk is increasingly recognized by consumers due to its high nutritional value, richness in short- and medium-chain fatty acids, and richness in polyunsaturated fatty acids (PUFA). Exogenous supplementation of docosahexaenoic acid (DHA) is an important approach to increasing the content of PUFA in goat milk. Several studies have reported benefits of dietary DHA in terms of human health, including potential against chronic diseases and tumors. However, the mechanisms whereby an increased supply of DHA regulates mammary cell function is unknown. In this study, we investigated the effect of DHA on lipid metabolism processes in goat mammary epithelial cells (GMEC) and the function of H3K9ac epigenetic modifications in this process. Supplementation of DHA promoted lipid droplet accumulation increased the DHA content and altered fatty acid composition in GMEC. Lipid metabolism processes were altered by DHA supplementation through transcriptional programs in GMEC. ChIP-seq analysis revealed that DHA induced genome-wide H3K9ac epigenetic changes in GMEC. Multiomics analyses (H3K9ac genome-wide screening and RNA-seq) revealed that DHA-induced expression of lipid metabolism genes (FASN, SCD1, FADS1, FADS2, LPIN1, DGAT1, MBOAT2), which were closely related with changes in lipid metabolism processes and fatty acid profiles, were regulated by modification of H3K9ac. In particular, DHA increased the enrichment of H3K9ac in the promoter region of PDK4 and promoted its transcription, while PDK4 inhibited lipid synthesis and activated AMPK signaling in GMEC. The activation of the expression of fatty acid metabolism-related genes FASN, FADS2, and SCD1 and their upstream transcription factor SREBP1 by the AMPK inhibitor was attenuated in PDK4-overexpressing GMEC. In conclusion, DHA alters lipid metabolism processes via H3K9ac modifications and the PDK4-AMPK-SREBP1 signaling axis in goat mammary epithelial cells, providing new insights into the mechanism through which DHA affects mammary cell function and regulates milk fat metabolism.


Subject(s)
Docosahexaenoic Acids , Lipid Metabolism , Humans , Animals , Docosahexaenoic Acids/pharmacology , Docosahexaenoic Acids/metabolism , AMP-Activated Protein Kinases/genetics , Triglycerides/metabolism , Fatty Acids/metabolism , Fatty Acids, Unsaturated/metabolism , Epigenesis, Genetic , Goats/genetics , Goats/metabolism , Mammary Glands, Animal/metabolism , Epithelial Cells/metabolism , Phosphatidate Phosphatase/genetics , Phosphatidate Phosphatase/metabolism
7.
Int J Mol Sci ; 24(2)2023 Jan 16.
Article in English | MEDLINE | ID: mdl-36675264

ABSTRACT

Embryos contain a large number of lipid droplets, and lipid metabolism is gradually activated during embryonic development to provide energy. However, the regulatory mechanisms remain to be investigated. Stearoyl-CoA desaturase 1 (Scd1) is a fatty acid desaturase gene that is mainly involved in intracellular monounsaturated fatty acid production, which takes part in many physiological processes. Analysis of transcripts at key stages of embryo development revealed that Scd1 was important and expressed at an increased level during the cleavage and blastocyst stages. Knockout Scd1 gene by CRISPR/Cas9 from zygotes revealed a decrease in lipid droplets (LDs) and damage in the inner cell mass (ICM) formation of blastocyst. Comparative analysis of normal and knockout embryo transcripts showed a suppression of ribosome protein (RPs) genes, leading to the arrest of ribosome biogenesis at the 2-cell stage. Notably, the P53-related pathway was further activated at the blastocyst stage, which eventually caused embryonic development arrest and apoptosis. In summary, Scd1 helps in providing energy for embryonic development by regulating intra-embryonic lipid droplet formation. Moreover, deficiency activates the RPs-Mdm2-P53 pathway due to ribosomal stress and ultimately leads to embryonic development arrest. The present results suggested that Scd1 gene is essential to maintain healthy development of embryos by regulating energy support.


