1
|
Mao Y, Su C, Yang H, Ma X, Zhao F, Qu B, Yang Y, Hou X, Zhao B, Cui Y. PI3K/AKT/mTORC1 signalling pathway regulates MMP9 gene activation via transcription factor NF-κB in mammary epithelial cells of dairy cows. Anim Biotechnol 2024; 35:2314100. [PMID: 38343377 DOI: 10.1080/10495398.2024.2314100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2024]
Abstract
Matrix metalloproteinase 9 (MMP9) plays a pivotal role in mammary ductal morphogenesis, angiogenesis and glandular tissue architecture remodeling. However, the molecular mechanism of MMP9 expression in mammary epithelial cells of dairy cows remains unclear. This study aimed to explore the underlying mechanism of MMP9 expression. In this study, to determine whether the PI3K/AKT/mTORC1/NF-κB signalling pathway participates in the regulation of MMP9 expression, we treated mammary epithelial cells with specific pharmacological inhibitors of PI3K (LY294002), mTORC1 (Rapamycin) or NF-κB (Celastrol), respectively. Western blotting results indicated that LY294002, Rapamycin and Celastrol markedly decreased MMP9 expression and P65 nuclear translocation. Furthermore, we found that NF-κB (P65) overexpression resulted in elevated expression of MMP9 protein and activation of MMP9 promoter. In addition, we observed that Celastrol markedly decreases P65-overexpression-induced MMP9 promoter activity. Moreover, the results of the promoter assay indicated that the core regulation sequence for MMP9 promoter activation may be located at -420 ∼ -80 bp downstream from the transcription start site. These observations indicated that the PI3K/AKT/mTORC1 signalling pathway is involved in MMP9 expression by regulating MMP9 promoter activity via NF-κB in the mammary epithelial cells of dairy cows.
Collapse
Affiliation(s)
- Yongjin Mao
- Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin, PR China
| | - Chen Su
- Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin, PR China
| | - Huilin Yang
- Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin, PR China
| | - Xiaocong Ma
- Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin, PR China
| | - Feng Zhao
- Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin, PR China
| | - Bo Qu
- Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin, PR China
| | - Yang Yang
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin, PR China
| | - Xiaoming Hou
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin, PR China
| | - Bing Zhao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, PR China
| | - Yingjun Cui
- Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin, PR China
| |
Collapse
|
2
|
Zeng WH, Wen ZY, Wei XY, He Y, Zhou L, Hu P, Shi QC, Qin CJ, Wang J, Li R, Jing XY, Hu W, Yuan HW, Fan JD, Zhang C, Jiang W, Fu P, Shi Q. Molecular characterization, spatio-temporal expression patterns of crtc2 gene and its immune roles in yellow catfish (Pelteobagrus fulvidraco). FISH & SHELLFISH IMMUNOLOGY 2024; 154:109877. [PMID: 39245185 DOI: 10.1016/j.fsi.2024.109877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 08/30/2024] [Accepted: 09/02/2024] [Indexed: 09/10/2024]
Abstract
cAMP response element binding (CREB) protein 2 (CRTC2) is a transcriptional coactivator of CREB and plays an important role in the immune system. Thus far, the physiological roles of Crtc2 in teleost are still poorly understood. In this study, the crtc2 gene was identified and characterized from yellow catfish (Pelteobagrus fulvidraco; therefore, the gene is termed as pfcrtc2), and its evolutionary and molecular characteristics as well as potential immunity-related roles were investigated. Our results showed that the open reading frame of pfcrtc2 was 2346 bp in length, encoding a protein with 781 amino acids. Gene structure analysis revealed its existence of 14 exons and 13 introns. A phylogenetic analysis proved that the tree of crtc2 was clustered into five groups, exhibiting a similar evolutionary topology with species evolution. Multiple protein sequences alignment demonstrated high conservation of the crtc2 in various vertebrates with similar structure. Syntenic and gene structural comparisons further established that crtc2 was highly conserved, implying its similar roles in diverse vertebrates. Tissue distribution pattern detected by quantitative real-time PCR showed that the pfcrtc2 gene was almost expressed in all detected tissues except for eyes, with the highest expression levels in the gonad, indicating that Crtc2 may play important roles in various tissues. In addition, pfcrtc2 was transcribed at all developmental stages in yellow catfish, showing the highest expression levels at 12 h after fertilization. Finally, the transcriptional profiles of crtc2 were significantly increased in yellow catfishes injected with Aeromonas hydrophila or Poly I:C, which shared a consistent change pattern with four immune-related genes including IL-17A, IL-10, MAPKp38, and NF-κBp65, suggesting pfCrtc2 may play critical roles in preventing both exogenous bacteria and virus invasion. In summary, our findings lay a solid foundation for further studies on the functions of pfcrtc2, and provide novel genetic loci for developing new strategies to control disease outbreak in teleost.
Collapse
Affiliation(s)
- Wan-Hong Zeng
- Key Laboratory of Sichuan Province for Fishes Conservation and Utilization in the Upper Reaches of the Yangtze River, Neijiang Normal University, Neijiang, 641100, China; College of Life Science, Neijiang Normal University, Neijiang, 641100, China; School of Animal Science, Yangtze University, Jingzhou, 424020, China
| | - Zheng-Yong Wen
- Key Laboratory of Sichuan Province for Fishes Conservation and Utilization in the Upper Reaches of the Yangtze River, Neijiang Normal University, Neijiang, 641100, China; College of Life Science, Neijiang Normal University, Neijiang, 641100, China; School of Animal Science, Yangtze University, Jingzhou, 424020, China; Shenzhen Key Lab of Marine Genomics, Guangdong Provincial Key Lab of Molecular Breeding in Marine Economic Animals, BGI Academy of Marine Sciences, BGI Marine, BGI, Shenzhen, 518083, China.
| | - Xiu-Ying Wei
- Key Laboratory of Sichuan Province for Fishes Conservation and Utilization in the Upper Reaches of the Yangtze River, Neijiang Normal University, Neijiang, 641100, China; College of Life Science, Neijiang Normal University, Neijiang, 641100, China
| | - Yu He
- Key Laboratory of Sichuan Province for Fishes Conservation and Utilization in the Upper Reaches of the Yangtze River, Neijiang Normal University, Neijiang, 641100, China; College of Life Science, Neijiang Normal University, Neijiang, 641100, China; School of Animal Science, Yangtze University, Jingzhou, 424020, China
| | - Luo Zhou
- Key Laboratory of Sichuan Province for Fishes Conservation and Utilization in the Upper Reaches of the Yangtze River, Neijiang Normal University, Neijiang, 641100, China; College of Life Science, Neijiang Normal University, Neijiang, 641100, China; School of Animal Science, Yangtze University, Jingzhou, 424020, China
| | - Peng Hu
- Key Laboratory of Sichuan Province for Fishes Conservation and Utilization in the Upper Reaches of the Yangtze River, Neijiang Normal University, Neijiang, 641100, China; College of Life Science, Neijiang Normal University, Neijiang, 641100, China
| | - Qing-Chao Shi
- Key Laboratory of Sichuan Province for Fishes Conservation and Utilization in the Upper Reaches of the Yangtze River, Neijiang Normal University, Neijiang, 641100, China; College of Life Science, Neijiang Normal University, Neijiang, 641100, China
| | - Chuan-Jie Qin
- Key Laboratory of Sichuan Province for Fishes Conservation and Utilization in the Upper Reaches of the Yangtze River, Neijiang Normal University, Neijiang, 641100, China; College of Life Science, Neijiang Normal University, Neijiang, 641100, China
| | - Jun Wang
- Key Laboratory of Sichuan Province for Fishes Conservation and Utilization in the Upper Reaches of the Yangtze River, Neijiang Normal University, Neijiang, 641100, China; College of Life Science, Neijiang Normal University, Neijiang, 641100, China
| | - Rui Li
- Key Laboratory of Sichuan Province for Fishes Conservation and Utilization in the Upper Reaches of the Yangtze River, Neijiang Normal University, Neijiang, 641100, China; College of Life Science, Neijiang Normal University, Neijiang, 641100, China
| | - Xiao-Ying Jing
- Key Laboratory of Sichuan Province for Fishes Conservation and Utilization in the Upper Reaches of the Yangtze River, Neijiang Normal University, Neijiang, 641100, China; College of Life Science, Neijiang Normal University, Neijiang, 641100, China
| | - Wei Hu
- School of Animal Science, Yangtze University, Jingzhou, 424020, China
| | - Han-Wen Yuan
- School of Animal Science, Yangtze University, Jingzhou, 424020, China
| | - Jun-De Fan
- Chongqing Fisheries Science Research Institute, Chongqing, 400020, China
| | - Chuang Zhang
- Yueyang Yumeikang Biotechnology Co. Ltd., Yueyang, 414100, China
| | - Wei Jiang
- Yueyang Yumeikang Biotechnology Co. Ltd., Yueyang, 414100, China
| | - Peng Fu
- Yueyang Yumeikang Biotechnology Co. Ltd., Yueyang, 414100, China.
| | - Qiong Shi
- Key Laboratory of Sichuan Province for Fishes Conservation and Utilization in the Upper Reaches of the Yangtze River, Neijiang Normal University, Neijiang, 641100, China; College of Life Science, Neijiang Normal University, Neijiang, 641100, China; Shenzhen Key Lab of Marine Genomics, Guangdong Provincial Key Lab of Molecular Breeding in Marine Economic Animals, BGI Academy of Marine Sciences, BGI Marine, BGI, Shenzhen, 518083, China; Laboratory of Aquatic Genomics, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518060, China.
| |
Collapse
|
3
|
Meng Y, Lyu CC, He YT, Che HY, Jiang H, Zhang JB, Tang HY, Yuan B. ALG5 Regulates STF-62247-Induced Milk Fat Synthesis via the mTOR Signaling Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:14620-14629. [PMID: 38885170 DOI: 10.1021/acs.jafc.3c07812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
Milk fat content is a critical indicator of milk quality. Exploring the key regulatory genes involved in milk fat synthesis is essential for enhancing milk fat content. STF-62247 (STF), a thiazolamide compound, has the potential to bind with ALG5 and upregulate lipid droplets in fat synthesis. However, the effect of STF on the process of milk fat synthesis and whether it acts through ALG5 remains unknown. In this study, the impact of ALG5 on milk fat synthesis and its underlying mechanism were investigated using bovine mammary epithelial cells (BMECs) and mouse models through real-time PCR, western blotting, Oil Red O staining, and triglyceride analysis. Experimental findings revealed a positive correlation between STF and ALG5 with the ability to synthesize milk fat. Silencing ALG5 led to decreased expression of FASN, SREBP1, and PPARγ in BMECs, as well as reduced phosphorylation levels in the PI3K/AKT/mTOR signaling pathway. Moreover, the phosphorylation levels of the PI3K/AKT/mTOR signaling pathway were restored when ALG5 silencing was followed by the addition of STF. These results suggest that STF regulates fatty acid synthesis in BMECs by affecting the PI3K/AKT/mTOR signaling pathway through ALG5. ALG5 is possibly a new factor in milk fat synthesis.
