Hostname: page-component-848d4c4894-hfldf Total loading time: 0 Render date: 2024-05-30T04:48:07.464Z Has data issue: false hasContentIssue false

Comparative transcriptomic analysis of mammary gland tissues reveals the critical role of GPR110 in palmitic acid-stimulated milk protein and fat synthesis

Published online by Cambridge University Press:  22 March 2023

Minghui Zhang
Affiliation:
College of Animal Science, Yangtze University, Jingmi Road 88, Jingzhou 434025, People’s Republic of China
Zonghua Ma
Affiliation:
College of Animal Science, Yangtze University, Jingmi Road 88, Jingzhou 434025, People’s Republic of China
Hao Qi
Affiliation:
College of Animal Science, Yangtze University, Jingmi Road 88, Jingzhou 434025, People’s Republic of China
Xu Cui
Affiliation:
College of Animal Science, Yangtze University, Jingmi Road 88, Jingzhou 434025, People’s Republic of China
Rui Li
Affiliation:
College of Animal Science, Yangtze University, Jingmi Road 88, Jingzhou 434025, People’s Republic of China
Xuejun Gao*
Affiliation:
College of Animal Science, Yangtze University, Jingmi Road 88, Jingzhou 434025, People’s Republic of China
*
*Corresponding author: Xuejun Gao, Email: gaoxj53901@163.com

Abstract

The G protein-coupled receptors (GPCR) sensing nutritional signals (amino acids, fatty acids, glucose, etc.) are not fully understood. In this research, we used transcriptome sequencing to analyse differentially expressed genes (DEG) in mouse mammary gland tissues at puberty, lactation and involution stages, in which eight GPCR were selected out and verified by qRT-PCR assay. It was further identified the role of GPR110-mediating nutrients including palmitic acid (PA) and methionine (Met) to improve milk synthesis using mouse mammary epithelial cell line HC11. PA but not Met affected GPR110 expression in a dose-dependent manner. GPR110 knockdown decreased milk protein and fat synthesis and cell proliferation and blocked the stimulation of PA on mechanistic target of rapamycin (mTOR) phosphorylation and sterol-regulatory element binding protein 1c (SREBP-1c) expression. In summary, these experimental results disclose DEG related to lactation and reveal that GPR110 mediates PA to activate the mTOR and SREBP-1c pathways to promote milk protein and fat synthesis.

Type
Research Article
Copyright
© The Author(s), 2023. Published by Cambridge University Press on behalf of The Nutrition Society