Subject(s)
Lipid Metabolism , Tumor Suppressor Protein p53 , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Lipid Metabolism/genetics , Fatty Acids, Monounsaturated/metabolism , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Blastocyst/metabolism , Stearoyl-CoA Desaturase/genetics , Stearoyl-CoA Desaturase/metabolism
8.
J Anim Sci ; 1012023 Jan 03.
Article in English | MEDLINE | ID: mdl-36694375

ABSTRACT

Goat milk provides benefits to human health due to its richness in bioactive components, such as polyunsaturated fatty acids (PUFAs). The fatty acid desaturase 2 (FADS2) is the first rate-limiting enzyme in PUFAs biosynthesis. However, its role and transcriptional regulation mechanisms in fatty acid metabolism in dairy goat remain unclear. Here, our study revealed that the FADS2 gene was highly expressed during the peak lactation compared with the dry period, early lactation, and late lactation. The content of triacylglycerol (TAG) was enhanced with the increasing mRNA expression of TAG synthesis genes (diacylglycerol acyltransferase 1/2, DGAT1/2) in FADS2-overexpressed goat mammary epithelial cells (GMECs). Overexpression of FADS2 was positively correlated with the elevated concentrations of dihomo-gamma-linolenic acid (DGLA) and docosahexaenoic acid (DHA) in GMECs. BODIPY staining showed that FADS2 promoted lipid droplet accumulation in GMECs. To clarify the transcriptional regulatory mechanisms of FADS2, 2,226 bp length of FADS2 promoter was obtained. Deletion mutation assays revealed that the core region of FADS2 promoter was located between the -375 and -26 region, which contained SRE1 (-361/-351) and SRE2 (-191/-181) cis-acting elements of transcription factor sterol regulatory element-binding protein 1 (SREBP1). Overexpression of SREBP1 enhanced relative luciferase activity of the single mutant of SRE1 or SRE2, vice versa, and failed to alter the relative luciferase activity of the joint mutant of SRE1 and SRE2. Chromatin immunoprecipitation (ChIP) and site-directed mutation assays further demonstrated that SREBP1 regulated the transcription of the FADS2 gene by binding to SRE sites in vivo and in vitro. In addition, the mRNA levels of FADS2 were significantly decreased by targeting SRE1 and SRE2 sites in the genome via the CRISPR interference (CRISPRi) system. These findings establish a direct role for FADS2 regulating TAG and fatty acid synthesis by SREBP1 transcriptional regulation in dairy goat, providing new insights into fatty acid metabolism in mammary gland of ruminants.


The fatty acid desaturase 2 (FADS2) is the first rate-limiting enzyme in polyunsaturated fatty acids (PUFAs) biosynthesis in mammals. This study aimed to investigate the function and transcriptional regulation mechanism of FADS2 in goat mammary epithelial cells (GMECs). The content of triacylglycerol (TAG) was enhanced with lipid droplet accumulation in FADS2-overexpressed GMECs. Overexpression of FADS2 was positively correlated with elevated concentrations of docosahexaenoic acid (DHA) in GMECs. Furthermore, site-directed mutation and chromatin immunoprecipitation (ChIP) assays simultaneously demonstrated that FADS2 was directly regulated by SREBP1 transcriptional factor binding to sterol regulatory element (SRE) in vitro and in vivo. In addition, genetic ablation of SRE1 and SRE2 in the genome resulted in a significant reduction in the mRNA levels of FADS2 via the CRISPR interference (CRISPRi) system. Altogether, this study discovered that the SREBP1 exerts control on FADS2 to regulate milk fatty acids, and provides a theoretical approach for improving milk quality via genetic approaches.


Subject(s)
Fatty Acid Desaturases , Goats , Mammary Glands, Animal , Sterol Regulatory Element Binding Protein 1 , Animals , Female , Epithelial Cells/metabolism , Fatty Acid Desaturases/genetics , Fatty Acid Desaturases/metabolism , Fatty Acids/metabolism , Fatty Acids, Unsaturated/metabolism , Goats/genetics , Goats/metabolism , Luciferases/metabolism , Mammary Glands, Animal/metabolism , RNA, Messenger/genetics , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism
9.
J Anim Sci ; 1012023 Jan 03.
Article in English | MEDLINE | ID: mdl-36547378