Collapse
Affiliation(s)
- Yu Meng
- Department of Laboratory Animals, College of Animal Sciences, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun 130062 Jilin, P. R. China
| | - Chen-Chen Lyu
- Department of Laboratory Animals, College of Animal Sciences, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun 130062 Jilin, P. R. China
| | - Yun-Tong He
- Department of Laboratory Animals, College of Animal Sciences, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun 130062 Jilin, P. R. China
| | - Hao-Yu Che
- Department of Laboratory Animals, College of Animal Sciences, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun 130062 Jilin, P. R. China
| | - Hao Jiang
- Department of Laboratory Animals, College of Animal Sciences, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun 130062 Jilin, P. R. China
| | - Jia-Bao Zhang
- Department of Laboratory Animals, College of Animal Sciences, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun 130062 Jilin, P. R. China
| | - Hong-Yu Tang
- College of Animal Sciences, Jilin University, Changchun 130062, Jilin, P. R. China
| | - Bao Yuan
- Department of Laboratory Animals, College of Animal Sciences, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun 130062 Jilin, P. R. China
| |
Collapse
|
4
|
Wu HY, Ji ZH, Xie WY, Guo HX, Zheng Y, Gao W, Yuan B. KLF4 promotes milk fat synthesis by regulating the PI3K-AKT-mTOR pathway and targeting FASN activation in bovine mammary epithelial cells. iScience 2024; 27:109850. [PMID: 38779481 PMCID: PMC11108978 DOI: 10.1016/j.isci.2024.109850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 03/18/2024] [Accepted: 04/26/2024] [Indexed: 05/25/2024] Open
Abstract
Milk fat is an important indicator for evaluating the quality of cow's milk. In this study, we used bovine mammary epithelial cells (BMECs) to investigate the role and molecular mechanism of KLF4 in the regulation of milk fat synthesis. The results showed that KLF4 was more highly expressed in mammary tissues of high-fat cows compared with low-fat cows. KLF4 positively regulated the expression of genes related to milk fat synthesis in BMECs, increasing intracellular triglycerides content, and KLF4 promoted milk fat synthesis by activating the PI3K-AKT-mTOR signaling pathway. Furthermore, the results of animal experiments also confirmed that knockdown of KLF4 inhibited milk fat synthesis. In addition, yeast one-hybrid assays and dual-luciferase reporter gene assays confirmed that KLF4 directly targets and binds to the fatty acid synthase (FASN) promoter region to promote FASN transcription. These results demonstrate that KLF4 is a key transcription factor for milk fat synthesis in BMECs.
Collapse
Affiliation(s)
- Hong-Yu Wu
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun, Jilin 130062, China
- Jilin Academy of Agricultural Sciences, Jilin 132101, China
| | - Zhong-Hao Ji
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun, Jilin 130062, China
- Department of Basic Medicine, Changzhi Medical College, Changzhi 046000, Shanxi, China
| | - Wen-Yin Xie
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun, Jilin 130062, China
| | - Hai-Xiang Guo
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun, Jilin 130062, China
| | - Yi Zheng
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun, Jilin 130062, China
| | - Wei Gao
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun, Jilin 130062, China
| | - Bao Yuan
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun, Jilin 130062, China
| |
Collapse
|
5
|
Guo Y, Wei Z, Zhang Y, Cao J. Research Progress on the Mechanism of Milk Fat Synthesis in Cows and the Effect of Conjugated Linoleic Acid on Milk Fat Metabolism and Its Underlying Mechanism: A Review. Animals (Basel) 2024; 14:204. [PMID: 38254373 PMCID: PMC10812695 DOI: 10.3390/ani14020204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 12/29/2023] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
Milk fat synthesis in cows mainly includes the synthesis of short- and medium-chain fatty acids, the uptake, transport, and activation of long-chain fatty acids (LCFAs), the synthesis of triglycerides, and the synthesis of the genes, transcription factors, and signaling pathways involved. Although the various stages of milk fat synthesis have been outlined in previous research, only partial processes have been revealed. CLA consists of an aggregation of positional and geometric isomers of linoleic fatty acid, and the accumulated evidence suggests that the two isomers of the active forms of CLA (cis-9, trans-11 conjugated linoleic acid and trans-10, cis-12 conjugated linoleic acid, abbreviated as c9, t11-CLA and t10, c12-CLA) can reduce the fat content in milk by regulating lipogenesis, fatty acid (FA) uptake, oxidation, and fat synthesis. However, the mechanism through which CLA inhibits milk fat synthesis is unique, with most studies focusing only on the effects of CLA on one of the genes, transcription factors, or signaling pathways involved. In this study, we summarized the structure and function of classic genes and pathways (mTOR, SREBP, AMPK, and PPARG) and new genes or pathways (THRSP, METTL3, ELOVL, and LPIN1) involved in each stage of milk fat synthesis and demonstrated the interactions between genes and pathways. We also examined the effects of other substances (melanin, nicotinic acid, SA, etc.). Furthermore, we evaluated the influence of β-sitosterol, sodium butyrate, Met arginine, and Camellia oleifera Abel on milk fat synthesis to improve the mechanism of milk fat synthesis in cows and provide a mechanistic reference for the use of CLA in inhibiting milk fat biosynthesis.
Collapse
Affiliation(s)
- Yuanyin Guo
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (Y.G.); (Z.W.)
| | - Ziang Wei
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (Y.G.); (Z.W.)
| | - Yi Zhang
- College of Animal Science and Technology, China Agricultural University, Beijing 100193, China;
| | - Jie Cao
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (Y.G.); (Z.W.)
| |
Collapse
|
6
|
Qi H, Lin G, Guo S, Guo X, Yu C, Zhang M, Gao X. Met stimulates ARID1A degradation and activation of the PI3K-SREBP1 signaling to promote milk fat synthesis in bovine mammary epithelial cells. Anim Biotechnol 2023; 34:4094-4104. [PMID: 37837279 DOI: 10.1080/10495398.2023.2265167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2023]
Abstract
Methionine (Met) can promote milk fat synthesis in bovine mammary epithelial cells (BMECs), but the potential molecular mechanism is largely unknown. In this report, we aim to explore the role and molecular mechanism of AT-rich interaction domain 1A (ARID1A) in milk fat synthesis stimulated by Met. ARID1A knockdown and activation indicated that ARID1A negatively regulated the synthesis of triglycerides, cholesterol and free fatty acids and the formation of lipid droplets in BMECs. ARID1A also negatively regulated the phosphorylation of PI3K and AKT proteins, as well as the expression and maturation of SREBP1. Met stimulated the phosphorylation of PI3K and AKT proteins, as well as the expression and maturation of SREBP1, while ARID1A gene activation blocked the stimulatory effects of Met. We further found that ARID1A was located in the nucleus of BMECs, and Met reduced the nuclear localization and expression of ARID1A. ARID1A gene activation blocked the stimulation of PI3K and SREBP1 mRNA expression by Met. In summary, our data suggests that ARID1A negatively regulates milk fat synthesis stimulated by Met in BMECs through inhibiting the PI3K-SREBP1 signaling pathway, which may provide some new perspectives for improving milk fat synthesis.
Collapse
Affiliation(s)
- Hao Qi
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, College of Life Science and Engineering, Foshan University, Foshan, Guangdong, China
| | - Gang Lin
- College of Animal Science, Yangtze University, Jingzhou, Hubei, China
| | - Siqi Guo
- College of Life Science, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Xudong Guo
- College of Animal Science, Yangtze University, Jingzhou, Hubei, China
| | - Congying Yu
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, College of Life Science and Engineering, Foshan University, Foshan, Guangdong, China
| | - Minghui Zhang
- College of Animal Science, Yangtze University, Jingzhou, Hubei, China
| | - Xuejun Gao
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, College of Life Science and Engineering, Foshan University, Foshan, Guangdong, China
- College of Animal Science, Yangtze University, Jingzhou, Hubei, China
| |
Collapse
|
7
|
Huang B, Khan MZ, Kou X, Chen Y, Liang H, Ullah Q, Khan N, Khan A, Chai W, Wang C. Enhancing Metabolism and Milk Production Performance in Periparturient Dairy Cattle through Rumen-Protected Methionine and Choline Supplementation. Metabolites 2023; 13:1080. [PMID: 37887405 PMCID: PMC10608895 DOI: 10.3390/metabo13101080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/07/2023] [Accepted: 10/10/2023] [Indexed: 10/28/2023] Open
Abstract
For dairy cattle to perform well throughout and following lactations, precise dietary control during the periparturient phase is crucial. The primary issues experienced by periparturient dairy cows include issues like decreased dry matter intake (DMI), a negative energy balance, higher levels of non-esterified fatty acids (NEFA), and the ensuing inferior milk output. Dairy cattle have always been fed a diet high in crude protein (CP) to produce the most milk possible. Despite the vital function that dairy cows play in the conversion of dietary CP into milk, a sizeable percentage of nitrogen is inevitably expelled, which raises serious environmental concerns. To reduce nitrogen emissions and their production, lactating dairy cows must receive less CP supplementation. Supplementing dairy cattle with rumen-protected methionine (RPM) and choline (RPC) has proven to be a successful method for improving their ability to use nitrogen, regulate their metabolism, and produce milk. The detrimental effects of low dietary protein consumption on the milk yield, protein yield, and dry matter intake may be mitigated by these nutritional treatments. In metabolic activities like the synthesis of sulfur-containing amino acids and methylation reactions, RPM and RPC are crucial players. Methionine, a limiting amino acid, affects the production of milk protein and the success of lactation in general. According to the existing data in the literature, methionine supplementation has a favorable impact on the pathways that produce milk. Similarly, choline is essential for DNA methylation, cell membrane stability, and lipid metabolism. Furthermore, RPC supplementation during the transition phase improves dry matter intake, postpartum milk yield, and fat-corrected milk (FCM) production. This review provides comprehensive insights into the roles of RPM and RPC in optimizing nitrogen utilization, metabolism, and enhancing milk production performance in periparturient dairy cattle, offering valuable strategies for sustainable dairy farming practices.
