Next Article in Journal
A Morphological and Ultrastructural Study of the Anterior Digestive Tract of Adult Nile Tilapia Oreochromis niloticus
Previous Article in Journal
Right Pulmonary Artery Distensibility Index in Heartworm Infected Dogs: Are the Different Methods Leading to Same Results?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Enhancing Milk Production by Nutrient Supplements: Strategies and Regulatory Pathways

1
Laboratory of Nutrition and Functional Food, College of Food Science and Engineering, Jilin University, Changchun 130062, China
2
Key Laboratory of Healthy Freshwater Aquaculture, Ministry of Agriculture and Rural Affairs, Huzhou 313001, China
3
Key Laboratory of Fish Health and Nutrition of Zhejiang Province, Zhejiang Institute of Freshwater Fisheries, Huzhou 313001, China
4
Key Laboratory of Genetics and Breeding, Zhejiang Institute of Freshwater Fisheries, Huzhou 313001, China
5
Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun 130062, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Animals 2023, 13(3), 419; https://doi.org/10.3390/ani13030419
Submission received: 24 October 2022 / Revised: 10 January 2023 / Accepted: 23 January 2023 / Published: 26 January 2023
(This article belongs to the Section Cattle)

Abstract

:

Simple Summary

The demand for the milk and dairy products keeps growing due to the growing population, the rising consumption of protein-enriched food, and other factors. Thus, enhancing milk production becomes a critical issue for the world. Of all the factors related to promoting milk production, nutrient supplementation is widely investigated. First, researchers provided mammary epithelial cells or animals with appropriate nutrients such as amino acids, peptides, lipids, carbohydrates, and others. Then, they detected the increase in milk protein expression or milk production. Finally, the researcheres further investigated the essential pathways on which the nutrients act. As much effort has been made on this topic, we summarize recent work on enhancing milk production through nutrient supplements. We hope this review provides a systematic reference for researchers in related fields and can help understand the mechanism of milk production promotion by nutrients.

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.

1. Introduction

Dairy production is an essential economic characteristic of dairy animals. Milk and dairy products are consumed all over the world. In 2021, the world’s milk production reached nearly 928 million tons [1], and the demand will continue to increase [2]. It is predicted that world milk production will grow at an annual rate of 1.7%. Milk production will reach 1.020 billion tons by 2030, faster than most other major agricultural products [3]. However, the average milk production per cow per lactation period varies from 1.3 to 10 tons. For example, the average milk production of dairy cows in Saudi Arabia and Israel is more than 10 tons, while in India, it is only 1.3 tons [4]. Though genetics greatly influence milk production, nutrient supplements still play an essential role.
In recent years, we have taken traditional and molecular breeding efforts, especially in breed registration, productivity determination, body size identification, artificial insemination, genome-wide association analysis (GWAS) and somatic cell cloning, achieving significant increases in milk production in dairy cows [5,6,7,8,9,10]. For example, in developing countries such as India, a Holstein cow’s average daily milk production increased from 1259 kg in 2009–2010 to 1700 kg in 2018–2019 [11]. However, it is becoming increasingly difficult to further improve milk quality and yield traits through conventional and molecular breeding methods.
A promising and upcoming strategy for improving milk production and quality in developing countries is providing high-quality feed and balanced nutrition [12]. The strategy involves adding appropriate nutrients (amino acids, peptides, lipids) to the cell culture medium or feed that promotes milk quality and production. This work summarizes recent studies on nutrients enhancing milk production and relevant pathways to mammal milk production.

2. The Enhancement of Milk Production by Amino Acids and Peptides

2.1. Amino Acid

The most fundamental pathway for amino acids to enhance milk synthesis is the mTOR pathway (Table 1). The mammalian target of rapamycin (mTOR) is an essential signaling factor widely distributed in mammalian cells. It is essential in cell growth and protein synthesis [13]. Phosphatidylinositol-3 Kinase (PI3K) exists in the cytoplasm and has dual protein kinase and phospholipid kinase activities. PI3K is eventually converted to phosphatidylinositol 3,4,5-triphosphate (PIP3) after activation [14,15]. The serine/threonine kinase AKT, also known as protein kinase B (PKB), is a key downstream protein of PI3K. It exists in the cytoplasm, and they play a vital role in regulating cell growth, proliferation, survival, and metabolism [16,17]. The PI3K-Akt pathway is upstream of the mTOR signaling pathway, so factors that stimulate this pathway can also activate the mTOR pathway (Figure 1). PIP3 can bind to the PH domain of AKT proteins as a second messenger when PI3K is activated. The activated AKT is transferred from the cell membrane to the cytoplasm or nucleus and then continues to target and regulate downstream signaling molecules-mTOR [18]. Therefore, it further regulates downstream proteins after mTOR signaling is activated through the PI3K/AKT pathway. The phosphorylated mTOR can activate ribosomal protein S6 kinase 1 (S6K1) and eukaryotic translation initiation factor 4e-binding protein 1 (4EBP1), promoting their involvement in translation and protein synthesis [19].
It has been reported that amino acid addition activates the mTORC1 pathway via the Septin6 factor in dairy bovine mammary epithelial cells (BMECs), promoting cell growth and milk protein synthesis [20]. Amino acid supplementation in BMECs can also stimulate mTOR signaling through Seryl-tRNA synthetase (SARS) and Glycyl-tRNA synthetase (GlyRS), thereby exerting positive effects on cell proliferation and casein synthesis [21,22]. In addition, the optimal ratio of essential amino acids added to BMECs, and bovine mammary tissue explants (MTE) can stimulate β-casein synthesis [23]. Among all the amino acids, researchers focus on several amino acids, including methionine, leucine, valine and lysine (Figure 1). We review these works in the following part.

2.1.1. Methionine

Among all the amino acids, methionine attracts the most attention (Table 1). The methionine supply can increase milk protein synthesis by increasing the expression of related proteins in early lactation [24,25]. In addition, methionine can promote milk fat synthesis by promoting the mTORC1 signaling pathway. Amino-acid transporter 2 (ASCT2) is involved in SARS-mediated β-casein synthesis in BMECs via the mTOR signalling pathway [26]. Methionine positively regulates milk protein and fat synthesis in BMECs through the sodium-coupled neutral amino acid transporter 2 (SNAT2)-PI3K signaling pathway [27]. The PI3K-Akt-mTOR signaling pathway can regulate lipid synthesis by regulating the expression of the lipid synthesis-related factor sterol response element-binding protein (SREBP1) and its related esterases [28]. SREBP-1c acts as a transcription factor that controls the expression of lipid synthesis genes. Therefore, some factors activating SREBP-1c can also stimulate milk fat synthesis. For example, fatty acid binding protein 5 (FABP5), an intracellular lipid transporter, has been confirmed to be a crucial regulator of methionine- and estrogen-induced SREBP-1c gene expression in BMECs [29]. Besides, nuclear receptor co-activator 5 (NCOA5) and glucose-regulated protein 78 (GRP78) can also act as regulators of methionine-stimulated mTOR expression and activation [30,31]. They both function upstream of mTOR.
Supplementing 2.5 g/d of rumen-protected methionine per lactating Saanen goat led to significantly increased milk production [32]. The addition of methionine at 42 g/d per Holstein cow positively affected milk yield [33]. Studies have shown that adding microencapsulated DL-Met (0, 11.0, 19.3 and 27.5 g/d) to diets deficient in metabolizable methionine (10 g/d) can also increase milk production. It was confirmed that the increase rate of milk protein yield was the highest with the addition of 19.3 g/d methionine [34]. Holstein cows fed rumen-protected methionine increased milk production (from 1.43 kg/d to 1.48 kg/d) [35]. Studies have shown that milk production increases when Holstein cows are fed on methionine 10 g/day in their corn grain and soybean meal concentrate diets [36].