ABSTRACT

CCAAT/enhancer binding protein α (C/EBPα) is the key transcription factor involved in lipid metabolism, however, the role of C/EBPα in milk fat synthesis of dairy goats remains unknown. The objective of the present research was to clarify the function of C/EBPα in goat mammary epithelial cells (GMECs) and its impact on peroxisome proliferator-activated receptor gamma (PPARG) promoter activity. In this study, C/EBPα overexpression increased its mRNA and protein levels by 42-fold and 6-fold, respectively. In contrast, transfecting siRNA targeting C/EBPα decreased its mRNA level to 20% and protein abundance to 80% of the basal level. The contents of lipid droplets, triacylglycerol (TAG), and cholesterol were increased (P < 0.05) in C/EBPα-overexpressing GMECs, and knockdown of C/EBPα led to the opposite results. Overexpression of C/EBPα significantly increased the expression levels of genes involved in TAG synthesis (AGPAT6, DGAT2, P < 0.01), lipid droplet formation (PLIN2, P < 0.01), and fatty acid synthesis (FADS2, P < 0.05; ELOVL6, P < 0.01). Knockdown of C/EBPα decreased (P < 0.05) the expression levels of AGPAT6, DGAT1, DGAT2, PLIN2, FADS2, and ELOVL6. C/EBPα upregulated the expression level of PPARG (P < 0.05), and four C/EBPα binding regions were identified in the PPARG promoter at -1,112 to -1,102 bp, -734 to -724 bp, -248 to -238 bp, and -119 to -109 bp. Knockdown of C/EBPα reduced (P < 0.05) the PPARG promoter activity when the C/EBPα binding regions were mutated at -1,112 to -1,102 bp, -734 to -724 bp, and -248 to -238 bp locations of the promoter. However, the promoter activity did not change when the mutation was located at -119 bp. In conclusion, our results suggest that C/EBPα can promote TAG synthesis in GMECs through its effects on mRNA abundance of genes related to lipid metabolism and regulation of the PPARG promoter activity via C/EBPα binding regions.


Goat milk is beneficial for human health because of its nutritional value, especially milk fat, which is plentiful in goat milk. The molecular mechanism of milk fat synthesis is of great importance for developing processing technology and using genetic approaches to improve goat milk quality. The purpose of this study was to identify the role of CCAAT/enhancer binding protein α (C/EBPα) in goat mammary gland epithelial cells (GMECs) to provide support for understanding the mechanism of milk fat synthesis. Overexpression of C/EBPα increased the contents of lipid droplets, triacylglycerol, and cholesterol in GMECs. The expression levels of genes related to lipid metabolism were influenced after C/EBPα overexpression or inhibition. The promoter transcriptional activity of peroxisome proliferator-activated receptor gamma, which is the key transcription factor in milk fat synthesis, was regulated by C/EBPα through its binding regions. Our results indicate that C/EBPα affects lipid metabolism in GMECs by regulating PPARG transcription. This study provides support for milk fat synthesis regulation and improvement of milk quality in goats.


Subject(s)
Fatty Acids , PPAR gamma , Animals , PPAR gamma/genetics , PPAR gamma/metabolism , Fatty Acids/metabolism , Goats/genetics , CCAAT-Enhancer-Binding Protein-alpha/genetics , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Mammary Glands, Animal/metabolism , Triglycerides/metabolism , Epithelial Cells/metabolism , RNA, Messenger/genetics
10.
PeerJ ; 10: e13945, 2022.
Article in English | MEDLINE | ID: mdl-36124130