Collapse
Affiliation(s)
- Bingjian Huang
- Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, Liaocheng University, Liaocheng 252000, China
- College of Life Sciences, Liaocheng University, Liaocheng 252059, China
| | - Muhammad Zahoor Khan
- Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, Liaocheng University, Liaocheng 252000, China
- Faculty of Veterinary and Animal Sciences, University of Agriculture, Dera Ismail Khan 29220, Pakistan
| | - Xiyan Kou
- Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, Liaocheng University, Liaocheng 252000, China
| | - Yinghui Chen
- Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, Liaocheng University, Liaocheng 252000, China
| | - Huili Liang
- Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, Liaocheng University, Liaocheng 252000, China
| | - Qudrat Ullah
- Faculty of Veterinary and Animal Sciences, University of Agriculture, Dera Ismail Khan 29220, Pakistan
| | - Nadar Khan
- Livestock and Dairy Development (Research) Department Khyber Pakhtunkhwa, Peshawar 25120, Pakistan
| | - Adnan Khan
- Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 511464, China
| | - Wenqiong Chai
- Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, Liaocheng University, Liaocheng 252000, China
| | - Changfa Wang
- Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, Liaocheng University, Liaocheng 252000, China
| |
Collapse
|
8
|
Li Q, Chen J, Liu J, Lin T, Liu X, Zhang S, Yue X, Zhang X, Zeng X, Ren M, Guan W, Zhang S. Leucine and arginine enhance milk fat and milk protein synthesis via the CaSR/G i/mTORC1 and CaSR/G q/mTORC1 pathways. Eur J Nutr 2023; 62:2873-2890. [PMID: 37392244 DOI: 10.1007/s00394-023-03197-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 06/23/2023] [Indexed: 07/03/2023]
Abstract
BACKGROUND AND AIMS Amino acids (AAs) not only constitute milk protein but also stimulate milk synthesis through the activation of mTORC1 signaling, but which amino acids that have the greatest impact on milk fat and protein synthesis is still very limited. In this study, we aimed to identify the most critical AAs involved in the regulation of milk synthesis and clarify how these AAs regulate milk synthesis through the G-protein-coupled receptors (GPCRs) signaling pathway. METHODS In this study, a mouse mammary epithelial cell line (HC11) and porcine mammary epithelial cells (PMECs) were selected as study subjects. After treatment with different AAs, the amount of milk protein and milk fat synthesis were detected. Activation of mTORC1 and GPCRs signaling induced by AAs was also investigated. RESULTS In this study, we demonstrate that essential amino acids (EAAs) are crucial to promote lactation by increasing the expression of genes and proteins related to milk synthesis, such as ACACA, FABP4, DGAT1, SREBP1, α-casein, β-casein, and WAP in HC11 cells and PMECs. In addition to activating mTORC1, EAAs uniquely regulate the expression of calcium-sensing receptor (CaSR) among all amino-acid-responsive GPCRs, which indicates a potential link between CaSR and the mTORC1 pathway in mammary gland epithelial cells. Compared with other EAAs, leucine and arginine had the greatest capacity to trigger GPCRs (p-ERK) and mTORC1 (p-S6K1) signaling in HC11 cells. In addition, CaSR and its downstream G proteins Gi, Gq, and Gβγ are involved in the regulation of leucine- and arginine-induced milk synthesis and mTORC1 activation. Taken together, our data suggest that leucine and arginine can efficiently trigger milk synthesis through the CaSR/Gi/mTORC1 and CaSR/Gq/mTORC1 pathways. CONCLUSION We found that the G-protein-coupled receptor CaSR is an important amino acid sensor in mammary epithelial cells. Leucine and arginine promote milk synthesis partially through the CaSR/Gi/mTORC1 and CaSR/Gq/mTORC1 signaling systems in mammary gland epithelial cells. Although this mechanism needs further verification, it is foreseeable that this mechanism may provide new insights into the regulation of milk synthesis.
Collapse
Affiliation(s)
- Qihui Li
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Jiaming Chen
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Jiaxin Liu
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Tongbin Lin
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Xinghong Liu
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Shuchang Zhang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Xianhuai Yue
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Xiaoli Zhang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Xiangfang Zeng
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture and Rural Affairs Feed Industry Center, China Agricultural University, Beijing, China
| | - Man Ren
- Anhui Provincial Key Laboratory of Animal Nutritional Regulation and Health, College of Animal Science, Anhui Science and Technology University, Fengyang, China
| | - Wutai Guan
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, China
| | - Shihai Zhang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China.
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, 510642, China.
- Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, China.
| |
Collapse
|
9
|
Li B, Khan MZ, Khan IM, Ullah Q, Cisang ZM, Zhang N, Wu D, Huang B, Ma Y, Khan A, Jiang N, Zahoor M. Genetics, environmental stress, and amino acid supplementation affect lactational performance via mTOR signaling pathway in bovine mammary epithelial cells. Front Genet 2023; 14:1195774. [PMID: 37636261 PMCID: PMC10448190 DOI: 10.3389/fgene.2023.1195774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 06/26/2023] [Indexed: 08/29/2023] Open
Abstract
Mammary glands are known for their ability to convert nutrients present in the blood into milk contents. In cows, milk synthesis and the proliferation of cow mammary epithelial cells (CMECs) are regulated by various factors, including nutrients such as amino acids and glucose, hormones, and environmental stress. Amino acids, in particular, play a crucial role in regulating cell proliferation and casein synthesis in mammalian epithelial cells, apart from being building blocks for protein synthesis. Studies have shown that environmental factors, particularly heat stress, can negatively impact milk production performance in dairy cattle. The mammalian target of rapamycin complex 1 (mTORC1) pathway is considered the primary signaling pathway involved in regulating cell proliferation and milk protein and fat synthesis in cow mammary epithelial cells in response to amino acids and heat stress. Given the significant role played by the mTORC signaling pathway in milk synthesis and cell proliferation, this article briefly discusses the main regulatory genes, the impact of amino acids and heat stress on milk production performance, and the regulation of mTORC signaling pathway in cow mammary epithelial cells.
Collapse
Affiliation(s)
- Bin Li
- Institute of Animal Husbandry and Veterinary, Tibet Autonomous Regional Academy of Agricultural Sciences, Lhasa, China
| | - Muhammad Zahoor Khan
- Liaocheng Research Institute of Donkey High‐Efficiency Breeding and Ecological Feeding, Agricultural Science and Engineering School, Liaocheng University, Liaocheng, China
- Faculty of Veterinary and Animal Sciences, The University of Agriculture, Dera Ismail Khan, Pakistan
| | - Ibrar Muhammad Khan
- Anhui Province Key Laboratory of Embryo Development and Reproduction Regulation, Anhui Province Key Laboratory of Environmental Hormone and Reproduction, School of Biological and Food Engineering, Fuyang Normal University, Fuyang, China
| | - Qudrat Ullah
- Faculty of Veterinary and Animal Sciences, The University of Agriculture, Dera Ismail Khan, Pakistan
| | - Zhuo-Ma Cisang
- Institute of Animal Husbandry and Veterinary, Tibet Autonomous Regional Academy of Agricultural Sciences, Lhasa, China
| | - Nan Zhang
- Tibet Autonomous Region Animal Husbandry Station, Lhasa, China
| | - Dan Wu
- Institute of Animal Husbandry and Veterinary, Tibet Autonomous Regional Academy of Agricultural Sciences, Lhasa, China
| | - Bingjian Huang
- Liaocheng Research Institute of Donkey High‐Efficiency Breeding and Ecological Feeding, Agricultural Science and Engineering School, Liaocheng University, Liaocheng, China
- College of Life Sciences, Liaocheng University, Liaocheng, China
| | - Yulin Ma
- State Key Laboratory of Animal Nutrition, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Adnan Khan
- Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Nan Jiang
- Institute of Animal Husbandry and Veterinary, Tibet Autonomous Regional Academy of Agricultural Sciences, Lhasa, China
| | - Muhammad Zahoor
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| |
Collapse
|
10
|
Lin G, Qi H, Guo X, Wang W, Zhang M, Gao X. ARID1B blocks methionine-stimulated mTOR activation to inhibit milk fat and protein synthesis in and proliferation of mouse mammary epithelial cells. J Nutr Biochem 2023; 114:109274. [PMID: 36681308 DOI: 10.1016/j.jnutbio.2023.109274] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 12/03/2022] [Accepted: 01/13/2023] [Indexed: 01/20/2023]
Abstract
Met can function through the mTOR signaling pathway, but the molecular mechanism is not fully understood. Here we investigated the role of ARID1B in this regulatory process. ARID1B knockdown promoted milk fat and protein synthesis in and cell proliferation of HC11 cells and increased mTOR mRNA expression and protein phosphorylation, whereas ARID1B gene activation had the opposite effects. ARID1B gene activation totally blocked Met's stimulation on mTOR mRNA expression. ARID1B bound to one region of the mTOR promoter, and Met reduced the binding of ARID1B on this promoter. LY294002 blocked Met-induced reduction of ARID1B mRNA and protein level. Cycloheximide treatment did not affect the decrease of ARID1B by Met. MG132 but not chloroquine restored ARID1B degradation induced by Met. Our data reveal that ARID1B is a key negative regulator of milk fat and protein synthesis in and proliferation of HC11 cells, and blocks Met-stimulated mTOR gene transcription.