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

Footnotes

These authors contributed equally to this work

References

Slepicka, PF, Somasundara, AVH & Dos Santos, CO (2021) The molecular basis of mammary gland development and epithelial differentiation. Semin Cell Dev Biol 114, 93112.10.1016/j.semcdb.2020.09.014CrossRefGoogle ScholarPubMed
Capuco, AV & Choudhary, RK (2020) Symposium review: determinants of milk production: understanding population dynamics in the bovine mammary epithelium. J Dairy Sci 103, 29282940.10.3168/jds.2019-17241CrossRefGoogle ScholarPubMed
Eberlé, D, Hegarty, B, Bossard, P, et al. (2004) SREBP transcription factors: master regulators of lipid homeostasis. Biochimie 86, 839848.10.1016/j.biochi.2004.09.018CrossRefGoogle ScholarPubMed
Osorio, JS, Lohakare, J & Bionaz, M (2016) Biosynthesis of milk fat, protein, and lactose: roles of transcriptional and posttranscriptional regulation. Physiol Genomics 48, 231256.10.1152/physiolgenomics.00016.2015CrossRefGoogle ScholarPubMed
Zheng, L, Zhang, W, Zhou, Y, et al. (2016) Recent advances in understanding amino acid sensing mechanisms that regulate mTORC1. Int J Mol Sci 17, 1636.10.3390/ijms17101636CrossRefGoogle ScholarPubMed
Nelson, G, Chandrashekar, J, Hoon, MA, et al. (2002) An amino-acid taste receptor. Nature 416, 199202.10.1038/nature726CrossRefGoogle Scholar
Pi, M, Nishimoto, SK & Quarles, LD (2017) GPRC6A: jack of all metabolism (or master of none). Mol Metab 6, 185193.10.1016/j.molmet.2016.12.006CrossRefGoogle ScholarPubMed
Li, X, Li, P, Wang, L, et al. (2019) Lysine enhances the stimulation of fatty acids on milk fat synthesis via the GPRC6A-PI3K-FABP5 signaling in bovine mammary epithelial cells. J Agric Food Chem 67, 70057015.10.1021/acs.jafc.9b02160CrossRefGoogle ScholarPubMed
Díaz-Soto, G, Rocher, A, García-Rodríguez, C, et al. (2016) The calcium-sensing receptor in health and disease. Int Rev Cell Mol Biol 327, 321369.10.1016/bs.ircmb.2016.05.004CrossRefGoogle ScholarPubMed
Luo, C, Yu, M, Li, S, et al. (2020) Methionine stimulates GlyRS phosphorylation via the GPR87-CDC42/Rac1-MAP3K10 signaling pathway. Biochem Biophys Res Commun 523, 847852.10.1016/j.bbrc.2019.12.124CrossRefGoogle ScholarPubMed
Yu, M, Wang, Y, Wang, Z, et al. (2019) Taurine promotes milk synthesis via the GPR87-PI3K-SETD1A signaling in BMECs. J Agric Food Chem 67, 19271936.10.1021/acs.jafc.8b06532CrossRefGoogle ScholarPubMed
Kimura, I, Ichimura, A, Ohue-Kitano, R, et al. (2020) Free fatty acid receptors in health and disease. Physiol Rev 100, 171210.10.1152/physrev.00041.2018CrossRefGoogle ScholarPubMed
Itoh, Y, Kawamata, Y, Harada, M, et al. (2003) Free fatty acids regulate insulin secretion from pancreatic β cells through GPR40. Nature 422, 173176.10.1038/nature01478CrossRefGoogle ScholarPubMed
Hirasawa, A, Tsumaya, K, Awaji, T, et al. (2005) Free fatty acids regulate gut incretin glucagon-like peptide-1 secretion through GPR120. Nat Med 11, 9094.10.1038/nm1168CrossRefGoogle ScholarPubMed
Luscombe, VB, Lucy, D, Bataille, CJR, et al. (2020) 20 Years an Orphan: is GPR84 a plausible medium-chain fatty acid-sensing receptor? DNA Cell Biol 39, 19261937.10.1089/dna.2020.5846CrossRefGoogle ScholarPubMed
Qi, H, Meng, C, Jin, X, et al. (2018) Methionine promotes milk protein and fat synthesis and cell proliferation via the SNAT2-PI3K signaling pathway in bovine mammary epithelial cells. J Agric Food Chem 66, 1102711033.10.1021/acs.jafc.8b04241CrossRefGoogle ScholarPubMed