ABSTRACT

Background: Lipid synthesis is an indispensable process during embryo and growth development. Abnormal lipid synthesis metabolism can cause multiple metabolic diseases including obesity and hyperlipidemia. Stearoyl-Coenzyme A desaturase 1 (SCD1) is responsible for catalyzing the synthesis of monounsaturated fatty acids (MUFA) and plays an essential role in lipid metabolism. The aim of our study was to evaluate the effects of SCD1 on embryo development and lipid synthesis in a knockout mice model. Methods: We used the CRISPR/Cas9 system together with microinjection for the knockout mouse model generation. Ten-week-old female C57BL/6 mice were used for zygote collection. RNase-free water was injected into mouse zygotes at different cell phases in order to select the optimal time for microinjection. Five sgRNAs were designed and in vitro transcription was performed to obtain sgRNAs and Cas9 mRNA. RNase-free water, NC sgRNA/Cas9 mRNA, and Scd1 sgRNA/Cas9 mRNA were injected into zygotes to observe the morula and blastocyst formation rates. Embryos that were injected with Scd1 sgRNA/Cas9 mRNA and developed to the two-cell stage were used for embryo transfer. Body weight, triacylglycerol (TAG), and cholesterol in Scd1 knockout mice serum were analyzed to determine the effects of SCD1 on lipid metabolism. Results: Microinjection performed during the S phase presented with the highest zygote survival rate (P < 0.05). Of the five sgRNAs targeted to Scd1, two sgRNAs with relatively higher gene editing efficiency were used for Scd1 knockout embryos and mice generation. Genome sequence modification was observed at Scd1 exons in embryos, and Scd1 knockout reduced blastocyst formation rates (P < 0.05). Three Scd1 monoallelic knockout mice were obtained. In mice, the protein level of SCD1 decreased (P < 0.05), and the body weight and serum TAG and cholesterol contents were all reduced (P < 0.01).


Subject(s)
CRISPR-Cas Systems , Embryonic Development , Animals , Female , Mice , CRISPR-Cas Systems/genetics , Mice, Inbred C57BL , Mice, Knockout , Embryonic Development/genetics , Fatty Acids, Monounsaturated/metabolism , Triglycerides/metabolism , Fatty Acid Desaturases/metabolism , Water/metabolism
11.
Front Genet ; 13: 980463, 2022.
Article in English | MEDLINE | ID: mdl-36160020

ABSTRACT

Goat milk is rich in fat and protein, thus, has high nutritional values and benefits human health. However, goaty flavour is a major concern that interferes with consumer acceptability of goat milk and the 4-alkyl-branched-chain fatty acids (vBCFAs) are the major substances relevant to the goaty flavour in goat milk. Previous research reported that the acyl-coenzyme A synthetases (ACSs) play a key role in the activation of fatty acids, which is a prerequisite for fatty acids entering anabolic and catabolic processes and highly involved in the regulation of vBCFAs metabolism. Although ACS genes have been identified in humans and mice, they have not been systematically characterized in goats. In this research, we performed genome-wide characterization of the ACS genes in goats, identifying that a total of 25 ACS genes (without ACSM2A) were obtained in the Capra hircus and each ACS protein contained the conserved AMP-binding domain. Phylogenetic analysis showed that out of the 25 genes, 21 belonged to the ACSS, ACSM, ACSL, ACSVL, and ACSBG subfamilies. However, AACS, AASDH, ACSF, and ACSF3 genes were not classified in the common evolutionary branch and belonged to the ACS superfamily. The genes in the same clade had similar conserved structures, motifs and protein domains. The expression analysis showed that the majority of ACS genes were expressed in multi tissues. The comparative analysis of expression patterns in non-lactation and lactation mammary glands of goat, sheep and cow indicated that ACSS2 and ACSF3 genes may participate in the formation mechanisms of goaty flavour in goat milk. In conclusion, current research provides important genomic resources and expression information for ACSs in goats, which will support further research on investigating the formation mechanisms of the goaty flavour in goat milk.

12.
Foods ; 11(14)2022 Jul 07.
Article in English | MEDLINE | ID: mdl-35885255

ABSTRACT

In nonruminants, microRNA (miRNA)-24 plays an important role in lipid metabolism in adipose tissue and the liver. Although the abundance of miR-24 in ruminant mammary glands is the highest during peak lactation, its potential role in regulating the synthesis and secretion of fat into milk is unclear. This study aimed to identify the function of miR-24 in these processes using CRISPR/Cas9 technology in primary goat mammary epithelial cells (GMEC). A single clone containing a 66-nucleotide deletion between two sgRNAs mediating double-strand break (DSB) sites was obtained. The abundance of miR-24-3p and miR-24-5p encoded by the deleted sequence was decreased, whereas the target genes INSIG1 and FASN increased. In addition, miR-24 knockout reduced the gene abundance of genes associated with fatty acid and TAG synthesis and transcription regulator. Similarly, the content of cholesterol and monounsaturated fatty acid (MUFA) C18:1 decreased, whereas that of polyunsaturated fatty acids (PUFA) C18:2, C20:3, C20:4 and C20:5 increased. Subsequently, knocking down of INSIG1 but not FASN reversed the effect of miR-24 knockout, indicating that miR-24 modulated cholesterol and fatty acid synthesis mainly by targeting INSIG1. Overall, the present in vitro data demonstrated a critical role for miR-24 in regulating lipid and fatty acid synthesis and highlighted the possibility of manipulating milk components in dairy goats.