2.1.2. Leucine

Leucine affects the expression of mTORC1 through system L and system A transporters such as L-type amino acid transporter 1 (LAT1) and SNAT2 [37]. It has been confirmed that mTOR is a target molecule of leucine. Leucine promotes mTORC1 expression in cells through the Sestrin2/GATOR2/GATOR1 pathway [38]. Meanwhile, leucine can promote cellular processes through the PI3K/AKT/mTOR pathway as a signaling molecule [39]. It has been shown that leucine activates PI3K, thereby upregulating DEAD-Box Helicase 59 (DDX59). DDX59 positively regulates the mTOR and SREBP-1c signaling pathways leading to milk synthesis in primary BMECs [40]. Guanine nucleotide-binding protein subunit gamma-12 (GNG12) also mediates leucine regulation of mTORC1 signaling. It activates the mTORC1 pathway by interacting with the regulator, which can promote cow mammary epithelial cells (CMECs) growth and casein synthesis [41]. The addition of leucine to primary BMECs from mid-lactation Holstein cows promotes milk protein synthesis by affecting mTOR upstream factors NCOA5 and GRP78 [30,31].
Leucine can promote milk protein synthesis by the JAK2 (Janus kinase 2) and STAT5 (signal transducers and activators of transcription 5) pathways [42]. During gland development, JAK-STAT is essential for the survival of mammary gland secretory epithelial cells. The JAK-STAT pathway consists of three parts: tyrosine kinase-related receptors that receive signals, JAK that transmit signals, and STAT that produce effects [43]. When cytokines specifically bind to the corresponding receptors, the receptor molecules can be induced to dimerize. JAK kinases can interact closely with receptors and activate themselves. Following activation of the kinase, the receptor’s tyrosine residues can be stimulated and phosphorylated. After kinase phosphorylation, it can be recognized and bound by STAT. As a result, STAT tyrosine residues are phosphorylated to form dimers, enter the nucleus, bind to the target gene’s DNA regulatory region, regulate the target gene’s transcription, and produce corresponding biological effects [44]. Studies have confirmed that acetate, leucine and their interactions affect milk protein synthesis through JACK2/STAT5, mTOR and AMPK pathways in BMECs [42].

2.1.3. Valine

Valine can regulate milk protein synthesis [45]. Studies have confirmed that valine promotes milk protein synthesis in porcine mammary epithelial cells, and the regulatory pathways are mTOR, Ras/ERK and AKT [46,47]. The extracellular signal-regulated kinase (ERK) is a member of mitogen-activated protein kinase (MAPK) family. In the ERK-MAPK pathway, Ras GTPase is an upstream activator in the cascade response of MAPK (Ras-Raf-MEK-ERK). The Ras and ERK-MAPK pathways can be activated by growth factors, polypeptide hormones, neurotransmitters, etc [48]. Studies have shown that treatment with L-valine (L-enantiomer of valine) to porcine mammary epithelial cells can promote the expression of mTOR, Ras and ERK1/2 [46], thus playing a pivotal role in milk protein synthesis. The milk yield of each prolific late lactating Holstein cow fed of valine 40 g/d could reach 25.2 kg/d, which was higher than that of the control group (22.0 kg/d) [49]. It has also been demonstrated in lactating sows that adding 3.07% branched-chain amino acids to the diet can increase milk production by 21% [50].

2.1.4. Lysine

Lysine is the first limiting amino acid for milk protein synthesis in dairy cows. In the process of lysine-stimulated milk protein, GPRC6A-PI3K-FABP5 regulatory axis plays an important role [51]. GPRC6A (G protein-coupled receptor family C group 6 member A), a widely expressed G-protein coupled receptor, is a significant regulator of energy metabolism, cell proliferation and differentiation. Lysine also regulates milk protein synthesis through the mTOR and JAK2-STAT5 pathways. It has been shown that 1.0 mmol/L lysine stimulation significantly increases the expression of amino acid transporter B0,+ (ATB0,+), STAT5, and mTOR in BMECs [52]. Lysine promotes β-casein synthesis in BMECs via the SLC6A14-ERK-CDK1-mTOR signaling pathway [53]. Lysine also promotes mammary gland development in mice at puberty through the PI3K/AKT/mTOR signaling pathway [54]. The optimal ratio of supplemented Lys/Met is 3:1. This ratio can also regulate casein synthesis in BMECs, mainly through JAK2/ELF5, mTOR and its downstream ribosomal protein S6 kinase B1 (RPS6KB1) and eukaryotic translation initiation factor 4E binding protein 1 (EIF4EBP1) signaling pathways [55].