Collapse
Affiliation(s)
- Gang Lin
- College of Animal Science, Yangtze University, Jingzhou, China
| | - Hao Qi
- College of Animal Science, Yangtze University, Jingzhou, China
| | - Xudong Guo
- College of Animal Science, Yangtze University, Jingzhou, China
| | - Wenqiang Wang
- College of Animal Science, Yangtze University, Jingzhou, China
| | - Minghui Zhang
- College of Animal Science, Yangtze University, Jingzhou, China
| | - Xuejun Gao
- College of Animal Science, Yangtze University, Jingzhou, China.
| |
Collapse
|
11
|
Ke C, Zhao S, Wang L, Zhang M, Gao X. Chromatin remodeler BRM is a key mediator of leucine-stimulated mTOR gene transcription in mouse mammary epithelial cells. Biochem Biophys Res Commun 2023; 643:88-95. [PMID: 36587526 DOI: 10.1016/j.bbrc.2022.12.064] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 12/21/2022] [Indexed: 12/27/2022]
Abstract
Brahma (BRM) is one of the core ATPase subunits of SWI/SNF chromatin remodeling complex, and participates in various important cellular regulatory processes. However, the role of BRM in regulating gene expression of the mechanistic target of rapamycin (mTOR) still remains unknown. In this study, we explored the effects and the corresponding molecular mechanisms of BRM on Leucine (Leu)-stimulated mTOR activation in and proliferation of a mouse mammary epithelial cell (MEC) line (HC11 cell). Initially, we found that the abundance of BRM protein in mammary gland tissue during lactation was significantly higher than that during puberty and involution. BRM knockdown inhibited HC11 cell proliferation, mRNA expression of mTOR and subsequent protein phosphorylation, whereas BRM gene activation had the opposite effect. Leu affected the level of BRM protein and mTOR phospphorylation in a dose-dependent manner, and BRM knockdown totally blocked the stimulation of Leu on mTOR mRNA expression and protein phospphorylation. ChIP-PCR detected that BRM was bound to the -4368 ∼ -4591 bp site of the mTOR promoter, and ChIP-qPCR further detected that Leu stimulated BRM to bind to this site. In conclusion, these data reveal that BRM is a positive regulator of HC11 cell proliferation and mediates Leu's stimulation on mTOR gene transcription and protein phosphorylation. Our data provide a new theoretical basis for the involvement of BRM in cell proliferation and regulation of the mTOR signaling pathway.
Collapse
Affiliation(s)
- Changping Ke
- College of Animal Science, Yangtze University, Jingzhou, 434025, China
| | - Sunqi Zhao
- College of Animal Science, Yangtze University, Jingzhou, 434025, China
| | - Lulu Wang
- College of Animal Science, Yangtze University, Jingzhou, 434025, China
| | - Minghui Zhang
- College of Animal Science, Yangtze University, Jingzhou, 434025, China.
| | - Xuejun Gao
- College of Animal Science, Yangtze University, Jingzhou, 434025, China.
| |
Collapse
|
12
|
Pan F, Li P, Hao G, Liu Y, Wang T, Liu B. Enhancing Milk Production by Nutrient Supplements: Strategies and Regulatory Pathways. Animals (Basel) 2023; 13:ani13030419. [PMID: 36766308 PMCID: PMC9913681 DOI: 10.3390/ani13030419] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 01/10/2023] [Accepted: 01/23/2023] [Indexed: 01/28/2023] Open
Abstract
The enhancement of milk production is essential for dairy animals, and nutrient supplements can enhance milk production. This work summarizes the influence of nutrient supplements-including amino acids, peptides, lipids, carbohydrates, and other chemicals (such as phenolic compounds, prolactin, estrogen and growth factors)-on milk production. We also attempt to provide possible illuminating insights into the subsequent effects of nutrient supplements on milk synthesis. This work may help understand the strategy and the regulatory pathway of milk production promotion. Specifically, we summarize the roles and related pathways of nutrients in promoting milk protein and fat synthesis. We hope this review will help people understand the relationship between nutritional supplementation and milk production.
Collapse
Affiliation(s)
- Fengguang Pan
- Laboratory of Nutrition and Functional Food, College of Food Science and Engineering, Jilin University, Changchun 130062, China
| | - Peizhi Li
- Laboratory of Nutrition and Functional Food, College of Food Science and Engineering, Jilin University, Changchun 130062, China
| | - Guijie Hao
- Key Laboratory of Healthy Freshwater Aquaculture, Ministry of Agriculture and Rural Affairs, Huzhou 313001, China
- Key Laboratory of Fish Health and Nutrition of Zhejiang Province, Zhejiang Institute of Freshwater Fisheries, Huzhou 313001, China
| | - Yinuo Liu
- Key Laboratory of Genetics and Breeding, Zhejiang Institute of Freshwater Fisheries, Huzhou 313001, China
| | - Tian Wang
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun 130062, China
- Correspondence: (T.W.); (B.L.)
| | - Boqun Liu
- Laboratory of Nutrition and Functional Food, College of Food Science and Engineering, Jilin University, Changchun 130062, China
- Correspondence: (T.W.); (B.L.)
| |
Collapse
|
13
|
Luo C, Li N, Wang Q, Li C. Sodium acetate promotes fat synthesis by suppressing TATA element modulatory factor 1 in bovine mammary epithelial cells. ANIMAL NUTRITION 2023; 13:126-136. [PMID: 37123620 PMCID: PMC10130354 DOI: 10.1016/j.aninu.2023.01.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 01/03/2023] [Accepted: 01/06/2023] [Indexed: 01/13/2023]
Abstract
Short-chain fatty acids are important nutrients that regulate milk fat synthesis. They regulate milk synthesis via the sterol regulatory element binding protein 1 (SREBP1) pathway; however, the details are still unknown. Here, the regulation and mechanism of sodium acetate (SA) in milk fat synthesis in bovine mammary epithelial cells (BMECs) were assessed. BMECs were treated with SA supplementation (SA+) or without SA supplementation (SA-), and milk fat synthesis and activation of the SREBP1 pathway were increased (P = 0.0045; P = 0.0042) by SA+ and decreased (P = 0.0068; P = 0.0031) by SA-, respectively. Overexpression or inhibition of SREBP1 demonstrated that SA promoted milk fat synthesis (P = 0.0045) via the SREBP1 pathway. Overexpression or inhibition of TATA element modulatory factor 1 (TMF1) demonstrated that TMF1 suppressed activation of the SREBP1 pathway (P = 0.0001) and milk fat synthesis (P = 0.0022) activated by SA+. Overexpression or inhibition of TMF1 and SREBP1 showed that TMF1 suppressed milk fat synthesis (P = 0.0073) through the SREBP1 pathway. Coimmunoprecipitation analysis revealed that TMF1 interacted with SREBP1 in the cytoplasm and suppressed the nuclear localization of SREBP1 (P = 0.0066). The absence or presence of SA demonstrated that SA inhibited the expression of TMF1 (P = 0.0002) and the interaction between TMF1 and SREBP1 (P = 0.0001). Collectively, our research suggested that TMF1 was a new negative regulator of milk fat synthesis. In BMECs, SA promoted the SREBP1 pathway and milk fat synthesis by suppressing TMF1. This study enhances the current understanding of the regulation of milk fat synthesis and provides new scientific data for the regulation of milk fat synthesis.
Collapse
|
14
|
Wang F, van Baal J, Ma L, Gao X, Dijkstra J, Bu D. MRCKα is a novel regulator of prolactin-induced lactogenesis in bovine mammary epithelial cells. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2022; 10:319-328. [PMID: 35891685 PMCID: PMC9304597 DOI: 10.1016/j.aninu.2022.06.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 01/18/2022] [Accepted: 06/02/2022] [Indexed: 06/15/2023]
Abstract
Myotonic dystrophy-related Cdc42-binding kinase alpha (MRCKα) is an integral component of signaling pathways controlling vital cellular processes, including cytoskeletal reorganization, cell proliferation and cell survival. In this study, we investigated the physiological role of MRCKα in milk protein and fat production in dairy cows, which requires a dynamic and strict organization of the cytoskeletal network in bovine mammary epithelial cells (BMEC). Within a selection of 9 Holstein cows, we found that both mRNA and protein expression of MRCKα in the mammary gland were upregulated during lactation and correlated positively (r > 0.89) with the mRNA and protein levels of β-casein. Similar positive correlations (r > 0.79) were found in a primary culture of BMEC stimulated with prolactin for 24 h. In these cells, silencing of MRCKα decreased basal β-casein, sterol-regulatory element binding protein (SREBP)-1 and cyclin D1 protein level, phosphorylation of mTOR, triglyceride secretion, cell number and viability-while overexpression of MRCKα displayed the reversed effect. Notably, silencing of MRCKα completely prevented the stimulatory action of prolactin on the same parameters. These data demonstrate that MRCKα is a critical mediator of prolactin-induced lactogenesis via stimulation of the mTOR/SREBP1/cyclin D1 signaling pathway.
Collapse
Affiliation(s)
- Fang Wang
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
- Animal Nutrition Group, Wageningen University and Research, Wageningen, 6708, WD, the Netherlands
| | - Jürgen van Baal
- Animal Nutrition Group, Wageningen University and Research, Wageningen, 6708, WD, the Netherlands
| | - Lu Ma
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Xuejun Gao
- College of Animal Science, Yangtze University, Jingzhou, 434020, China
| | - Jan Dijkstra
- Animal Nutrition Group, Wageningen University and Research, Wageningen, 6708, WD, the Netherlands
| | - Dengpan Bu
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
- Joint Laboratory on Integrated Crop-Tree-Livestock Systems of the Chinese Academy of Agricultural Sciences (CAAS), Ethiopian Institute of Agricultural Research (EIAR) and World Agroforestry Center (ICRAF), Beijing, 100193, China
| |
Collapse
|
15
|
Jin X, Zhen Z, Wang Z, Gao X, Li M. GPRC6A is a key mediator of palmitic acid regulation of lipid synthesis in bovine mammary epithelial cells. Cell Biol Int 2022; 46:1747-1758. [PMID: 35979663 DOI: 10.1002/cbin.11886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 12/29/2021] [Accepted: 03/07/2022] [Indexed: 11/07/2022]
Abstract
Fatty acids (FAs) can promote lipid synthesis in the mammary gland via stimulating lipogenic gene expression, but the underlying molecular mechanism is still not fully understood. Here, we showed the dose-dependent effects of palmitic acid (PA) on lipid synthesis in primary bovine mammary epithelial cells (BMECs) and explored the corresponding molecular mechanism. BMECs were treated with PA (0, 50, 100, 150, and 200 μM), and the 100 μM treatment had the best stimulatory effect on lipid synthesis and expression and maturation of sterol regulatory element-binding protein 1c (SREBP-1c) in cells. Inhibition of phosphatidylinositol 3-kinase (PI3K) almost totally blocked the stimulation of PA on SREBP-1c expression, whereas protein kinase Cα (PKCα) knockdown only partially decreased the stimulation of PA on SREBP-1c expression but abolished the stimulation of PA on its maturation. Knockdown of GPR120 did not change the stimulation of PA on the SREBP-1c signaling. G protein-coupled receptor family C group 6 member A (GPRC6A) knockdown almost totally blocked the stimulation of FA on PI3K and PKCα phosphorylation as well as SREBP-1c expression and maturation. Furthermore, PA dose-dependently promoted GPRC6A expression and plasma membrane localization. Together, these above results reveal that GPRC6A is a key mediator of PA signaling to lipid synthesis in BMECs via the PI3K/PKCα-SREBP-1c pathways.