13.
Foods ; 11(9)2022 Apr 29.
Article in English | MEDLINE | ID: mdl-35564013

ABSTRACT

Malonyl/acetyltransferase (MAT) is a crucial functional domain of fatty acid synthase (FASN), which plays a vital role in the de novo synthesis of fatty acids in vivo. Milk fatty acids are secreted by mammary epithelial cells. Mammary epithelial cells are the units of mammary gland development and function, and it is a common model for the study of mammary gland tissue development and lactation. This study aimed to investigate the effects of MAT deletion on the synthesis of triacylglycerol and medium-chain fatty acids. The MAT domain was knocked out by CRISPR/Cas9 in the goat mammary epithelial cells (GMECs), and in MAT knockout GMECs, the mRNA level of FASN was decreased by approximately 91.19% and the protein level decreased by 51.83%. The results showed that MAT deletion downregulated the contents of triacylglycerol and medium-chain fatty acids (p < 0.05) and increased the content of acetyl-Coenzyme A (acetyl-CoA) (p < 0.001). Explicit deletion of MAT resulted in significant drop of FASN, which resulted in downregulation of LPL, GPAM, DGAT2, PLIN2, XDH, ATGL, LXRα, and PPARγ genes in GMECs (p < 0.05). Meanwhile, mRNA expression levels of ACC, FASN, DGAT2, SREBP1, and LXRα decreased following treatment with acetyl-CoA (p < 0.05). Our data reveals that FASN plays critical roles in the synthesis of medium-chain fatty acids and triacylglycerol in GMECs.

14.
J Agric Food Chem ; 70(13): 4030-4043, 2022 Apr 06.
Article in English | MEDLINE | ID: mdl-35343224

ABSTRACT

Goat milk contains a rich source of nutrients, especially unsaturated fatty acids. However, the regulatory mechanism of milk fat and fatty acid synthesis remains unclear. Stearoyl-CoA desaturase 1 (SCD1) is the key enzyme catalyzing monounsaturated fatty acid synthesis and is essential for milk lipid metabolism. To explore milk lipid synthesis mechanism in vivo, SCD1-knockout goats were generated through CRISPR/Cas9 technology for the first time. SCD1 deficiency did not influence goat growth or serum biochemistry. Plasma phosphatidylcholines increased by lipidomics after SCD1 knockout in goats. Whole-blood RNA-seq indicated alterations in biosynthesis of unsaturated fatty acid synthesis, cAMP, ATPase activity, and Wnt signaling pathways. In SCD1-knockout goats, milk fat percentage and unsaturated fatty acid levels were reduced but other milk components were unchanged. Milk lipidomics revealed decreased triacylglycerols and diacylglycerols levels, and the differential abundance of lipids were enriched in glycerolipid, glycerophospholipids, and thermogenesis metabolism pathways. In milk fat globules, the expression levels of genes related to fatty acid and TAG synthesis including SREBP1 were reduced. ATP content and AMPK activity were promoted, and p-p70S6K protein level was suppressed in SCD1-knockout goat mammary epithelial cells, suggesting that SCD1 affected milk lipid metabolism by influencing AMPK-mTORC1/p70S6K-SREBP1 pathway. The integrative analysis of gene expression levels and lipidomics of milk revealed a crucial role of SCD1 in glycerolipids and glycerophospholipids metabolism pathways. Our observations indicated that SCD1 regulated the synthesis of milk fat and unsaturated fatty acid in goat by affecting lipid metabolism gene expression and lipid metabolic pathways. These findings would be essential for improving goat milk nutritional value which is beneficial to human health.