2.1.5. Other Amino Acids

Other amino acids can also have a positive effect on milk production (Table 1). Arginine supply affects mTORC1 expression and AMPK pathway, thereby affecting α-s1-casein synthesis in BMECs [56]. Histidine and threonine can promote milk protein synthesis through the mTOR pathway [57,58,59]. Taurine, 2-aminoethanesulfonic acid, can regulate insulin secretion, and the pathways that have been confirmed are PI3K/AKT, AKT/FOXO1, JAK2/STAT3 and mTOR/AMPK [60,61]. Taurine also plays an essential role in milk protein synthesis. Studies have shown that taurine promotes milk synthesis through the GPR87-PI3K-SETD1A signaling pathway [62]. GPCRs can sense amino acid signals to activate downstream pathways [63,64]. Therefore, taurine stimulates the expression of GPR87 to activate PI3K, activating the downstream signaling SET domain containing 1A (SETD1A) of the pathway.
Each lactating Holstein cow was fed 10 mg/kg 5-aminolevulinic acid for 14 days, and milk protein content increased [65]. Studies have shown that the continuous addition N-carbamoylglutamate (NCG) of 20 g/d per cow to the feed from 4 weeks before calving to 10 weeks after calving improves the lactation performance of postpartum cows [56]. Previous studies have demonstrated that feeding lactating sows (Large White) with 1% glutamine for 14 days has a positive effect on milk production (+ 3.34 kg) [66]. Supplementation with 1.2 g/day of rumen-protective gamma-aminobutyric acid increased feed intake and milk protein production in Holstein cows [67]. The addition of three rumen-protected methionines (Met, Lys, and His) have the potential to increase milk production [68].
We can conclude that adding amino acids to the feed has a specific positive effect on milk yield. In recent years, researchers focused on exploring the functional genes and key pathways related to the promotion of milk production at the cellular level. Amino acids can positively affect the proliferation and milk protein synthesis of mammary epithelial cells. After amino acids are transported into cells, they stimulate the expression of certain genes involved in milk protein synthesis. The mTOR is involved in multiple biological processes (Figure 1). Septin6, SARS, GlyRS, DDX59, and GNG12 are the upstream genes of mTOR pathway. After being stimulated by amino acids, this pathway will be activated, thereby promoting milk protein synthesis. The PI3K/AKT is the upstream pathway of mTOR, so certain amino acids exert their effects by stimulating upstream factors of PI3K, such as NCOA5. In addition, leucine and lysine can also affect the JAK2/STAT5 pathway and thus affect milk protein synthesis.
Table 1. Effects of amino acids on milk production and its potential regulatory pathways.
Table 1. Effects of amino acids on milk production and its potential regulatory pathways.
ItemsTreatmentsFunctionsPotential Signaling PathwaysReferences
Amino acid (+)Dairy cow mammary epithelial cellsCell growth ↑
Casein synthesis ↑
Septin6-mTOR signaling pathway[20]
Essential amino acids (+)Bovine mammary epithelial cellsCell proliferation ↑
β-casein production ↑
SARS- mTOR signaling pathway[22]
Amino acid (+)Bovine mammary epithelial cellsMilk protein ↑
Fat synthesis ↑
GlyRS-mTOR signaling pathway[21]
Essential amino acids (+)Bovine mammary epithelial cell line, mammary tissue explantsβ-casein expression ↑mTOR signaling[23]
Methionine (+)Cow mammary gland tissueMilk protein synthesis ↑AKT phosphorylation[24]
Methionine (+)Bovine mammary epithelial cells (0.6 mmol/L)Milk protein ↑
Fat synthesis ↑
Cell proliferation ↑
SNAT2-PI3K signaling pathway[27]
Methionine (+)Bovine mammary epithelial cells (0.6 mmol/L)Cell growth ↑
β-casein synthesis ↑
ASCT2 expression ↑
ASCT2/SARS/mTOR signaling pathway[28]
Methionine (+)Bovine mammary epithelial cellsLipid droplet formation ↑
β-casein ↑
FABP5-SREBP-1c signaling pathway[29]
Methionine (+)Lactating Saanen goatsMilk production ↑
Milk protein ↑
——[32]
Methionine (+)Holstein cowsMilk yield ↑——[33]
Methionine (+)Holstein cowsMilk production ↑
Milk protein ↑
——[34]
Methionine (+)Holstein cowsMilk protein production ↑——[35]
Methionine (+)Holstein cowsMilk production ↑——[36]
Leucine (+)Bovine mammary epithelial cells (0.75 mmol/L)Milk protein ↑
Milk fat synthesis ↑
PI3K-DDX59 signaling pathway[40]
Leucine (+)Cow mammary epithelial cellsCell growth ↑
Casein synthesis ↑
GNG12-mTORC1 signaling pathway[41]
Leucine and methionine (+)Bovine mammary epithelial cellsMilk Fat ↑mTOR-CRTC2-SREBP-1c signaling pathway[69]
Leucine and methionine (+)Bovine mammary epithelial cellsmTOR phosphorylation ↑
β-casein synthesis ↑
NCOA5- PI3K-mTOR signaling pathway[31]
Leucine and methionine (+)Bovine mammary epithelial cellsMilk synthesis ↑
Cell proliferation ↑
AnxA2 PI3K-mTOR-SREBP-1c/Cyclin D1 signaling
pathway
[70]
Leucine and methionine (+)Bovine mammary epithelial cellsβ-casein, triglycerides,
Lactose synthesis ↑
Cell viability ↑
Cell proliferation ↑
mTOR-SREBP-1c signaling pathway[71]
Leucine and methionine (+)Bovine mammary epithelial cellsMilk protein ↑
Milk fat ↑
Cell proliferation ↑
GRP78-mTOR signaling pathway[30]
Leucine, acetate, and their interaction (+)Bovine mammary epithelial cells (1.8 mmol/L Leucine or 8–10 mmol/L acetate)Milk protein ↑JACK2/STAT5, mTOR, and AMPK pathway[42]
ValinePrimiparous gilts (total lysine: lysine = 0.93:1)Milk fat synthesis ↑——[45]
L-Valine (+)Porcine mammary epithelial cells (0.5 mmol/L)Cell numbers ↑
Protein synthesis ↑
mTOR and Ras/ERK signaling pathways[46]
Valine (+)Porcine mammary epithelial cellsFatty acids synthesis ↑
Intracellular triacylglycerol content ↑
AKT-mTOR-SREBP1 signaling pathway[47]
Valine (+)Holstein cowsMilk production ↑——[49]
Branched-chain amino acids (+)Multiparous sows (Yorkshire × Landrace)Milk production ↑——[50]
Lysine (+)Bovine mammary epithelial cells (0.70 mmol/L)Cells numbers ↑
Milk fat synthesis ↑
GPRC6A-PI3K-FABP5 signaling pathway[51]
Lysine (+)Bovine mammary epithelial cells (1.0 mmol/L)Protein synthesis ↑ATB0,+,mTOR and JAK2-STAT5 pathways[52]
Lysine (+)Bovine mammary epithelial cells (0.70 mmol/L)β-casein synthesis ↑SLC6A14-ERK-CDK1-mTOR signaling pathway[53]
Lysine (+)Mouse mammary epithelial cellsCell proliferation ↑PI3K/AKT/mTOR signal axis[54]
Lysine, methionine (+)Bovine mammary epithelial cells (Lys/Met ratio = 3:1, 1.2 mmol/L Lys, 0.4 mmol/L Met)Casein biosynthesis ↑JAK2/ELF5, mTOR, and its downstream RPS6KB1 and EIF4EBP1 signaling[55]
Methionine and arginine (+)Bovine mammary epithelial cellsα-s1-casein abundance ↑mTOR signaling; AMPK pathways[25]
Leucine or arginine (-)Mid-lactation Holstein cows (5 day continuous Leucine-163 g/d; arginine-158 g/d)Milk yield ↓
Milk protein yield ↓
——[72]
N-carbamoylglutamate (+)Holstein cows (20 g/d per cow, n = 15)Milk production ↑——[56]
5-aminolevulinic acid (+)Holstein cows (10 mg/kg per cow)Milk protein ↑;
Milk casein contents ↑
——[65]
Glutamine (+)Lactating sows (Large White) (1%)Milk yield ↑——[66]
Taurine (+)Bovine mammary epithelial cellsMilk protein ↑
Fat synthesis ↑
mTOR phosphorylation ↑
SREBP-1c expression ↑
GPR87-PI3K-SETD1A signaling pathway[62]
Leucine and histidine (+)Immortalized bovine mammary epithelial cell (0.45–10.80 mmol/L Leucine 0.15–9.60 mmol/L histidineMilk protein ↑mTOR signaling pathway[59]
Histidine, lysine, methionine, leucineImmortalized bovine mammary epithelial cell line (His: Lys: Met: Leu = 5:6:1:7)β-casein expression ↑mTOR signaling pathway[57]
Threonine, isoleucine, valine, leucineImmortalized bovine mammary epithelial cell line (Lysine: valine = 1.12:1 when Lysine: methionine is ideal)β-casein expression ↑mTOR signaling pathway[58]
Rumen-protected-methionine, lysine, histidine (+)Holstein cowsMilk production ↑——[68]
Rumen-protected gamma-aminobutyric acidHolstein dairy cowsMilk protein yield ↑——[67]
↑: increase; ↓: decrease; ——: reported but not fully confirmed.

2.2. Peptides

Peptides are formed by the dehydration condensation of two or more amino acids. It is involved in hormones, nerves, cell growth and reproduction and is highly significant for biologically active functions [73]. The proton-coupled oligopeptide transporter family (SLC15) plays an essential role in intracellular di- and tri-peptide uptake. The SLC15 family consists of four members, including PepT1 (SLC15A1), PepT2 (SLC15A2), Pht1 (SLC15A4) and Pht2 (SLC15A3) [74], of which both PepT1 and PepT2 play crucial roles in the mammary gland [75].
Peptides can enhance mammalian (bovine, goat) milk protein and affect its potential signaling pathway. Among them, mTOR still plays an important role. It has been confirmed that ghrelin, kisspeptin-10, dipeptide (methionyl-methionine), and substrate (threonyl-phenylalanyl-phenylalanine) are of great significance for promoting mammalian milk fat synthesis (Table 2). Ghrelin is a polypeptide hormone containing 28 amino acids isolated from the stomach. It is available in both acylated ghrelin (AG) and unacylated ghrelin (UAG) forms [76]. Ghrelin stimulates β-casein expression in cultured goat primary mammary epithelial cells [77]. In BMECs, both AG and UAG act on ERK1/2 and AKT signaling pathways to promote CSN2 expression [78]. ERKs are closely related to cell growth, proliferation, and differentiation [79]. Kisspeptin-10 is a neuropeptide hormone, a short peptide formed by the cleavage of Kisspeptin-54 [80]. A study has shown that 100 nmol/L of Kisspeptin-10 promotes β-casein synthesis in Holstein cow mammary epithelial cells through GPR54 and its downstream signaling pathways mTOR, ERK1/2, STAT5 and AKT [81]. Kisspeptins are the products of the Kiss-1 gene, and the Kiss-1/GPR54 system is expressed in mammalian mammary glands [82,83], thereby promoting downstream signaling. Dipeptide (Methionyl-Methionine) positively affects JAK2-STAT5 and mTOR signaling pathways in BMECs after being absorbed by PepT2, thereby promoting β-casein synthesis and cell proliferation [84]. Studies also showed that after stimulation of BMECs with tripeptides and lactogenic hormones, the abundance of PepT1 and PepT2 can be increased, thereby promoting milk fat synthesis [85,86]. In addition, it has been shown that OPH3-1, a fraction of the octopus protein hydrolysis product bioactive peptide, promotes mammary epithelial cell proliferation and casein synthesis in mice [87].
Compared with amino acids, there are relatively few studies on peptides promoting milk production. We find that the mechanism of peptide promoting milk production is similar to amino acids. The mTOR and JAK2-STAT5 signaling pathways are also important in peptide-promoted milk fat synthesis. In these pathways, related factors such as GPR54, ERK1/2 and AKT all affect milk fat synthesis. They will increase the expression of β-casein and PepT in mammary epithelial cells through the pathway and finally achieve the effect of promoting lactation.