Collapse
Affiliation(s)
- Xin Jin
- College of Life Science, Northeast Agricultural University, Harbin, China
| | - Zhen Zhen
- College of Life Science, Northeast Agricultural University, Harbin, China
| | - Zhaoxiong Wang
- College of Animal Science, Yangtze University, Jingzhou, China
| | - Xuejun Gao
- College of Animal Science, Yangtze University, Jingzhou, China
| | - Meng Li
- College of Life Science, Northeast Agricultural University, Harbin, China
| |
Collapse
|
16
|
Hao Q, Wang Z, Wang L, Han M, Zhang M, Gao X. Isoleucine stimulates mTOR and SREBP-1c gene expression for milk synthesis in mammary epithelial cells through BRG1-mediated chromatin remodelling. Br J Nutr 2022; 129:1-11. [PMID: 35593529 DOI: 10.1017/s0007114522001544] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Several amino acids can stimulate milk synthesis in mammary epithelial cells (MEC); however, the regulatory role of isoleucine (Ile) and underlying molecular mechanism remain poorly understood. In this study, we aimed to evaluate the regulatory effects of Ile on milk protein and fat synthesis in MEC and reveal the mediation mechanism of Brahma-related gene 1 (BRG1) on this regulation. Ile dose dependently affected milk protein and fat synthesis, mechanistic target of rapamycin (mTOR) phosphorylation, sterol regulatory element binding protein 1c (SREBP-1c) expression and maturation, and BRG1 protein expression in bovine MEC. Phosphatidylinositol 3 kinase (PI3K) inhibition by LY294002 treatment blocked the stimulation of Ile on BRG1 expression. BRG1 knockdown and gene activation experiments showed that it mediated the stimulation of Ile on milk protein and fat synthesis, mTOR phosphorylation, and SREBP-1c expression and maturation in MEC. ChIP-PCR analysis detected that BRG1 bound to the promoters of mTOR and SREBP-1c, and ChIP-qPCR further detected that these bindings were increased by Ile stimulation. In addition, BRG1 positively regulated the binding of H3K27ac to these two promoters, while it negatively affected the binding of H3K27me3 to these promoters. BRG1 knockdown blocked the stimulation of Ile on these two gene expressions. The expression of BRG1 was higher in mouse mammary gland in the lactating period, compared with that in the puberty or dry period. Taken together, these experimental data reveal that Ile stimulates milk protein and fat synthesis in MEC via the PI3K-BRG1-mTOR/SREBP-1c pathway.
Collapse
Affiliation(s)
- Qi Hao
- College of Animal Science, Yangtze University, Jingzhou434023, People's Republic of China
| | - Zhe Wang
- College of Animal Science, Yangtze University, Jingzhou434023, People's Republic of China
- College of Life Science, Northeast Agricultural University, Harbin150030, People's Republic of China
| | - Lulu Wang
- College of Animal Science, Yangtze University, Jingzhou434023, People's Republic of China
| | - Meihong Han
- College of Animal Science, Yangtze University, Jingzhou434023, People's Republic of China
| | - Minghui Zhang
- College of Animal Science, Yangtze University, Jingzhou434023, People's Republic of China
- College of Life Science, Northeast Agricultural University, Harbin150030, People's Republic of China
| | - Xuejun Gao
- College of Animal Science, Yangtze University, Jingzhou434023, People's Republic of China
| |
Collapse
|
17
|
Johnson CT, de Abreu GHD, Mackie K, Lu HC, Bradshaw HB. Cannabinoids accumulate in mouse breast milk and differentially regulate lipid composition and lipid signaling molecules involved in infant development. BBA ADVANCES 2022; 2:100054. [PMID: 36643901 PMCID: PMC9835790 DOI: 10.1016/j.bbadva.2022.100054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Maternal cannabis use during lactation may expose developing infants to cannabinoids (CBs) such as Δ-9-tetrahydrocannabinol (THC) and cannabidiol (CBD). CBs modulate lipid signaling molecules in the central nervous system in age- and cell-dependent ways, but their influence on the lipid composition of breast milk has yet to be established. This study investigates the effects of THC, CBD, or their combination on milk lipids by analyzing the stomach contents of CD1 mouse pups that have been nursed by dams injected with CBs on postnatal days (PND) 1 -10. Stomach contents were collected 2 hours after the last injection on PND10 and HPLC/MS/MS was used to identify and quantify over 80 endogenous lipid species and cannabinoids in the samples. We show that CBs differentially accumulate in milk, lead to widespread decreases in free fatty acids, decreases in N-acyl methionine species, increases N-linoleoyl species, as well as modulate levels of endogenous CBs (eCBs) AEA, 2-AG, and their structural congeners. Our data indicate the passage of CBs to pups through breast milk and that maternal CB exposure alters breast milk lipid compositions.
Collapse
Affiliation(s)
- Clare T Johnson
- Psychological and Brain Sciences, Indiana University, Bloomington IN, United States
| | | | - Ken Mackie
- Psychological and Brain Sciences, Indiana University, Bloomington IN, United States
- Gill Center for Molecular Neuroscience, Indiana University, Bloomington IN, United States
| | - Hui-Chen Lu
- Psychological and Brain Sciences, Indiana University, Bloomington IN, United States
- Gill Center for Molecular Neuroscience, Indiana University, Bloomington IN, United States
| | - Heather B Bradshaw
- Psychological and Brain Sciences, Indiana University, Bloomington IN, United States
- Corresponding author.
| |
Collapse
|
18
|
Yang Q, Mao Y, Wang J, Yu H, Zhang X, Pei X, Duan Z, Xiao C, Ma M. Gestational bisphenol A exposure impairs hepatic lipid metabolism by altering mTOR/CRTC2/SREBP1 in male rat offspring. Hum Exp Toxicol 2022; 41:9603271221129852. [PMID: 36137816 DOI: 10.1177/09603271221129852] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Lipid metabolism is an important biochemical process in the body. Recent studies have found that environmental endocrine disruptors play an important role in the regulation of lipid metabolism. Bisphenol A (BPA), a common environmental endocrine disruptor, has adverse effects on lipid metabolism, but the mechanism is still unclear. This study aimed to investigate the effects of gestational BPA exposure on hepatic lipid metabolism and its possible mechanism in male offspring. The pregnant Sprague-Dawley rats were exposed to BPA (0, 0.05, 0.5, 5 mg/kg/day) from day 5 to day 19 of gestation to investigate the levels of triglyceride (TG) and total cholesterol (TC), and the expression of liver lipid metabolism-related genes in male offspring rats. The results showed that compared with the control group, the TG and TC levels in serum and liver in BPA-exposed groups was increased. And the expressions of liver fatty acid oxidation related genes, such as peroxisome proliferators-activated receptor α (PPARα) and carnitine palmitoyl transferase 1α (CPT1α), were down-regulated. However, the expressions of fatty acid synthesis related genes, such as sterol regulatory element binding proteins 1 (SREBP-1), acetyl-CoA carboxylase 1 (ACC1), fatty acid synthase (FAS) and stearoyl-CoA desaturase 1 (SCD-1), were up-regulated. The increased protein levels of mTOR and p-CRTC2 suggested that CREB-regulated transcription coactivator 2 (CRTC2) might be an important mediator in the mTOR/SREBP-1 pathway. In conclusion, these results demonstrated that mTOR/CRTC2/SREBP-1 could be affected by gestational BPA exposure, which may involve in the lipid metabolic disorders in later life.
Collapse
Affiliation(s)
- Q Yang
- Department of Toxicology, School of Public Heath, 70577Shenyang Medical College, Shenyang, China
| | - Y Mao
- Department of Toxicology, School of Public Heath, 70577Shenyang Medical College, Shenyang, China
| | - J Wang
- Department of Toxicology, School of Public Heath, 70577Shenyang Medical College, Shenyang, China
| | - H Yu
- Department of Toxicology, School of Public Heath, 70577Shenyang Medical College, Shenyang, China
| | - X Zhang
- Department of Toxicology, School of Public Heath, 70577Shenyang Medical College, Shenyang, China
| | - X Pei
- Department of Toxicology, School of Public Heath, 70577Shenyang Medical College, Shenyang, China
| | - Z Duan
- Department of Toxicology, School of Public Heath, 70577Shenyang Medical College, Shenyang, China
| | - C Xiao
- Department of Key Laboratory of Environmental Pollution and Microecology, 70577Shenyang Medical College, Shenyang, China
| | - M Ma
- Department of Toxicology, School of Public Heath, 70577Shenyang Medical College, Shenyang, China.,Department of Key Laboratory of Environmental Pollution and Microecology, 70577Shenyang Medical College, Shenyang, China
| |
Collapse
|
19
|
Methionine and leucine induce ARID1A degradation to promote mTOR expression and milk synthesis in mammary epithelial cells. J Nutr Biochem 2021; 101:108924. [PMID: 34843932 DOI: 10.1016/j.jnutbio.2021.108924] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 09/26/2021] [Accepted: 11/12/2021] [Indexed: 12/13/2022]
Abstract
Amino acids can activate mTOR to promote milk synthesis in mammary epithelial cells (MECs), but the underlying molecular mechanism is still largely unknown. The objective is to investigate the regulatory mechanism of amino acids (Met and Leu) in stimulating mRNA expression of mTOR in MECs. We found that the protein abundance of AT-rich interaction domain 1A (ARID1A) was poorly expressed in mouse mammary gland tissues of lactating period. ARID1A knockdown and gene activation experiments detected whether ARID1A negatively regulated milk protein and fat synthesis in bovine MECs, cell proliferation and the expression and activation of mTOR. ChIP-PCR detected that ARID1A, H3K27ac, H3K27me3 and H3K4me3 all bound to the mTOR promoter at -548∼-793 nt. Knockdown or gene activation of ARID1A enhanced or weakened the binding of H3K27ac on the mTOR promoter, respectively. The stimulation of Met and Leu on mTOR expression and phosphorylation were eliminated by ARID1A gene activation. Furthermore, Met and Leu decreased the protein level of ARID1A through ubiquitination and proteasomal degradation. TRIM21 bound to ARID1A, and TRIM21 knockdown blocked the stimulation of Met and Leu on ARID1A degradation. In summary, these data reveal that ARID1A blocks Met and Leu signaling to mTOR gene transcription through inhibiting H3K27ac deposition on its promoter, and Met and Leu decrease ARID1A protein level through TRIM21-mediated ubiquitination and proteasomal degradation. Our findings uncover that Met and Leu promote mTOR expression for milk synthesis through the TRIM21-ARID1A signaling pathway, providing a novel theoretical basis for the application of amino acids in milk production.