Carmon, KS, Gong, X, Lin, Q, et al. (2011) R-spondins function as ligands of the orphan receptors LGR4 and LGR5 to regulate Wnt/β-catenin signaling. Proc Natl Acad Sci USA 108, 1145211457.10.1073/pnas.1106083108CrossRefGoogle ScholarPubMed
Yue, Z, Yuan, Z, Zeng, L, et al. (2018) LGR4 modulates breast cancer initiation, metastasis, and cancer stem cells. FASEB J 32, 24222437.10.1096/fj.201700897RCrossRefGoogle ScholarPubMed
Quon, T, Lin, LC, Ganguly, A, et al. (2020) Therapeutic opportunities and challenges in targeting the orphan G protein-coupled receptor GPR35. ACS Pharmacol Transl Sci 3, 801812.10.1021/acsptsci.0c00079CrossRefGoogle ScholarPubMed
Kaya, B, Melhem, H & Niess, JH (2021) GPR35 in intestinal diseases: from risk gene to function. Front Immunol 12, 717392.10.3389/fimmu.2021.717392CrossRefGoogle ScholarPubMed
Kindt, KS, Akturk, A, Jarysta, A, et al. (2021) EMX2-GPR156-Gαi reverses hair cell orientation in mechanosensory epithelia. Nat Commun 12, 2861.10.1038/s41467-021-22997-1CrossRefGoogle ScholarPubMed
Zarei Ghobadi, M, Emamzadeh, R, Teymoori-Rad, M, et al. (2021) Decoding pathogenesis factors involved in the progression of ATLL or HAM/TSP after infection by HTLV-1 through a systems virology study. Virol J 18, 175.10.1186/s12985-021-01643-8CrossRefGoogle ScholarPubMed
Gong, X, Carmon, KS, Lin, Q, et al. (2012) LGR6 is a high affinity receptor of R-spondins and potentially functions as a tumor suppressor. PLoS One 7, e37137.10.1371/journal.pone.0037137CrossRefGoogle ScholarPubMed
Chiang, N, Libreros, S, Norris, PC, et al. (2019) Maresin 1 activates LGR6 receptor promoting phagocyte immunoresolvent functions. J Clin Invest 129, 52945311.10.1172/JCI129448CrossRefGoogle ScholarPubMed
Kong, Y, Ou, X, Li, X, et al. (2020) LGR6 promotes tumor proliferation and metastasis through Wnt/β-Catenin signaling in triple-negative breast cancer. Mol Ther Oncolytics 18, 351359.10.1016/j.omto.2020.06.020CrossRefGoogle ScholarPubMed
Liu, SL, Zhou, YM, Tang, DB, et al. (2019) LGR6 promotes osteogenesis by activating the Wnt/β-catenin signaling pathway. Biochem Biophys Res Commun 519, 17.10.1016/j.bbrc.2019.08.122CrossRefGoogle ScholarPubMed
Hamann, J, Hsiao, CC, Lee, CS, et al. (2016) Adhesion GPCRs as modulators of immune cell function. Handb Exp Pharmacol 234, 329350.10.1007/978-3-319-41523-9_15CrossRefGoogle ScholarPubMed
Röthe, J, Thor, D, Winkler, J, et al. (2019) Involvement of the adhesion GPCRs latrophilins in the regulation of insulin release. Cell Rep 26, 15731584.e5.10.1016/j.celrep.2019.01.040CrossRefGoogle Scholar
Kakarala, KK & Jamil, K (2014) Sequence-structure based phylogeny of GPCR Class A Rhodopsin receptors. Mol Phylogenet Evol 74, 6696.10.1016/j.ympev.2014.01.022CrossRefGoogle Scholar
Zaccagnino, P, Saltarella, M, D’Oria, S, et al. (2009) N-arachidonylglycine causes ROS production and cytochrome c release in liver mitochondria. Free Radic Biol Med 47, 585592.10.1016/j.freeradbiomed.2009.05.038CrossRefGoogle ScholarPubMed
Zhao, L, Hart, S, Cheng, J, et al. (2004) Mammary gland remodeling depends on gp130 signaling through Stat3 and MAPK. J Biol Chem 279, 4409344100.10.1074/jbc.M313131200CrossRefGoogle ScholarPubMed
Lee, JW, Huang, BX, Kwon, H, et al. (2016) Orphan GPR110 (ADGRF1) targeted by N-docosahexaenoylethanolamine in development of neurons and cognitive function. Nat Commun 7, 13123.10.1038/ncomms13123CrossRefGoogle ScholarPubMed