3. The Enhancement of Milk Production by Lipids

Lipids include triglycerides and lipoids (phospholipids, sterols). Fatty acids are an essential source of energy and affect cellular and tissue metabolism, among others [88]. The mTOR pathway plays an important role in the promotion of milk fat synthesis by fatty acids. In addition, some studies have confirmed the positive effect of JAK2-STAT5 (Table 3). It is affected by many fatty acid-related factors in milk fat synthesis, such as SREBP1, peroxisome proliferators-activated receptor gamma (PPARG), fatty acid binding protein 3 (FABP3), etc. SREBPS is a significant factor regulating fat synthesis in animals. It regulates the activities of lipogenesis-related enzymes by regulating the gene transcription of lipogenesis-related enzymes, thereby controlling fat synthesis [89]. It has been shown that overexpression of SREBP1 promotes milk fat synthesis in cow and goat mammary epithelial cells [90,91]. PPAR can protect cells from oxidative damage, regulate glucose and lipid metabolism, and maintain the balance of inflammation in the body [92,93,94]. PPARG has an important link with the transcriptional regulation of fatty acids in the mammary gland [95]. FABP is a member of the intracellular lipid-binding protein superfamily. It plays a significant role in intracellular long-chain fatty acid uptake, transport and metabolism [96]. FABP3, one of the target genes of SREBP1, is the main protein for the rapid diffusion of long-chain fatty acids and the selective targeting of specific organelles for metabolism.
We can still find that researchers are exploring the effects of fatty acids on pathways related to milk fat synthesis at the cellular level (Table 3). Short-chain fatty acids such as acetic acid can promote BMECs proliferation and milk fat synthesis. Adding 6 mmol/L acetic acid to BMECs can stimulate the expression of fatty acid-related genes (such as SREBP1, mTOR, FABP3, etc.), and it is proved that the pathway is mTOR/eIF4E [97]. Acetic acid and β-hydroxybutyric acid work together to promote dairy cow mammary epithelial cells (DCMECs) triglyceride synthesis [98]. Studies of long-chain saturated and unsaturated fatty acids on milk fat synthesis have shown that saturated fatty acids, especially C18:0, can regulate milk fat synthesis in goats via PPARG [99]. Some C18 unsaturated fatty acids (e.g., oleic, linoleic, linolenic, palmitic, stearic, and palmitic) have also been identified. Oleic acid, stearic acid and palmitic acid can increase the accumulation of lipid droplets in BMECs by affecting the expression of FABP3, thereby upregulating SREBP1 and PPARG to promote milk fat synthesis [100]. Different ratios of unsaturated fatty acids (oleic, linoleic, and linolenic) can affect the synthesis of related proteins in BMECs [101]. Adding volatile fatty acids to Holstein dairy cattle feed can also increase the mRNA expression of PPARG, SREBPF1, FABP3 and other genes, and the optimal addition amount is 60 g BCVFA/head cow/day [102]. In addition, β-sitosterol is essential for milk fat synthesis in BMECs in the concentration range of 0.1-10 µmol/L, and the potential pathways are JAK2/STAT5 and mTOR signaling pathways [103].
In vivo, there are also many studies on increasing milk production by adding relevant fatty acids to the feed (Table 3). Compared to the addition of stearic acid, the addition of 2% palmitic acid to the feed increased milk fat concentration (+0.11%), milk yield (+0.09 kg/d), and 3.5% fat-corrected milk yield (+1.9 kg /d) in Holstein cows [104]. In a study, adding palmitic acid (C16:0) and changing the dietary n-6/n-3 fatty acid (FA) ratio increased milk production in Holstein dairy cows [105]. In several studies, the addition of linseed to feed was shown to improve milk production and milk quality [106,107,108]. Supplementation of soybean oil or linseed oil 20 mL/d to lactating Anglo-Nubian goat diets, respectively, increased milk production (+15% and 11.7%) [109]. A study confirmed that adding plant oils (2% rapeseed oil, 2% peanut oil and 2% sunflower oil) to the basal feed positively affected milk production. The addition of rapeseed oil had the most significant effect [110]. Adding fish oil can increase the milk production of Holstein cows [111]. It was demonstrated that adding 2% dietary fish oil increased milk production (+2.23 kg/d) in Polish Holstein-Friesian cows [112]. In addition, microalgae rich in polyunsaturated fatty acids are also of interest. Studies have shown that adding microalgae to the diet of ruminants can improve the fatty acid content of milk and dairy products. Dietary addition of 20 g Schizochytrium spp./ewe/day improved the fatty acid profile of milk [113]. Dietary intake of 10 g/head/day of Schizochytrium limacinum in goats significantly increased lactogenic acid and docosahexaenoic acid fatty acid concentrations in milk [114]. The inclusion of docosahexaenoic acid-rich microalgae (1.22 to 2.90 g/kg dry matter) in the feed may be beneficial for milk, fat-corrected milk and energy-corrected milk yield [115].
Therefore, we found a lot of literature to support that fatty acids promote the synthesis of milk yield, which is of great significance for improving milk yield. At the same time, we can learn that some fatty acids can also promote the proliferation of related cells and the synthesis of milk fat. Most fatty acids, such as acetic acid, oleic acid, palmitic acid, etc., have a positive effect on the regulation of milk fat synthesis. They can stimulate the expression of genes related to milk fat syntheses such as SREBP1, PPARG and FABP3. In addition, mTOR and JAK2-STAT5 signaling pathways still play an active role in promoting milk fat synthesis by β-sitosterol.
Table 3. Effects of fatty acids on milk production and its potential regulatory pathways.
Table 3. Effects of fatty acids on milk production and its potential regulatory pathways.
ItemsTreatmentsFunctionsPotential Signaling PathwaysReferences
Acetate (+)Bovine mammary epithelial cells (6 mmol/L)Milk fat synthesis ↑
Cell proliferation ↑
mTOR/eIF4E signaling pathway[97]
Acetate, β-hydroxybutyrate and their interaction (+)Dairy cow mammary epithelial cellsTriglyceride contents ↑
Lipid droplet formation ↑
SREBP1 signaling[98]
Long Chain Fatty Acids (+)Goat mammary epithelial cellsPPARG expression ↑——[99]
Branched-chain volatile fatty acids (+)Chinese Holstein cows (60 g BCVFA per cow
per day)
Milk fat synthesis ↑——[102]
β-sitosterol (+)Bovine mammary epithelial cells (0.1 to 10 μmol/L)β-casein synthesis ↑JAK2/STAT5 and mTOR signaling pathways[103]
Oleic acid, stearic acid, and palmitic acid (+)Dairy cow mammary epithelial cellsTriglyceride contents ↑
FABP3-SREBP1/PPARG signaling[100]
Oleic acid, linoleic acid, and linolenic acidBovine mammary epithelial cells (The ratio is 2:13.3:1)Fat and protein synthesis ↑——[101]
Palmitic acid (+)Holstein cowsMilk production ↑
Milk fat ↑
——[102]
Palmitic acid, n-6/n-3 fatty acids (+)Holstein cowsMilk production ↑——[103]
Linseed (+)Cilentana dairy goats (20%)Milk production ↑——[108]
Linseed (+)Italian Friesian dairy cows (700 g/head/d)Milk production ↑——[107]
Soybean, flaxseed oils (+)Anglo-Nubian goatsMilk production ↑——[109]
Linseed, verbascoside, vitamin E (+)Lacaune ewesMilk production ↑——[106]
Rapeseed oil (+)Holstein cowsMilk production ↑——[110]
Fish oil (+)Polish holstein-friesian cowsMilk production ↑——[112]
Fish oil (+)Holstein cowsMilk production ↑——[111]
Microalgae
Schizochytrium spp. (+)
Crossbred dairy ewes [Lacaune x Local (Greek) breed] Improves
milks’ fatty acid profile
——[113]
Schizochytrium limacinum marine algae (+)Multiparous Alpine goatsDHA and rumenic
acid concentration↑
——[114]
Docosahexaenoic acid-rich microalgae (+)Holstein cowsImproves
milks’ fatty acid profile
——[115]
↑: increase; ↓: decrease; ——: reported but not fully confirmed.