Collapse
|
20
|
Evodiamine Relieve LPS-Induced Mastitis by Inhibiting AKT/NF-κB p65 and MAPK Signaling Pathways. Inflammation 2021; 45:129-142. [PMID: 34401976 DOI: 10.1007/s10753-021-01533-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/28/2021] [Accepted: 08/03/2021] [Indexed: 01/15/2023]
Abstract
Evodiamine, an alkaloid component in the fruit of Evodia, has been shown to have biological functions such as antioxidant and anti-inflammatory. But whether evodiamine plays an improvement role on mastitis has not been studied. To investigate the effect and mechanism of evodiamine on lipopolysaccharide (LPS)-induced mastitis was the purpose of this study. In animal experiments, the mouse mastitis model was established by injecting LPS into the canals of the mammary gland. The results showed that evodiamine could significantly relieve the pathological injury of breast tissue and the production of pro-inflammatory cytokines and inhibit the activation of inflammation-related pathways such as AKT, NF-κB p65, ERK1/2, p38, and JNK. In cell experiments, the mouse mammary epithelial cells (mMECs) were incubated with evodiamine for 1 h and then stimulated with LPS. Next, pro-inflammatory mediators and inflammation-related signal pathways were detected. As expected, our results showed that evodiamine notably ameliorated the inflammatory reaction and inhibit the activation of related signaling pathways of mMECs. All the results suggested that evodiamine inhibited inflammation by inhibiting the phosphorylation of AKT, NF-κBp65, ERK1/2, p38, and JNK thus the LPS-induced mastitis was ameliorated. These findings suggest that evodiamine maybe a potential drug for mastitis because of its anti-inflammatory effects.
Collapse
|
21
|
Kan X, Hu G, Huang B, Guo W, Huang Y, Chen Y, Xu P, Cai X, Fu S, Liu J. Pedunculoside protects against LPS-induced mastitis in mice by inhibiting inflammation and maintaining the integrity of blood-milk barrier. Aging (Albany NY) 2021; 13:19460-19474. [PMID: 34383710 PMCID: PMC8386561 DOI: 10.18632/aging.203357] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 06/14/2021] [Indexed: 04/17/2023]
Abstract
Mastitis is a disease that seriously threatens the health of the mammary gland after delivery. Pedunculoside (PE) is the main bioactive component of Aquifoliaceae. The purpose of this experiment is to explore the effects of PE on mastitis and its underlying mechanisms. Our research results showed that PE could significantly inhibit the increase in the levels of inflammatory mediators such as TNF-α, IL-6, IL-1β, MPO and iNOS during mastitis. Mechanism studies have found that PE could significantly inhibit the phosphorylation of AKT protein and binds to the ASP-184 site. Further research found that PE also inhibited the activation of AKT's downstream pro-inflammatory signals NF-κB and MAPK. In addition, PE effectively promote the expression of tight junction proteins occludin and claudin-3 during inflammation, maintaining the integrity of the blood-milk barrier. In summary, our research shows that PE inhibits the phosphorylation of AKT/NF-κB and MAPK signals; It also relieves mastitis by repairing the blood-milk barrier.
Collapse
Affiliation(s)
- Xingchi Kan
- Department of Theoretic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Guiqiu Hu
- Department of Theoretic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Bingxu Huang
- Department of Theoretic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Wenjin Guo
- Department of Theoretic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Yaping Huang
- Department of Theoretic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Yingsheng Chen
- Department of Theoretic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Ping Xu
- Department of Theoretic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Xiangyu Cai
- Department of Theoretic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Shoupeng Fu
- Department of Theoretic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Juxiong Liu
- Department of Theoretic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| |
Collapse
|
22
|
Han M, Zhang M. The regulatory mechanism of amino acids on milk protein and fat synthesis in mammary epithelial cells: a mini review. Anim Biotechnol 2021; 34:402-412. [PMID: 34339350 DOI: 10.1080/10495398.2021.1950743] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Mammary epithelial cell (MEC) is the basic unit of the mammary gland that synthesizes milk components including milk protein and milk fat. MECs can sense to extracellular stimuli including nutrients such as amino acids though different sensors and signaling pathways. Here, we review recent advances in the regulatory mechanism of amino acids on milk protein and fat synthesis in MECs. We also highlight how these mechanisms reflect the amino acid requirements of MECs and discuss the current and future prospects for amino acid regulation in milk production.
Collapse
Affiliation(s)
- Meihong Han
- College of Animal Science, Yangtze University, Jingzhou, China
| | - Minghui Zhang
- College of Animal Science, Yangtze University, Jingzhou, China
| |
Collapse
|
23
|
Luo C, Peng W, Kang J, Chen C, Peng J, Wang Y, Tang Q, Xie H, Li Y, Pan X. Glutamine Regulates Cell Growth and Casein Synthesis through the CYTHs/ARFGAP1-Arf1-mTORC1 Pathway in Bovine Mammary Epithelial Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:6810-6819. [PMID: 34096300 DOI: 10.1021/acs.jafc.1c02223] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
In the dairy industry, glutamine (Gln) is often used as a feed additive to increase milk yield and quality; however, the molecular regulation underneath needs further clarification. Here, with bovine mammary epithelial cells (BMECs), the effects and mechanisms of Gln on cell growth and casein synthesis were assessed. When Gln was added or depleted from BMECs, both cell growth and β-casein (CSN2) expression were increased or decreased, respectively. Overexpressing or inhibiting the mechanistic target of rapamycin (mTOR) revealed that Gln regulated cell growth and CSN2 synthesis through the mTORC1 pathway. A similar intervention of ADP-ribosylation factor 1 (Arf1) uncovered that Gln activated the mTORC1 pathway through Arf1. We next observed that both guanine nucleotide exchange factors, Cytohesin-1/2/3 (CYTH1/2/3, CYTHs) and ADP-ribosylation factor GTPase activating protein 1 (ARFGAP1), interacted with Arf1. Inhibiting CYTHs or ARFGAP1 showed that Gln supplement or depletion activated or inactivated Arf1 through CYTHs or ARFGAP1, respectively. Collectively, this study demonstrated that Gln positively regulated cell growth and casein synthesis in BMECs, which works through the CYTHs/ARFGAP1-Arf1-mTORC1 pathway. These results greatly enhanced current understanding regarding the regulation of the mTOR pathway and provided new insights for the processes of cell growth and casein synthesis by amino acids, particularly Gln.
Collapse
|
24
|
Toledo MZ, Stangaferro ML, Gennari RS, Barletta RV, Perez MM, Wijma R, Sitko EM, Granados G, Masello M, Van Amburgh ME, Luchini D, Giordano JO, Shaver RD, Wiltbank MC. Effects of feeding rumen-protected methionine pre- and postpartum in multiparous Holstein cows: Lactation performance and plasma amino acid concentrations. J Dairy Sci 2021; 104:7583-7603. [PMID: 33865588 DOI: 10.3168/jds.2020-19021] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 02/18/2021] [Indexed: 12/16/2022]
Abstract
Objectives were to evaluate the effect of feeding rumen-protected methionine (RPM) in pre- and postpartum total mix ration (TMR) on lactation performance and plasma AA concentrations in dairy cows. A total of 470 multiparous Holstein cows [235 cows at University of Wisconsin (UW) and 235 cows at Cornell University (CU)] were enrolled approximately 4 wk before parturition, housed in close-up dry cow and replicated lactation pens. Pens were randomly assigned to treatment diets (pre- and postpartum, respectively): UW control (CON) diet = 2.30 and 2.09% of Met as percentage of metabolizable protein (MP) and RPM diet = 2.83 and 2.58% of Met as MP; CU CON = 2.22 and 2.19% of Met as percentage of MP, and CU RPM = 2.85 and 2.65% of Met as percentage of MP. Treatments were evaluated until 112 ± 3 d in milk (DIM). Milk yield was recorded daily. Milk samples were collected at wk 1 and 2 of lactation, and then every other week, and analyzed for milk composition. For lactation pens, dry matter intake (DMI) was recorded daily. Body weight and body condition score were determined from 4 ± 3 DIM and parturition until 39 ± 3 and 49 DIM, respectively. Plasma AA concentrations were evaluated within 3 h after feeding during the periparturient period [d -7 (±4), 0, 7 (±1), 14 (±1), and 21 (±1); n = 225]. In addition, plasma AA concentrations were evaluated (every 3 h for 24 h) after feeding in cows at 76 ± 8 DIM (n = 16) and within 3 h after feeding in cows at 80 ± 3 DIM (n = 72). The RPM treatment had no effect on DMI (27.9 vs. 28.0 kg/d) or milk yield (48.7 vs. 49.2 kg/d) for RPM and CON, respectively. Cows fed the RPM treatment had increased milk protein concentration (3.07 vs. 2.95%) and yield (1.48 vs. 1.43 kg/d), and milk fat concentration (3.87 vs. 3.77%), although milk fat yield did not differ. Plasma Met concentrations tended to be greater for cows fed RPM at 7 d before parturition (25.9 vs. 22.9 µM), did not differ at parturition (22.0 vs. 20.4 µM), and were increased on d 7 (31.0 vs. 21.2 µM) and remained greater with consistent concentrations until d 21 postpartum (d 14: 30.5 vs. 19.0 µM; d 21: 31.0 vs. 17.8 µM). However, feeding RPM decreased Leu, Val, Asn, and Ser (d 7, 14, and 21) and Tyr (d 14). At a later stage in lactation, plasma Met was increased for RPM cows (34.4 vs. 16.7 µM) consistently throughout the day, with no changes in other AA. Substantial variation was detected for plasma Met concentration (range: RPM = 8.9-63.3 µM; CON = 7.8-28.8 µM) among cows [coefficient of variation (CV) > 28%] and within cow during the day (CV: 10.5-27.1%). In conclusion, feeding RPM increased plasma Met concentration and improved lactation performance via increased milk protein production.