Subject(s)
Goats , Milk , Stearoyl-CoA Desaturase , Animals , CRISPR-Cas Systems , Fatty Acids/metabolism , Fatty Acids, Unsaturated/metabolism , Goats/metabolism , Milk/chemistry , Stearoyl-CoA Desaturase/genetics , Stearoyl-CoA Desaturase/metabolism
15.
J Agric Food Chem ; 69(18): 5353-5362, 2021 May 12.
Article in English | MEDLINE | ID: mdl-33939400

ABSTRACT

αS1-Casein (encoded by the CSN1S1 gene) is associated with higher rates of allergy than other milk protein components for humans. microRNAs (miRNAs) as small noncoding RNA molecules regulate gene expression and influence diverse biological processes. However, little is known about the regulation of milk protein synthesis by miRNAs in ruminants. In this study, we aim to investigate the regulatory roles of miR-204 family members (miR-204-5p and miR-211) on αS1-casein in goat mammary epithelial cells (GMEC). Here, we observed that the CSN1S1 mRNA level is upregulated, while miR-204-5p and miR-211 (miR-204-5p/-211) abundance is downregulated during peak lactation compared with middle lactation of dairy goats. We found that miR-204-5p/-211 synergistically inhibit αS1-casein expression via directly binding to the 3'-untranslated region (3'UTR) of CSN1S1 in GMEC. miR-204-5p/-211 increase ß-casein mRNA (CSN2) and protein abundance, as well as the signal transducer and activator of transcription 5a (STAT5a) activity. Further, miR-204-5p/-211 enhance ß-casein expression via the CSN1S1-STAT5a signaling axis and promote ß-casein transcription by activating the STAT5 response element located in the CSN2 promoter. In conclusion, miR-204-5p/-211 regulate αS1-casein and ß-casein synthesis via targeting CSN1S1 in GMEC, which provide the strategy for manipulating miR-204 family members to reduce milk allergy potential and improve ruminant milk quality for human consumption.


Subject(s)
Hypersensitivity , MicroRNAs , Animals , Caseins/genetics , Female , Goats/genetics , MicroRNAs/genetics , Milk
16.
J Dairy Sci ; 103(10): 9561-9572, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32828499

ABSTRACT

Compared with other milk proteins, αS1-casein (encoded by CSN1S1) is associated with higher rates of allergies in humans. Although some studies have revealed that CSN1S1 affects a variety of cellular physiological processes such as immune response and proliferation in various cells, whether CSN1S1 regulates other milk proteins in ruminants is not known. In this study, we observed a negative Pearson correlation between the contents of αS1-casein and ß-casein in goat milk. Thus, we used isolated primary goat mammary epithelial cells along with adenoviral infection or small interference RNA to alter abundance of CSN1S1 and examine its role in milk protein synthesis regulation. Overexpressing CSN1S1 through adenoviral transfection decreased ß-casein mRNA (CSN2) and protein abundance, whereas interference of CSN1S1 resulted in a significant increase in ß-casein abundance. CSN1S1 reduced phosphorylation level of Janus kinase 2 (JAK2) and signal transducer and activator of transcription 5a (STAT5a). The transcription factor STAT5a activates CSN2 transcription by binding with its promoter region and then promotes ß-casein synthesis. Furthermore, CSN1S1 inhibited CSN2 promoter activity and ß-casein synthesis by modulating the JAK2/STAT5a signaling pathway. No changes in abundance were detected for αS2-casein (CSN1S2), κ-casein (CSN3) and major whey proteins (LALBA, BLG). Overall, our results underscored a mechanism whereby CSN1S1 inhibits ß-casein synthesis via inhibition of STAT5a. The data suggested that knocking down CSN1S1 not only reduced the content of αS1-casein, but also increased the abundance of major milk proteins including ß-casein. Thus, the present study provides a theoretical basis for manipulating αS1-casein mRNA to improve quality of ruminant milk for human consumption.