4. The Enhancement of Milk Production by Carbohydrates

Carbohydrates are tightly related to energy metabolism, and glucose is one of the most important carbohydrates. It is an essential precursor for lactose synthesis in the lactating mammary gland, providing fuel for milk synthesis and immune function in dairy cows [116]. The utilization of glucose by the mammary gland includes (1) synthesis of lactose; (2) generation of nicotinamide adenine dinucleotide phosphate (NADPH); (3) synthesis of milk fat; (4) production of energy; (5) synthesis of nucleic acids and amino acids [117]. In the mammalian mammary gland, two independent transport mechanisms are involved in glucose uptake: a facilitative transport mediated by the glucose transporter family and sodium-dependent transport mediated by the sodium+/glucose co-transporter [118]. The most important in glucose-promoted milk protein synthesis is the AMPK-mTOR pathway (Table 4). There is an interaction between mTOR and AMPK signaling pathways [119]. The adenosine monophosphate (AMP)-activated protein kinase (AMPK) can maintain energy balance under metabolic stress at the cellular and physiological levels and is closely related to intracellular energy metabolism [120]. AMPK is an upstream factor of mTOR. When AMPK is activated by other factors, mTOR signaling pathway is inhibited, leading to a reduction in the synthesis of related proteins [121].
The previous study has shown that the concentration of total amino acids in milk is highest when 60 g/d glucose is administered to lactating goats. During this process, added glucose may regulate the utilization of amino acids and the synthesis of related proteins through the AMPK-mTOR pathway [122]. In BMECs, adding glucose can regulate casein synthesis through the AMPK/mTOR signaling pathway [55]. In addition, adding chitosan to feed can alter rumen fermentation and increase milk production in lactating dairy cows [123].
It can be seen that there are not many studies on the effect of carbohydrates on lactation, but the studies show that glucose and chitosan can promote the synthesis of related proteins and milk production in dairy cows. They are linked to energy-related genes, including AMPK. As an upstream factor of mTOR, AMPK acts through the AMPK/mTOR pathway under the stimulation of glucose.

5. The Enhancement of Milk Production by Other Chemicals

In addition to the main nutritional factors related to milk protein synthesis, other chemicals—including phenolic compounds and lactation-related hormones (prolactin, estrogen, growth factors) [124]—also affect the synthesis of milk components (Table 5). Phenolic compounds have aromatic rings, and at least one hydroxyl group and phenolic derivatives can be divided into flavonoids and non-flavonoids. A study has shown that the 20 μmol/L daidzein (an isoflavone extract from soy)-treated BMECs can promote casein production and stimulate milk fat synthesis-related protein synthesis and cell proliferation through ERa-dependent NF-κB 1 activation [125]. Adding phenolic compounds extracted from pistacia lentiscus to BMECs can also increase milk content [126].
Several hormones, alone or in synergy, can play a crucial role in milk protein synthesis. Hormones generally act on breast cells to promote cell growth, promoting the synthesis of related milk proteins [124]. In the process of hormone-promoted milk synthesis, the related pathways still have mTOR and JAK-STAT signaling pathways (Table 5). The JAK-STAT signaling pathway is related to the transcriptional level and regulates the synthesis of mammary milk proteins, while the mTOR pathway affects the synthesis of related proteins at the translational level.
Prolactin is a polypeptide hormone. Prolactin can interact with specific receptors on mammary epithelial cells. Its receptors are located on the cell membrane, and the receptor sites on the membrane have a high affinity for hormones [127]. Estrogen is a steroid hormone, and its receptors include estrogen nuclear receptors and estrogen membrane receptors. It is also closely related to prolactin content and progesterone in breast cells [128]. Prolactin can stimulate the expression of LAT1 to increase milk protein synthesis [129]. The LAT1, also known as SLC7A5, is part of the SLC family and is closely related to amino acid transport [130]. Prolactin can activate Tudor staphylococcal nuclease (Tudor - SN), thereby regulating milk protein synthesis [131]. Estrogen-stimulated milk fat synthesis has been demonstrated in BMECs. The interaction between methionine and estrogen can promote milk fat synthesis and positively regulate SREBP-1c expression through FABP5 [29]. As previously described, FABP5 and SREBP-1c are associated with fatty acids promoting milk fat synthesis. The interaction between prolactin, estrogen and amino acids can encourage cell and milk synthesis. The validated pathways are AnxA2-mediated PI3K-mTOR-SREBP-1c/Cyclin D1, GRP78/mTOR, and U2AF65-mediated mTOR-SREBP-1c signaling pathway [30,70,71]. Annexin A2 (AnxA2) can interact with hormone receptors to increase the phosphorylation of PI3K, thereby affecting its downstream pathways [132]. Prolactin and epidermal growth factor (EGF) stimulate β-casein synthesis in mouse mammary epithelial cell line HC11 through the PI3-kinase/Akt/mTOR signaling pathway [133].
In addition, studies have shown that sodium butyrate can activate G protein-coupled receptor 41 (GPR41) and its downstream signaling pathways [134]. Sodium butyrate often replaces unstable butyric acid. GPR4 is associated with lipid metabolism and can be activated by short-chain fatty acids [135,136], thereby promoting the AMPK/mTOR/S6K signaling pathway and increasing the nuclear translocation of SREBP1 to promote milk fat synthesis in BMECs. Camellia seed oil and all-trans retinoic acid can promote casein and milk fat synthesis in BMECs, and the mTOR and JAK2/STAT5 signaling pathways can explain the mechanism [137,138]. Supplementation of 120 mg/500 kg of folic acid in dairy cows can increase milk production in peripartum cows [139]. Adding 20 mg GSPE (grape seed proanthocyanidin extract) /kg body weight/d to the diet can significantly improve the milk production of early lactating dairy cows [140]. Lutein-feeding lactating Holstein cows can promote lactose synthesis and metabolism, which may provide clues for the mechanism of lutein regulation of lactation in dairy cows [141]. In brief, phenolic compounds, lactation-related hormones and other chemicals can enhance milk synthesis. The mTOR pathway remains essential in these studies. These compounds can either activate the synthesis of related enzymes to enhance milk protein synthesis or stimulate factors related to the mTOR pathway.
Table 5. Effects of other chemicals on milk production and its potential signaling pathways.
Table 5. Effects of other chemicals on milk production and its potential signaling pathways.
ItemsTreatmentsFunctionsPotential Signaling PathwaysReferences
Daidzein (+)Primary bovine mammary epithelial cells (20 µmol/L)α- and β-casein ↑
Lipid synthesis ↑
Cell amount ↑
ERα-dependent NFƘB1 signaling[125]
Polyphenols from lentisk ethanolic extract (+)Bovine mammary epithelial cellsLactose synthesis ↑
Secretion of whey proteins ↑
Casein contents ↑
——[126]
Prolactin (+)Bovine mammary epithelial cellsMilk protein synthesis ↑LAT1 signaling[129]
Prolactin (+)Bovine mammary epithelial cellsMilk protein synthesis ↑
Tudor-SN expression ↑
——[131]
Estrogen (+)Bovine mammary epithelial cellsMilk fat synthesis ↑FABP5/SREBP-1c signaling[29]
Estrogen or prolactin (+)Bovine mammary epithelial cellsMilk synthesis ↑
Cell proliferation ↑
PI3K-mTOR-SREBP-1c/Cyclin D1 signaling pathway[70]
Estrogen and prolactin (+)Bovine mammary epithelial cellsMilk protein synthesis ↑
Milk fat synthesis ↑
Cell proliferation ↑
GRP78/mTOR signaling pathway[30]
Prolactin and β-estradiol (+)Bovine mammary epithelial cellsβ-casein synthesis ↑
Triglyceride synthesis ↑, Lactose synthesis ↑;
Cell proliferation ↑
U2AF65/mTOR-SREBP-1c
signaling pathway
[71]
Prolactin and epidermal growth factor (+)Mouse mammary epithelial cell line HC11β-casein ↑PI3K/Akt/mTOR signaling pathways[133]
Sodium butyrate (+)Bovine mammary epithelial cellsMilk fat synthesis ↑GPR41/AMPK/mTOR/S6K- SREBP1signaling pathway[134]
Camellia seed oil (+)Differentiated bovine mammary epithelial cellsβ-casein ↑PI3K-mTOR-S6K1 and JAK2-STAT5 signaling pathways[137]
All-trans retinoic acid (+)Bovine mammary epithelial cellsCasein synthesis ↑
Fatty acid composition ↑
JAK2/STAT5 pathway and downstream mTOR signaling pathway[138]
Folic acid (+)Lactating cows (120 mg/500 kg per cow)Milk production ↑——[139]
Grape seed proanthocyanidin extract (+)Holstein dairy cattle (20 mg GSPE/kg of body weight/day)Milk yield ↑——[140]
Lutein (+)Lactating Holstein cowsMilk lactose synthesis ↑——[141]
↑: increase; ↓: decrease; ——: reported but not fully confirmed.