Collapse
Affiliation(s)
- Mateus Z Toledo
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison 53706
| | | | - Rodrigo S Gennari
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison 53706
| | - Rafael V Barletta
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison 53706
| | - Martin M Perez
- Department of Animal Science, Cornell University, Ithaca, NY 14853
| | - Robert Wijma
- Department of Animal Science, Cornell University, Ithaca, NY 14853
| | - Emily M Sitko
- Department of Animal Science, Cornell University, Ithaca, NY 14853
| | - German Granados
- Department of Animal Science, Cornell University, Ithaca, NY 14853
| | | | | | | | - Julio O Giordano
- Department of Animal Science, Cornell University, Ithaca, NY 14853
| | - Randy D Shaver
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison 53706
| | - Milo C Wiltbank
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison 53706.
| |
Collapse
|
25
|
Ao J, Ma Z, Li R, Zhang S, Gao X, Zhang M. Phospho-Tudor-SN coordinates with STAT5 to regulate prolactin-stimulated milk protein synthesis and proliferation of bovine mammary epithelial cells. Anim Biotechnol 2021; 33:1161-1169. [PMID: 33849380 DOI: 10.1080/10495398.2021.1879824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Tudor staphylococcal nuclease (Tudor-SN) participates in milk synthesis and cell proliferation in response to prolactin (PRL) and plays a regulatory role on mTOR phosphorylation. However, the complicated molecular mechanism of Tudor-SN regulating milk protein synthesis and cell proliferation still remains to be illustrated. In present study, we observed that the proteins level of phosphorylated Tudor-SN and phosphorylated STAT5 were simultaneously enhanced upon PRL treatment in bovine mammary epithelial cells (BMECs). Tudor-SN overexpression and knockdown experiment showed that Tudor-SN positively regulated the synthesis of milk protein, cell proliferation and the phosphorylation of STAT5, which was dependent on Tudor-SN phosphorylation. STAT5 knockdown experiment showed that Tudor-SN stimulated mTOR pathway through regulating STAT5 activation, which was required for PRL to activate the mTOR pathway. Thus, these results demonstrate the primary mechanism of Tudor-SN coordinating with STAT5 to regulate milk protein synthesis and cell proliferation under stimulation of PRL in BMECs, which may provide some new perspectives for increasing milk production.
Collapse
Affiliation(s)
- Jinxia Ao
- College of Life Science, Northeast Agricultural University, Heilongjiang, Harbin, China
| | - Zonghua Ma
- College of Life Science, Northeast Agricultural University, Heilongjiang, Harbin, China
| | - Rui Li
- College of Life Science, Northeast Agricultural University, Heilongjiang, Harbin, China
| | - Sen Zhang
- College of Life Science, Northeast Agricultural University, Heilongjiang, Harbin, China
| | - Xuejun Gao
- College of Animal Science, Yangtze University, Jingzhou, Hubei, China
| | - Minghui Zhang
- College of Life Science, Northeast Agricultural University, Heilongjiang, Harbin, China.,College of Animal Science, Yangtze University, Jingzhou, Hubei, China
| |
Collapse
|
26
|
Cardoso FF, Donkin SS, Pereira MN, Pereira RAN, Peconick AP, Santos JP, Silva RB, Caproni VR, Parys C, Danes MAC. Effect of protein level and methionine supplementation on dairy cows during the transition period. J Dairy Sci 2021; 104:5467-5478. [PMID: 33685687 DOI: 10.3168/jds.2020-19181] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 01/09/2021] [Indexed: 12/19/2022]
Abstract
Cows experience a significant negative protein balance during the first 30 d of lactation. Given the functional effects of AA on health, especially in challenging periods such as calving, higher levels of protein and specific AA in the diet may act to improve health and feed intake. The response of dairy cows to 3 protein supplementation strategies during the transition period and through the first 45 d in milk was evaluated. The final data set had 39 Holstein cows blocked based on parity (primiparous vs. multiparous) and expected calving and randomly assigned within each block to one of 3 dietary treatments: low protein (LP), high protein (HP), or high protein plus rumen-protected methionine (HPM). Treatments were offered from d -18 ± 5 to 45 d relative to parturition. Pre- and postpartum diets were formulated for high metabolizable protein (MP) supply from soybean meal, and HP and HPM provided higher MP balance than LP. Preplanned contrasts were LP versus HP+HPM and HP versus HPM. Significance was declared at P ≤ 0.05 and trends at 0.05 <P ≤ 0.10. Cows fed HP and HPM had greater fry matter intake (DMI) prepartum than LP (+2 kg/d), and there was a trend for greater DMI with HPM than with HP (+1.6 kg/d). Body weight and condition score before and after calving did not differ among treatments. High protein (HP and HPM) tended to increase milk yield during the first 45 d of lactation (+1.75 kg/d), increased milk lactose content and urea-N in milk and plasma, tended to increase blood BHB 14 d postpartum, and tended to reduce milk/DMI compared with LP. Blood concentrations of calcium at calving and of glucose, and nonesterified fatty acids pre- and postpartum did not differ. High protein induced lower concentration of plasma IL-1 at calving and lowered blood lymphocytes 21 d postpartum, suggestive of a reduced inflammatory status compared with LP. The concentrations of IL-10, tumor necrosis factor alpha, and other hemogram variables did not differ among treatments. Addition of rumen-protected methionine to the HP diet did not alter milk yield but increased fat and total solids concentrations. The rumen-protected methionine had no effect on blood metabolites and immunity markers, with the exception of increased pre-partum insulin concentrations. The data indicate that dairy cows around calving respond positively to an increase in the supply of MP and to rumen-protected methionine supplementation of the HP diet by increasing intake and improving immune status.
Collapse
Affiliation(s)
- Fabiana F Cardoso
- Departamento de Zootecnia, Universidade Federal de Lavras, Lavras, MG 37200-900, Brazil
| | - Shawn S Donkin
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907
| | - Marcos N Pereira
- Departamento de Zootecnia, Universidade Federal de Lavras, Lavras, MG 37200-900, Brazil.
| | - Renata A N Pereira
- Empresa de Pesquisa Agropecuária de Minas Gerais, Lavras, MG 37200-900, Brazil
| | - Ana Paula Peconick
- Departamento de Medicina Veterinária, Universidade Federal de Lavras, Lavras, MG 37200-900, Brazil
| | - Josiane P Santos
- Departamento de Medicina Veterinária, Universidade Federal de Lavras, Lavras, MG 37200-900, Brazil
| | - Rayana B Silva
- Better Nature Research Center, Ijaci, MG 37.218-000, Brazil
| | - Vitória R Caproni
- Departamento de Zootecnia, Universidade Federal de Lavras, Lavras, MG 37200-900, Brazil
| | - Claudia Parys
- Evonik Nutrition & Care GmbH, 63457 Hanau-Wolfgang, Germany
| | - Marina A C Danes
- Departamento de Zootecnia, Universidade Federal de Lavras, Lavras, MG 37200-900, Brazil.
| |
Collapse
|
27
|
Zhou C, Li P, Han M, Gao X. Daidzein stimulates fatty acid-induced fat deposition in C2C12 myoblast cells via the G protein-coupled receptor 30 pathway. Anim Biotechnol 2020; 33:851-863. [PMID: 33164657 DOI: 10.1080/10495398.2020.1842749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Fat deposition in skeletal muscle is an important aspect of improving meat quality. Isoflavones can promote animal anabolism, but whether and how they regulate muscle fat deposition remain largely unclear. In this study, we explored the role and corresponding molecular mechanism of one of the major isoflavones, daidzein, in fat deposition in C2C12 myoblast cells. In the absence of fatty acids (FAs), daidzein did not promote triglyceride synthesis and lipid droplet formation in cells but increased sterol regulatory element-binding protein 1c (SREBP-1c) expression and maturation. In the presence of FAs, daidzein enhanced FAs-induced fat deposition and the SREBP-1c signaling. Daidzein promoted FAs-induced nuclear factor κB1 (NFκB1) phosphorylation and activated the SREBP-1c signaling in a PI3K-dependent manner. G protein-coupled receptor 30 (GPR30) knockdown but not estrogen receptor α (ERα) knockdown blocked the stimulation of daidzein on the PI3K-NFκB1-SREBP-1c signaling pathway, while both knockdown did not affect the stimulation of FAs on this signaling. qRT-PCR and ChIP-qPCR further detected that daidzein stimulated NFκB1-targeted SREBP-1c transcription. Daidzein did not affect ERα expression in cells, but it stimulated GPR30 expression and cytoplasmic localization. These results reveal that daidzein promotes FAs-induced fat deposition through the GPR30 signaling in C2C12 myoblast cells.
Collapse
Affiliation(s)
- Chengjian Zhou
- College of Animal Science, Yangtze University, Jingzhou, China
| | - Ping Li
- College of Animal Science, Yangtze University, Jingzhou, China
| | - Meihong Han
- College of Animal Science, Yangtze University, Jingzhou, China
| | - Xuejun Gao
- College of Animal Science, Yangtze University, Jingzhou, China
| |
Collapse
|
28
|
NCOA5 is a master regulator of amino acid-induced mTOR activation and β-casein synthesis in bovine mammary epithelial cells. Biochem Biophys Res Commun 2020; 529:569-574. [DOI: 10.1016/j.bbrc.2020.05.193] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 05/25/2020] [Indexed: 12/14/2022]
|
29
|
Yu Y, Yuan X, Li P, Wang Y, Yu M, Gao X. Vaccarin promotes proliferation of and milk synthesis in bovine mammary epithelial cells through the Prl receptor-PI3K signaling pathway. Eur J Pharmacol 2020; 880:173190. [DOI: 10.1016/j.ejphar.2020.173190] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 05/07/2020] [Accepted: 05/10/2020] [Indexed: 12/23/2022]
|
30
|
Pszczolkowski VL, Halderson SJ, Meyer EJ, Lin A, Arriola Apelo SI. Pharmacologic inhibition of mTORC1 mimics dietary protein restriction in a mouse model of lactation. J Anim Sci Biotechnol 2020; 11:67. [PMID: 32612825 PMCID: PMC7322913 DOI: 10.1186/s40104-020-00470-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 05/11/2020] [Indexed: 12/20/2022] Open
Abstract
Background Understanding the mechanisms of N utilization for lactation can lead to improved requirement estimates and increased efficiency, which modern dairy diets currently fail to maximize. The mechanistic target of rapamycin complex 1 (mTORC1) is a central hub of translation regulation, processing extra- and intra-cellular signals of nutrient availability and physiological state, such as amino acids and energy. We hypothesized that dietary amino acids regulate lactation through mTORC1, such that inhibition of mTORC1 will lead to decreased lactation performance when amino acids are not limiting. Our objectives were to assess lactation performance in lactating mice undergoing dietary and pharmacologic interventions designed to alter mTORC1 activity. Methods First lactation mice (N = 18; n = 6/treatment) were fed an adequate protein diet (18% crude protein), or an isocaloric protein-restricted diet (9% crude protein) from the day after parturition until lactation day 13. A third group of mice was fed an adequate protein diet and treated with the mTORC1 inhibitor rapamycin (4 mg/kg every other day) intraperitoneally, with the first two groups treated with vehicle as control. Dams and pups were weighed daily, and feed intake was recorded every other day. Milk production was measured every other day beginning on lactation day 4 by the weigh-suckle-weigh method. Tissues were collected after fasting and refeeding. Results Milk production and pup weight were similarly decreased by both protein restriction and rapamycin treatment, with final production at 50% of control (P = 0.008) and final pup weight at 85% of control (P < 0.001). Mammary phosphorylation of mTORC1’s downstream targets were decreased by protein restriction and rapamycin treatment (P < 0.05), while very little effect was observed in the liver of rapamycin treated mice, and none by protein restriction. Conclusions Overall, sufficient supply of dietary amino acids was unable to maintain lactation performance status in mice with pharmacologically reduced mammary mTORC1 activity, as evidenced by diminished pup growth and milk production, supporting the concept that mTORC1 activation rather than substrate supply is the primary route by which amino acids regulate synthesis of milk components.