Subject(s)
Allergens/analysis , Caseins/genetics , Gene Expression Regulation , Milk/chemistry , Animals , Caseins/metabolism , Female , Goats/metabolism , Milk Proteins/analysis
17.
Am J Physiol Cell Physiol ; 318(6): C1284-C1293, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32320287

ABSTRACT

The present study aimed to elucidate the mechanisms by which leucine impacts the secretion of pancreatic enzymes, especially amylase, by studying the proteomics profiles of pancreatic acinar (PA) cells from dairy cows. PA cells, the experimental model, were treated with four concentrations of leucine (0, 0.23, 0.45, and 0.90 mM). The abundance of different proteins in the four leucine treatment groups was detected. Label-free proteomic analysis enabled the identification of 1,906 proteins in all four treatment groups, and 1,350 of these proteins showed common expression across the groups. The primary effects of leucine supplementation were increased (P < 0.05) citrate synthase and ATPase activity, which enlarged the cytosolic ATP pool, and the upregulation of secretory protein 61 (Sec61) expression, which promoted protein secretion. In summary, these results suggest that leucine increases citrate synthase in the TCA cycle and ATPase activity and promotes the Sec signaling pathway to increase the exocrine function of PA cells.


Subject(s)
Acinar Cells/drug effects , Citric Acid Cycle/drug effects , Leucine/pharmacology , Pancreas, Exocrine/drug effects , Secretory Pathway/drug effects , Signal Transduction/drug effects , alpha-Amylases/metabolism , Acinar Cells/enzymology , Adenosine Triphosphate/metabolism , Animals , Animals, Newborn , Cattle , Cells, Cultured , Citrate (si)-Synthase/metabolism , Dairying , Male , Mitochondria/drug effects , Mitochondria/metabolism , Pancreas, Exocrine/enzymology , Proteomics , SEC Translocation Channels/metabolism
18.
J Agric Food Chem ; 68(18): 5138-5146, 2020 May 06.
Article in English | MEDLINE | ID: mdl-32299216

ABSTRACT

MiR-145 modulates fatty acid metabolism by regulating the expression of fatty acid metabolism-related genes in goat mammary epithelial cells. Previous studies using RNAi methods have clarified the function of miR-145 in lipogenesis. However, there are limiting factors such as short-term and inconsistent inhibition efficiency in RNAi method. On the basis of previous miR-145 functional studies, this study aims to knock out miR-145 and validate the function using CRISPR/Cas9 technology. We successfully obtained the single cell clone which had single nucleotide deletion around the Drosha processing site. The expression of miR-145 was significantly decreased, and the mRNA and protein expression of target gene INSIG1 were both increased by RT-qPCR and Western blot. The expression of fatty acid metabolism-associated gene (DGAT1, AGPAT6, TIP47, ADFP, CD36, ACSL1, ATGL, ACOX, CPT1A, FADS2, ELOVL5, PPARA, SCD1, FASN, and ACACA) were decreased. The contents of triacylglycerol and cholesterol were significantly inhibited. The percentage of C17:0 and C18:0 saturated fatty acid increased. Taken together, these data suggested that knockout of miR-145 could inhibit TAG and cholesterol contents and affect fatty acid composition through regulating the expression of fatty acid metabolism-related genes. These findings provide a sufficient theoretical basis for improving goat milk quality by miR-145.


Subject(s)
Epithelial Cells/metabolism , Fatty Acids/metabolism , Goats/genetics , Intracellular Signaling Peptides and Proteins/genetics , Mammary Glands, Animal/metabolism , MicroRNAs/genetics , Animals , CRISPR-Cas Systems , Female , Gene Knockout Techniques , Goats/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , MicroRNAs/metabolism
19.
J Anim Sci ; 98(3)2020 Mar 01.
Article in English | MEDLINE | ID: mdl-32067038