6. STRING Database Analysis

In order to construct the interaction network between proteins and explore the core regulatory genes for milk production, we employ the STRING database (https://string-db.org/, accessed on 1 July 2022), a widely used database and online resource devoted to investigating functional protein association networks. We submitted 36 genes to the STRING database and specified the organism as cattle (Bos taurus). The result is shown in Figure 2.
The genes related to milk protein synthesis are widely investigated in milk production studies. The mammalian target of rapamycin (mTOR) is the most important regulator of these pathways. The mTOR is a class of serine/threonine kinases whose catalytic domains contain different phosphorylation sites [142]. The mTOR, together with its upstream genes (PIK3CB, MAPK1 and MAPK3) as well as its downstream genes (EIF4EBP1 and RPS6KB1), may enhance milk protein and nucleotide synthesis by inhibiting autophagy and other protein degradation pathway. Another complex pathway, the JAK2-STAT5A, may increase the RNA abundance of casein and thus enhance milk production.
The genes related to fat synthesis and metabolism are also crucial in milk production studies. Milk fat is the primary energy component of milk, and fatty acids are the essential components for milk fat synthesis. Milk fat synthesis and regulation are currently known to rely on SREBP1 and PPARG. The genes networks analysis showed that fatty acid metabolism genes, including FABP3 and FABP5 are in crosstalk with other essential genes and pathways mainly through PPARG. FABP3 and FABP5 belong to a fatty acid-binding proteins (FABPs) family, and the members of FABPs have formed proteins ranging from 14 to 15 kDa, which could participate in the uptake and transport of long-chain fatty acids. FABP3 and FABP5 genes are enriched in the KEGG pathways related to metabolism and glucose/energy metabolism, and Gene Ontology annotations related to transporter activity and cytoskeletal protein binding.
The transport systems that transport nutrients across membranes also play essential roles in promoting milk production. The genes—including SLC1A5, SLC6A14, SLC38A2, and SLC7A5—are all proven to be enrolled in the milk production process. SLC1A5 can transport all neutral amino acids, including glutamine, asparagine, and branched-chain and aromatic amino acids. SLC6A14 transports all amino acids with exception of the acidic ones: aspartate and glutamate. SLC38A2 transports neutral amino acids, and SLC7A5 uptakes sizeable neutral amino acids such as phenylalanine, tyrosine, L-DOPA, leucine, histidine, methionine and tryptophan. These results indicate that amino acids are necessary for milk production. As the need for protein and other nutrient increase during lactation, we assume the expression levels of transporters will be upregulated. If researchers further focus on the expression of transporters during lactation, we may know more about the nutrient needed during lactation. The study of transporter expression during lactation will become even more critical as the nutrients needed to improve milk quality during lactation become better understood.
We note that research on nutrient supplements to promote milk production is gaining traction. However, the entire pathway for milk production is unclear. Applying bioinformatics methods to the lactation process may help build the entire pathway network for milk production; mainly, the bioinformatics methods help to understand the effect of different nutrients, including amino lipid carbohydrates and other nutrients, on lactation. A better understanding of the associated trophic factors and signaling pathways may help us optimize the trophic factor profile in animal feeding.

7. Conclusion and Future Perspective

This review summarizes the enhancement of milk production by nutrient supplements and the related pathways. Most works focus on enhancing milk production with amino acids and fats. In contrast, a relatively small amount of work focuses on the enhancement of milk production with other chemicals, such as carbohydrates, phenols, and hormones. We found that the nutrients may influence the pathways related to protein synthesis as well as fat synthesis and metabolism. Additionally, the transport systems that transport nutrients across membranes also play essential roles in promoting milk production. Of all the pathways, mTOR, JAK2-STAT5A, SREBP1, and PPARG were frequently involved. We speculate that these pathways play essential roles in promoting milk production. However, the entire pathway for milk production still needs to be clarified. Further research should be carried out to interpret the enhancement of milk production by nutrient supplements.
Moreover, most current works employ mammary epithelial cells to investigate the enhancement of milk production. It is supposed that the results from dairy animals are more convincing than that from the cell model. As the genetic base of dairy animals has changed compared with decades ago, we suggest that future studies can be conducted again with the current breeds of dairy animals. We encourage validating cell-level experiments at the animal level in subsequent studies.

Author Contributions

F.P. and P.L.: writing—original draft preparation; G.H.: drawing pictures; Y.L.: sorting out forms; T.W. and B.L.: writing—review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the International Science and Technology Cooperation Project of Jilin Province (No. 20200801030GH), National Natural Science Foundation of China (No. 32101942), and Special project of Science and Technology Department of Zhejiang Province (No. 2022YSZX005).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We thank Arnold Boersma and Aditya Iyerfor for the suggestions and language corrections.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

The following abbreviations are used in this manuscript:
ATPAdenosine triphosphate
PI3KPhosphatidylinositol-3 Kinase
PKBProtein kinase B
mTORMammalian target of rapamycin
S6K1Ribosomal protein S6 kinase 1
4EBP14e-binding protein 1
mTORC1mTOR complex 1
SARSSeryl-tRNA synthetase
GlyRSGlycyl-tRNA synthetase
BMECsBovine mammary epithelial cells
SNAT2Sodium-coupled neutral amino acid transporter 2
ASCT2Amino-acid transporter 2
SREBP1Sterol response element-binding protein 1
FABP5Fatty acid binding protein 5
NCOA5Nuclear receptor co-activator 5
GRP78Glucose-regulated protein 78
LAT1L-type amino acid transporter 1
GATOR1GAP activity towards Rags 1
GATOR2GAP activity towards Rags 2
DDX59DEAD-Box Helicase 59
GNG12Guanine nucleotide-binding protein subunit gamma-12
JAK2Janus kinase 2
STAT5Signal transducers and activators of transcription 5
ERKExtracellular signal-regulated kinase
MAPKMitogen-activated protein kinase
GPRC6AG protein-coupled receptor family C group 6 member A
ELF5E74 Like ETS Transcription Factor 5)
RPS6KB1Ribosomal protein S6 kinase B1
EIF4EBP1Eukaryotic translation initiation factor 4E binding protein 1
NCGN-carbamoylglutamate
5-ALA5-aminolevulinic acid
FOXO1Forkhead Box O1
GPCRsG protein-coupled receptors
SETD1ASET domain containing 1A
SLC15Proton-coupled oligopeptide transporter family
AGAcylated ghrelin
UAGUnacylated ghrelin
MECsMammary epithelial cells
PPARGPeroxisome proliferators-activated receptor gamma
FABPFatty acid binding protein
NADPHNicotinamide adenine dinucleotide phosphate
AMPKAdenosine 5‘-monophosphate (AMP)-activated protein kinase
Tudor - SN  Tudor staphylococcal nuclease
U2AF65U2 snRNP auxiliary factor 65 kDa
AnxA2Annexin A2
EGFEpidermal growth factor
GPR41G protein-coupled receptor41
GSPEGrape seed proanthocyanidin extract
FFAR3Free fatty acid receptor 3