Collapse
Affiliation(s)
- Virginia L Pszczolkowski
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI USA.,Endocrinology and Reproductive Physiology Graduate Training Program, University of Wisconsin-Madison, Madison, WI USA
| | - Steven J Halderson
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI USA
| | - Emma J Meyer
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI USA
| | - Amy Lin
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI USA
| | - Sebastian I Arriola Apelo
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI USA.,Endocrinology and Reproductive Physiology Graduate Training Program, University of Wisconsin-Madison, Madison, WI USA
| |
Collapse
|
31
|
Zhang M, Ma Z, Li R, Guo S, Qiu Y, Gao X. Proteomic Analysis Reveals Proteins and Pathways Associated with Lactation in Bovine Mammary Epithelial Cell-Derived Exosomes. J Proteome Res 2020; 19:3211-3219. [DOI: 10.1021/acs.jproteome.0c00176] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Minghui Zhang
- College of Animal Science, Yangtze University, Nanhuan Road 1, Jingzhou, 434020 Hubei, China
| | - Zonghua Ma
- College of Life Science, Northeast Agricultural University, Changjiang Road 600, Xiangfang District, Harbin, 150030 Heilongjiang, China
| | - Rui Li
- College of Life Science, Northeast Agricultural University, Changjiang Road 600, Xiangfang District, Harbin, 150030 Heilongjiang, China
| | - Siqi Guo
- College of Life Science, Northeast Agricultural University, Changjiang Road 600, Xiangfang District, Harbin, 150030 Heilongjiang, China
| | - Youwen Qiu
- College of Life Science, Northeast Agricultural University, Changjiang Road 600, Xiangfang District, Harbin, 150030 Heilongjiang, China
| | - Xuejun Gao
- College of Animal Science, Yangtze University, Nanhuan Road 1, Jingzhou, 434020 Hubei, China
| |
Collapse
|
32
|
Yu M, Qi H, Gao X. Daidzein promotes milk synthesis and proliferation of mammary epithelial cells via the estrogen receptor α-dependent NFκB1 activation. Anim Biotechnol 2020; 33:43-52. [PMID: 32401613 DOI: 10.1080/10495398.2020.1763376] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Isoflavones possess a wide range of physiological effects. However, it is still unclear whether isoflavones can promote milk synthesis in mammary gland. This study aimed to determine the effects of a main soy isoflavone, daidzein, on milk synthesis and proliferation of mammary epithelial cells (MECs) and reveal the underlying molecular mechanism. Primary bovine MECs were treated with different concentrations of daidzein (0, 5, 10, 20, 40, and 80 μM). Daidzein dose-dependently promoted α- and β-casein and lipid synthesis, cell cycle transition, and cell amount, with the best stimulatory effect at 20 μM. Daidzein also stimulated mTOR activation and Cyclin D1 and SREBP-1c expression. Daidzein induced the expression and nuclear localization of estrogen receptor α (ERα), and ERα knockdown blocked the stimulation of daidzein on these above signaling pathways. ERα knockdown also abolished the stimulation of daidzein on NFκB1 expression and phosphorylation, and NFκB1 was required for daidzein to enhance the mTOR, Cyclin D1 and SREBP-1c signaling pathways. In summary, our findings reveal that daidzein stimulates milk synthesis and proliferation of MECs via the ERα-dependent NFκB1 activation.
Collapse
Affiliation(s)
- Mengmeng Yu
- College of Animal Science, Yangtze University, Jingzhou, Hubei, China
| | - Hao Qi
- College of Animal Science, Yangtze University, Jingzhou, Hubei, China
| | - Xuejun Gao
- College of Animal Science, Yangtze University, Jingzhou, Hubei, China
| |
Collapse
|
33
|
Potts SB, Scholte CM, Moyes KM, Erdman RA. Production responses to rumen-protected choline and methionine supplemented during the periparturient period differ for primi- and multiparous cows. J Dairy Sci 2020; 103:6070-6086. [PMID: 32359982 DOI: 10.3168/jds.2019-17591] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 02/14/2020] [Indexed: 12/19/2022]
Abstract
The objective of this experiment was to examine production performance responses to feeding rumen-protected choline (RPC) or methionine (RPM), or both, during the periparturient period. Fifty-four Holstein cows (25 primiparous, 29 multiparous) were used in a randomized block design experiment with a 2 × 2 factorial treatment structure. Cows were blocked by expected calving date and parity and assigned to 1 of 4 treatments: CON (no RPC or RPM); RPC (13.0 g/d of choline ion); RPM (9 g/d of dl-methionine prepartum; 13.5 g/d of dl-methionine postpartum); or RPC + RPM. Treatments were applied once daily as a top-dress from 3 wk before through 5 wk after calving. Dry matter intake and milk production were recorded daily, and milk samples were obtained once weekly. Data were analyzed for primi- and multiparous cows separately, using a repeated-measures mixed model that included random effects of cow and block and fixed effects of RPC, RPM, week, and their interactions; week served as the repeated effect. Initial BW and previous lactation milk yield were included as covariates in the statistical model for multiparous cows. Feeding RPC without RPM increased milk yield for multiparous cows by 8.7 kg/d, but this increase was not observed when RPC was fed with RPM. In multiparous cows, feeding RPM increased milk fat concentration and tended to increase milk fat yield. Because of this, RPM increased fat-corrected milk (FCM) by 2.8 kg/d at wk 2 postpartum, and this increase was sustained through wk 5 postpartum. In contrast, RPM did not affect overall milk fat yield and concentration for primiparous cows. Feeding RPC increased milk yield for primiparous cows by 3.5 kg/d irrespective of RPM inclusion, which is contrary to observations in multiparous cows, where RPC increased milk yield only in the absence of RPM. These results indicate that responses to RPC during the periparturient period may be dependent upon supply of methionine. Our observations also demonstrate that primi- and multiparous cows respond differently to RPC and RPM supplemented individually or simultaneously during the periparturient period. This variation in response could have been mediated by putative differences in choline and methionine requirements of primiparous versus multiparous cows, or by differences in the levels of milk production between the 2 groups (36 vs. 25 kg of FCM/d). However, cows in this study did not experience severe negative energy balance (mean nadirs of -6.6 and -5.0 Mcal/d for multiparous and primiparous cows, respectively), which likely affected their responses to RPC and RPM.
Collapse
Affiliation(s)
- S B Potts
- Department of Animal and Avian Sciences, University of Maryland, College Park 20742
| | - C M Scholte
- Department of Animal and Avian Sciences, University of Maryland, College Park 20742
| | - K M Moyes
- Department of Animal and Avian Sciences, University of Maryland, College Park 20742
| | - R A Erdman
- Department of Animal and Avian Sciences, University of Maryland, College Park 20742.
| |
Collapse
|
34
|
Zhu C, Wang L, Zhu J, Jiang Y, Du X, Duan Q, Yin H, Huang X, Song Y, Cao B, Li G, An X. OGR1 negatively regulates β-casein and triglyceride synthesis and cell proliferation via the PI3K/AKT/mTOR signaling pathway in goat mammary epithelial cells. Anim Biotechnol 2020; 32:627-636. [PMID: 32167419 DOI: 10.1080/10495398.2020.1737099] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Goat milk in some cases is less allergenic than cow milk, therefore, more people drink goat milk in the world, so it is necessary for us to improve the yield and quality of goat milk. Previous studies have shown that some genes are closely related to lactation. Ovarian cancer G protein-coupled 1 (OGR1) is a G protein-coupled receptor discovered recently. OGR1 is widely found in various tissues of organisms and is involved in cell skeleton reorganization, carcinogenesis, cell proliferation, and apoptosis by regulating multiple signaling pathways in cells. However, the modulating effect of OGR1 in lactation is still unknown. Therefore, the objective of this study is to investigate the function of OGR1 in goat mammary epithelial cells (GMECs). Flow cytometry, CCK8, EDU, enzyme-linked immunosorbent assay, and triglyceride test kit assays were performed and we found that OGR1 regulated Bcl-2/Bax ratio, Fas protein expression as well as the phosphorylation of AKT and mammalian target of rapamycin (mTOR). si-OGR1 could enhance the proliferation of GMECs by promoting G1/S phase progression and the synthesis of β-casein and triglyceride. By contrast, OGR1 repressed GMECs proliferation and down-regulated the synthesis of β-casein and triglyceride by blocking the PI3K/AKT/mTOR signaling pathway in GMECs.
Collapse
Affiliation(s)
- Chao Zhu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, P. R. China
| | - Lili Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, P. R. China
| | - Junru Zhu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, P. R. China
| | - Yue Jiang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, P. R. China
| | - Xiaoyan Du
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, P. R. China
| | - Quyu Duan
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, P. R. China
| | - Hao Yin
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, P. R. China
| | - Xinrui Huang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, P. R. China
| | - Yuxuan Song
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, P. R. China
| | - Binyun Cao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, P. R. China
| | - Guang Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, P. R. China
| | - Xiaopeng An
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, P. R. China
| |
Collapse
|
35
|
Luo C, Yu M, Li S, Huang X, Qi H, Gao X. Methionine stimulates GlyRS phosphorylation via the GPR87-CDC42/Rac1-MAP3K10 signaling pathway. Biochem Biophys Res Commun 2020; 523:847-852. [PMID: 31954518 DOI: 10.1016/j.bbrc.2019.12.124] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 12/24/2019] [Indexed: 02/08/2023]
|