Huang, BX, Hu, X, Kwon, HS, et al. (2020) Synaptamide activates the adhesion GPCR GPR110 (ADGRF1) through GAIN domain binding. Commun Biol 3, 109.10.1038/s42003-020-0831-6CrossRefGoogle ScholarPubMed
Lum, AM, Wang, BB, Beck-Engeser, GB, et al. (2010) Orphan receptor GPR110, an oncogene overexpressed in lung and prostate cancer. BMC Cancer 10, 40.10.1186/1471-2407-10-40CrossRefGoogle ScholarPubMed
Nam, HJ, Kim, YJ, Kang, JH, et al. (2022) GPR110 promotes progression and metastasis of triple-negative breast cancer. Cell Death Discov 8, 271.10.1038/s41420-022-01053-xCrossRefGoogle ScholarPubMed
Kwon, H, Kevala, K, Xin, H, et al. (2021) Ligand-induced GPR110 activation facilitates axon growth after injury. Int J Mol Sci 22, 3386.10.3390/ijms22073386CrossRefGoogle ScholarPubMed
Offermanns, S (2014) Free fatty acid (FFA) and hydroxy carboxylic acid (HCA) receptors. Annu Rev Pharmacol Toxicol 54, 407434.10.1146/annurev-pharmtox-011613-135945CrossRefGoogle ScholarPubMed
Triki, M, Rinaldi, G, Planque, M, et al. (2020) mTOR signaling and SREBP activity increase FADS2 expression and can activate sapienate biosynthesis. Cell Rep 31, 107806.10.1016/j.celrep.2020.107806CrossRefGoogle ScholarPubMed
Bai, D, Wu, Y, Deol, P, et al. (2021) Palmitic acid negatively regulates tumor suppressor PTEN through T366 phosphorylation and protein degradation. Cancer Lett 496, 127133.10.1016/j.canlet.2020.10.007CrossRefGoogle ScholarPubMed
Qiu, Y, Qu, B, Zhen, Z, et al. (2019) Leucine promotes milk synthesis in bovine mammary epithelial cells via the PI3K-DDX59 signaling. J Agric Food Chem 67, 88848895.10.1021/acs.jafc.9b03574CrossRefGoogle ScholarPubMed
Qi, H, Wang, L, Zhang, M, et al. (2022) Methionine and leucine induce ARID1A degradation to promote mTOR expression and milk synthesis in mammary epithelial cells. J Nutr Biochem 101, 108924.10.1016/j.jnutbio.2021.108924CrossRefGoogle ScholarPubMed
Lin, G, Qi, H, Guo, X, et al. (2023) ARID1B blocks methionine-stimulated mTOR activation to inhibit milk fat and protein synthesis in and proliferation of mouse mammary epithelial cells. J Nutr Biochem 114, 109274.10.1016/j.jnutbio.2023.109274CrossRefGoogle ScholarPubMed
Sztefko, K & Panek, J (2001) Serum free fatty acid concentration in patients with acute pancreatitis. Pancreatology 1, 230236.10.1159/000055816CrossRefGoogle ScholarPubMed
Moran, A, Basu, R, Milla, C, et al. (2004) Insulin regulation of free fatty acid kinetics in adult cystic fibrosis patients with impaired glucose tolerance. Metabolism 53, 14671472.10.1016/j.metabol.2004.06.015CrossRefGoogle ScholarPubMed
Yonezawa, T, Kurata, R, Yoshida, K, et al. (2013) Free fatty acids-sensing G protein-coupled receptors in drug targeting and therapeutics. Curr Med Chem 20, 38553871.10.2174/09298673113209990168CrossRefGoogle ScholarPubMed
Guo, X, Qi, H, Lin, G, et al. (2023) ARID3A plays a key regulatory role in palmitic acid-stimulated milk fat synthesis in mouse mammary epithelial cells. Cell Biol Int (Epublication ahead of print version 14 February 2023).10.1002/cbin.12003CrossRefGoogle Scholar
Jin, X, Zhen, Z, Wang, Z, et al. (2023) GPRC6A is a key mediator of palmitic acid regulation of lipid synthesis in bovine mammary epithelial cells. Cell Biol Int 46, 17471758.10.1002/cbin.11886CrossRefGoogle Scholar