References

  1. FAO. Dairy Market Review: Emerging Trends and Outlook. Available online: https://www.fao.org/3/cb7982en/cb7982en.pdf (accessed on 16 December 2021).
  2. Smith, N.W.; Fletcher, A.J.; Hill, J.P.; McNabb, W.C. Modeling the Contribution of Milk to Global Nutrition. Front. Nutr. 2021, 8, 716100. [Google Scholar] [CrossRef] [PubMed]
  3. FAO. OECD-FAO Agricultural Outlook 2021–2030. Available online: https://www.fao.org/3/cb5332en/Dairy.pdf (accessed on 2 September 2021).
  4. Hegde, N.G. Livestock Development for Sustainable Livelihood of Small Farmers. Asian J. Res. Anim. Vet. Sci. 2019, 3, 1–17. [Google Scholar]
  5. Miglior, F.; Fleming, A.; Malchiodi, F.; Brito, L.F.; Martin, P.; Baes, C.F. A 100-Year Review: Identification and genetic selection of economically important traits in dairy cattle. J. Dairy Sci. 2017, 100, 10251–10271. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Saha, A.; Bhattacharyya, S. Artificial insemination for milk production in India: A statistical insight. Indian J. Anim. Sci. 2020, 90, 1186–1190. [Google Scholar] [CrossRef]
  7. Adesogan, A.T.; Dahl, G.E. MILK Symposium Introduction: Dairy production in developing countries. J. Dairy Sci. 2020, 103, 9677–9680. [Google Scholar] [CrossRef]
  8. Jones, G.M.; Pearson, R.E.; Clabaugh, G.A. Relationships between somatic cell counts and milk production. J. Dairy Sci. 1984, 67, 1823–1831. [Google Scholar] [CrossRef]
  9. Otto, P.I.; Guimaraes, S.E.F.; Calus, M.P.L.; Vandenplas, J.; Machado, M.A.; Panetto, J.C.C.; da Silva, M. Single-step genome-wide association studies (GWAS) and post-GWAS analyses to identify genomic regions and candidate genes for milk yield in Brazilian Girolando cattle. J. Dairy Sci. 2020, 103, 10347–10360. [Google Scholar] [CrossRef]
  10. Teissier, M.; Sanchez, M.P.; Boussaha, M.; Barbat, A.; Hoze, C.; Robert-Granie, C.; Croiseau, P. Use of meta-analyses and joint analyses to select variants in whole genome sequences for genomic evaluation: An application in milk production of French dairy cattle breeds. J. Dairy Sci. 2018, 101, 3126–3139. [Google Scholar] [CrossRef] [Green Version]
  11. Sexton, F. Average Milk Production of a Holstein Cow Per Day in India. Available online: https://fatinsl.info (accessed on 4 January 2020).
  12. Tricarico, J.M.; Kebreab, E.; Wattiaux, M.A. MILK Symposium review: Sustainability of dairy production and consumption in low-income countries with emphasis on productivity and environmental impact. J. Dairy Sci. 2020, 103, 9791–9802. [Google Scholar] [CrossRef]
  13. Kimball, S.R.; Jefferson, L.S. New functions for amino acids: Effects on gene transcription and translation. Am. J. Clin. Nutr. 2006, 83, 500S–507S. [Google Scholar] [CrossRef] [Green Version]
  14. Vanhaesebroeck, B.; Guillermet-Guibert, J.; Graupera, M.; Bilanges, B. The emerging mechanisms of isoform-specific PI3K signalling. Nat. Rev. Mol. Cell Biol. 2010, 11, 329–341. [Google Scholar] [CrossRef]
  15. Cantley, L.C. The phosphoinositide 3-kinase pathway. Science 2002, 296, 1655–1657. [Google Scholar] [CrossRef]
  16. Kandel, E.S.; Hay, N. The regulation and activities of the multifunctional serine/threonine kinase Akt/PKB. Exp. Cell Res. 2000, 253, 210–229. [Google Scholar] [CrossRef]
  17. Engelman, J.A. Targeting PI3K signalling in cancer: Opportunities, challenges and limitations. Nat. Rev. Cancer 2009, 9, 550–562. [Google Scholar] [CrossRef]
  18. Yu, J.S.; Cui, W. Proliferation, survival and metabolism: The role of PI3K/AKT/mTOR signalling in pluripotency and cell fate determination. Development 2016, 143, 3050–3060. [Google Scholar] [CrossRef] [Green Version]
  19. Hay, N.; Sonenberg, N. Upstream and downstream of mTOR. Genes Dev. 2004, 18, 1926–1945. [Google Scholar] [CrossRef] [Green Version]
  20. Li, B.; Basang, Z.; Hu, L.; Liu, L.; Jiang, N. Septin6 regulates cell growth and casein synthesis in dairy cow mammary epithelial cells via mTORC1 pathway. J. Dairy Res. 2019, 86, 181–187. [Google Scholar] [CrossRef]
  21. Luo, C.; Qi, H.; Huang, X.; Li, M.; Zhang, L.; Lin, Y.; Gao, X. GlyRS is a new mediator of amino acid-induced milk synthesis in bovine mammary epithelial cells. J. Cell. Physiol. 2019, 234, 2973–2983. [Google Scholar] [CrossRef]
  22. Dai, W.T.; White, R.R.; Liu, J.X.; Liu, H.Y. Seryl-tRNA synthetase-mediated essential amino acids regulate beta-casein synthesis via cell proliferation and mammalian target of rapamycin (mTOR) signaling pathway in bovine mammary epithelial cells. J. Dairy Sci. 2018, 101, 10456–10468. [Google Scholar] [CrossRef] [Green Version]
  23. Li, S.S.; Loor, J.J.; Liu, H.Y.; Liu, L.; Hosseini, A.; Zhao, W.S.; Liu, J.X. Optimal ratios of essential amino acids stimulate beta-casein synthesis via activation of the mammalian target of rapamycin signaling pathway in MAC-T cells and bovine mammary tissue explants. J. Dairy Sci. 2017, 100, 6676–6688. [Google Scholar] [CrossRef]
  24. Ma, Y.F.; Batistel, F.; Xu, T.L.; Han, L.Q.; Bucktrout, R.; Liang, Y.; Coleman, D.N.; Parys, C.; Loor, J.J. Phosphorylation of AKT serine/threonine kinase and abundance of milk protein synthesis gene networks in mammary tissue in response to supply of methionine in periparturient Holstein cows. J. Dairy Sci. 2019, 102, 4264–4274. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Hu, L.; Chen, Y.; Cortes, I.M.; Coleman, D.N.; Dai, H.; Liang, Y.; Parys, C.; Fernandez, C.; Wang, M.; Loor, J.J. Supply of methionine and arginine alters phosphorylation of mechanistic target of rapamycin (mTOR), circadian clock proteins, and α-s1-casein abundance in bovine mammary epithelial cells. Food Funct. 2020, 11, 883–894. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Dai, W.T.; Zhao, F.Q.; Liu, J.X.; Liu, H.Y. ASCT2 Is Involved in SARS-Mediated beta-Casein Synthesis of Bovine Mammary Epithelial Cells with Methionine Supply. J. Agric. Food Chem. 2020, 68, 13038–13045. [Google Scholar] [CrossRef] [PubMed]
  27. Qi, H.; Meng, C.; Jin, X.; Li, X.; Li, P.; Gao, X. 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. 2018, 66, 11027–11033. [Google Scholar] [CrossRef]
  28. Porstmann, T.; Santos, C.R.; Griffiths, B.; Cully, M.; Wu, M.; Leevers, S.; Griffiths, J.R.; Chung, Y.L.; Schulze, A. SREBP activity is regulated by mTORC1 and contributes to Akt-dependent cell growth. Cell Metab. 2008, 8, 224–236. [Google Scholar] [CrossRef] [Green Version]
  29. Li, P.; Yu, M.; Zhou, C.; Qi, H.; Wen, X.; Hou, X.; Li, M.; Gao, X. FABP5 is a critical regulator of methionine- and estrogen-induced SREBP-1c gene expression in bovine mammary epithelial cells. J. Cell. Physiol. 2018, 234, 537–549. [Google Scholar] [CrossRef] [Green Version]
  30. Liu, Y.; Wang, X.; Zhen, Z.; Yu, Y.; Qiu, Y.; Xiang, W. GRP78 regulates milk biosynthesis and the proliferation of bovinemammaryepithelial cells through the mTOR signaling pathway. Cell. Mol. Biol. Lett. 2019, 24, 57. [Google Scholar] [CrossRef] [Green Version]
  31. Yuan, X.; Zhang, L.; Cui, Y.; Yu, Y.; Gao, X.; Ao, J. NCOA5 is a master regulator of amino acid-induced mTOR activation and beta-casein synthesis in bovine mammary epithelial cells. Biochem. Biophys. Res. Commun. 2020, 529, 569–574. [Google Scholar] [CrossRef]
  32. Flores, A.; Mendoza, G.; Pinos-Rodriguez, J.M.; Plata, F.; Vega, S.; Bárcena, R. Effects of rumen-protected methionine on milk production of dairy goats. Ital. J. Anim. Sci. 2016, 8, 271–275. [Google Scholar] [CrossRef]