ABSTRACT

A key member of the nuclear receptor superfamily is the peroxisome proliferator-activated receptor alpha (PPARA) isoform, which in nonruminants is closely associated with fatty acid oxidation. Whether PPARA plays a role in milk fatty acid synthesis in ruminants is unknown. The main objective of the present study was to use primary goat mammary epithelial cells (GMEC) to activate PPARA via the agonist WY-14643 (WY) or to silence it via transfection of small-interfering RNA (siRNA). Three copies of the peroxisome proliferator-activated receptor response element (PPRE) contained in a luciferase reporter vector were transfected into GMEC followed by incubation with WY at 0, 10, 20, 30, 50, or 100 µM. A dose of 50 µM WY was most effective at activating PPRE without influencing PPARA mRNA abundance. Transfecting siRNA targeting PPARA decreased its mRNA abundance to 20% and protein level to 50% of basal levels. Use of WY upregulated FASN, SCD1, ACSL1, DGAT1, FABP4, and CD36 (1.1-, 1.5-, 2-, 1.4-, 1.5-, and 5-fold, respectively), but downregulated DGAT2 and PGC1A (-20% and -40%, respectively) abundance. In contrast, triacylglycerol concentration decreased and the content and desaturation index of C16:1 and C18:1 increased. Thus, activation of PPARA via WY appeared to channel fatty acids away from esterification. Knockdown of PPARA via siRNA downregulated ACACA, SCD1, AGPAT6, CD36, HSL, and SREBF1 (-43%, -67%, -16%, -56%, -26%, and -29%, respectively), but upregulated ACSL1, DGAT2, FABP3, and PGC1A (2-, 1.4-, 1.3-, and 2.5-fold, respectively) mRNA abundance. A decrease in the content and desaturation index of C16:1 and C18:1 coupled with an increase in triacylglycerol content accompanied those effects at the mRNA level. Overall, data suggest that PPARA could promote the synthesis of MUFA in GMEC through its effects on mRNA abundance of genes related to fatty acid synthesis, oxidation, transport, and triacylglycerol synthesis.


Subject(s)
Fatty Acids, Monounsaturated/metabolism , Goats , Mammary Glands, Animal/metabolism , PPAR alpha/metabolism , Animals , Epithelial Cells/metabolism , Fatty Acid-Binding Proteins , Fatty Acids/metabolism , Female , Gene Expression Regulation , Lipid Metabolism , Lipogenesis/genetics , Peroxisome Proliferators/metabolism , Sterol Regulatory Element Binding Protein 1 , Triglycerides/metabolism , Up-Regulation
20.
J Agric Food Chem ; 66(38): 10041-10052, 2018 Sep 26.
Article in English | MEDLINE | ID: mdl-30180552

ABSTRACT

Stearoyl-CoA desaturase 1 (SCD1) is a fatty acid desaturase catalyzing cis-double-bond formation in the Δ9 position to produce monounsaturated fatty acids essential for the synthesis of milk fat. Previous studies using RNAi methods have provided support for a role of SCD1 in goat mammary epithelial cells (GMEC); however, RNAi presents several limitations that might preclude a truthful understanding of the biological function of SCD1. To explore the function of SCD1 on fatty acid metabolism in GMEC, we used CRISPR-Cas9-mediated SCD1 knockout through non-homologous end-joining (NHEJ) and homology-directed repair (HDR) pathways in GMEC. We successfully introduced nucleotide deletions and mutations in the SCD1 gene locus through the NHEJ pathway and disrupted its second exon via insertion of an EGFP-PuroR segment using the HDR pathway. In clones derived from the latter, gene- and protein-expression data indicated that we obtained a monoallelic SCD1 knockout. A T7EN1-mediated assay revealed no off-targets in the surveyed sites. The contents of triacylglycerol and cholesterol and the desaturase index were significantly decreased as a consequence of SCD1 knockout. The deletion of SCD1 decreased the expression of other genes involved in de novo fatty acid synthesis, including SREBF1 and FASN, as well the fatty acid transporters FABP3 and FABP4. The downregulation of these genes partly explains the decrease of intracellular triacylglycerols. Our results indicate a successful SCD1 knockout in goat mammary cells using CRISPR-Cas9. The demonstration of the successful use of CRISPR-Cas9 in GMEC is an important step to producing transgenic goats to study mammary biology in vivo.


Subject(s)
Animals, Genetically Modified/genetics , Epithelial Cells/enzymology , Fatty Acids/metabolism , Gene Knockout Techniques/methods , Goats/genetics , Mammary Glands, Animal/enzymology , Stearoyl-CoA Desaturase/deficiency , Animals , Animals, Genetically Modified/metabolism , CRISPR-Cas Systems , Epithelial Cells/metabolism , Female , Gene Deletion , Goats/metabolism , Lipogenesis , Mammary Glands, Animal/cytology , Mammary Glands, Animal/metabolism , Milk/chemistry , Milk/metabolism , Stearoyl-CoA Desaturase/genetics , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Triglycerides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...