  33. Sen, E.; Orman, A.; Kara, C.; Turkmen, I.I.; Cetin, I. Improved Lactational Performance in Dairy Cows Supplemented with Methionine or Rumen-Protected Choline During the Transition Period. Kafkas Univ. Vet. Fak. Derg. 2018, 24, 2. [Google Scholar]
  34. King, L.; Wickramasinghe, J.; Dooley, B.; McCarthy, C.; Branstad, E.; Grilli, E.; Baumgard, L.; Appuhamy, R. Effects of Microencapsulated Methionine on Milk Production and Manure Nitrogen Excretions of Lactating Dairy Cows. Animals 2021, 11, 3545. [Google Scholar] [CrossRef]
  35. Toledo, M.Z.; Stangaferro, M.L.; Gennari, R.S.; Barletta, R.V.; Perez, M.M.; Wijma, R.; Sitko, E.M.; Granados, G.; Masello, M.; Van Amburgh, M.E.; et al. 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. [Google Scholar] [CrossRef]
  36. Park, J.K.; Yeo, J.M.; Bae, G.S.; Kim, E.J.; Kim, C.H. Effects of supplementing limiting amino acids on milk production in dairy cows consuming a corn grain and soybean meal-based diet. J. Anim. Sci. Technol. 2020, 62, 485–494. [Google Scholar] [CrossRef]
  37. Dodd, K.M.; Tee, A.R. Leucine and mTORC1: A complex relationship. Am. J. Physiol. Endocrinol. Metab. 2012, 302, E1329–E1342. [Google Scholar] [CrossRef]
  38. Saxton, R.A.; Knockenhauer, K.E.; Wolfson, R.L.; Chantranupong, L.; Pacold, M.E.; Wang, T.; Schwartz, T.U.; Sabatini, D.M. Structural basis for leucine sensing by the Sestrin2-mTORC1 pathway. Science 2016, 351, 53–58. [Google Scholar] [CrossRef] [Green Version]
  39. Zhang, J.; Zhang, X.; Liu, Y.; Su, Z.; Dawar, F.U.; Dan, H.; He, Y.; Gui, J.F.; Mei, J. Leucine mediates autophagosome-lysosome fusion and improves sperm motility by activating the PI3K/Akt pathway. Oncotarget 2017, 8, 111807–111818. [Google Scholar] [CrossRef]
  40. Qiu, Y.; Qu, B.; Zhen, Z.; Yuan, X.; Zhang, L.; Zhang, M. Leucine Promotes Milk Synthesis in Bovine Mammary Epithelial Cells via the PI3K-DDX59 Signaling. J. Agric. Food Chem. 2019, 67, 8884–8895. [Google Scholar] [CrossRef]
  41. Luo, C.; Zhao, S.; Dai, W.; Zheng, N.; Wang, J. Proteomic analyses reveal GNG12 regulates cell growth and casein synthesis by activating the Leu-mediated mTORC1 signaling pathway. Biochim. Biophys. Acta Proteins Proteom. 2018, 1866, 1092–1101. [Google Scholar] [CrossRef]
  42. Zhao, Y.L.; Yan, S.M.; Chen, L.; Shi, B.L.; Guo, X.Y. Effect of interaction between leucine and acetate on the milk protein synthesis in bovine mammary epithelial cells. Anim. Sci. J. 2019, 90, 81–89. [Google Scholar] [CrossRef] [Green Version]
  43. Tian, M.; Qi, Y.G.; Zhang, X.L.; Wu, Z.H.; Chen, J.M.; Chen, F.; Guan, W.T.; Zhang, S.H. Regulation of the JAK2-STAT5 Pathway by Signaling Molecules in the Mammary Gland. Front. Cell Dev. Biol. 2020, 8, 604896. [Google Scholar] [CrossRef]
  44. Wagner, K.U.; Rui, H. Jak2/Stat5 signaling in mammogenesis, breast cancer initiation and progression. J Mammary Gland. Biol. 2008, 13, 93–103. [Google Scholar] [CrossRef] [PubMed]
  45. Che, L.; Xu, M.; Gao, K.; Wang, L.; Yang, X.; Wen, X.; Xiao, H.; Li, M.; Jiang, Z. Mammary tissue proteomics in a pig model indicates that dietary valine supplementation increases milk fat content via increased de novo synthesis of fatty acid. Food Sci. Nutr. 2021, 9, 6213–6223. [Google Scholar] [CrossRef] [PubMed]
  46. Zhang, J.; He, W.; Yi, D.; Zhao, D.; Song, Z.; Hou, Y.; Wu, G. Regulation of protein synthesis in porcine mammary epithelial cells by L-valine. Amino. Acids 2019, 51, 717–726. [Google Scholar] [CrossRef] [PubMed]
  47. Che, L.; Xu, M.; Gao, K.; Zhu, C.; Wang, L.; Yang, X.; Wen, X.; Xiao, H.; Jiang, Z.; Wu, D. Valine increases milk fat synthesis in mammary gland of gilts through stimulating AKT/MTOR/SREBP1 pathwaydagger. Biol. Reprod. 2019, 101, 126–137. [Google Scholar] [CrossRef] [PubMed]
  48. Roux, P.P.; Shahbazian, D.; Vu, H.; Holz, M.K.; Cohen, M.S.; Taunton, J.; Sonenberg, N.; Blenis, J. RAS/ERK signaling promotes site-specific ribosomal protein S6 phosphorylation via RSK and stimulates cap-dependent translation. J. Biol. Chem. 2007, 282, 14056–14064. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Hultquist, K.M.; Casper, D.P. Effects of feeding rumen-degradable valine on milk production in late-lactating dairy cows. J. Dairy Sci. 2016, 99, 1201–1215. [Google Scholar] [CrossRef] [Green Version]
  50. Rezaei, R.; Gabriel, A.S.; Wu, G. Dietary supplementation with branched-chain amino acids enhances milk production by lactating sows and the growth of suckling piglets. J Anim. Sci. Biotechnol. 2022, 13, 65. [Google Scholar] [CrossRef]
  51. Li, X.; Li, P.; Wang, L.; Zhang, M.; Gao, X. 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. 2019, 67, 7005–7015. [Google Scholar] [CrossRef]
  52. Lin, X.; Li, S.; Zou, Y.; Zhao, F.Q.; Liu, J.; Liu, H. Lysine Stimulates Protein Synthesis by Promoting the Expression of ATB0,+ and Activating the mTOR Pathway in Bovine Mammary Epithelial Cells. J. Nutr. 2018, 148, 1426–1433. [Google Scholar] [CrossRef] [Green Version]
  53. Cao, Y.; Hu, G.; Li, W.; Wang, J.; Ge, Y.; Li, F.; Guo, W.; Kan, X.; Fu, S.; Liu, J. Lysine promotes proliferation and beta-casein synthesis through the SLC6A14-ERK1/2-CDK1-mTOR signaling pathway in bovine primary mammary epithelial cells. J. Therm. Biol. 2022, 110, 103375. [Google Scholar] [CrossRef]
  54. Li, W.; Long, X.Y.; Li, F.; Cao, Y.; Liu, J.X.; Fu, S.P.; Guo, W.J.; Hu, G.Q. Lysine stimulates the development of the murine mammary gland at puberty via PI3K/AKT/mTOR signalling axis. J. Anim. Physiol. Anim. Nutr. 2022, 106, 1420–1430. [Google Scholar] [CrossRef]
  55. Wang, F.; van Baal, J.; Ma, L.; Loor, J.J.; Wu, Z.L.; Dijkstra, J.; Bu, D.P. Short communication: Relationship between lysine/methionine ratios and glucose levels and their effects on casein synthesis via activation of the mechanistic target of rapamycin signaling pathway in bovine mammary epithelial cells. J. Dairy Sci. 2019, 102, 8127–8133. [Google Scholar] [CrossRef]
  56. Gu, F.; Miao, C.; Jiang, L.; Wang, D.; Liu, H.; Liu, J. Dietary supplementation with N-carbamoylglutamate initiated from the prepartum stage improves lactation performance of postpartum dairy cows. Anim. Nutr. 2021, 7, 232–238. [Google Scholar] [CrossRef]
  57. Gao, H.N.; Zhao, S.G.; Zheng, N.; Zhang, Y.D.; Wang, S.S.; Zhou, X.Q.; Wang, J.Q. Combination of histidine, lysine, methionine, and leucine promotes beta-casein synthesis via the mechanistic target of rapamycin signaling pathway in bovine mammary epithelial cells. J. Dairy Sci. 2017, 100, 7696–7709. [Google Scholar] [CrossRef]
  58. Dong, X.; Zhou, Z.; Wang, L.; Saremi, B.; Helmbrecht, A.; Wang, Z.; Loor, J.J. Increasing the availability of threonine, isoleucine, valine, and leucine relative to lysine while maintaining an ideal ratio of lysine:methionine alters mammary cellular metabolites, mammalian target of rapamycin signaling, and gene transcription. J. Dairy Sci. 2018, 101, 5502–5514. [Google Scholar] [CrossRef] [Green Version]
  59. Gao, H.N.; Hu, H.; Zheng, N.; Wang, J.Q. Leucine and histidine independently regulate milk protein synthesis in bovine mammary epithelial cells via mTOR signaling pathway. J. Zhejiang Univ. Sci. B 2015, 16, 560–572. [Google Scholar] [CrossRef] [Green Version]
  60. Inam, U.L.; Piao, F.; Aadil, R.M.; Suleman, R.; Li, K.; Zhang, M.; Wu, P.; Shahbaz, M.; Ahmed, Z. Ameliorative effects of taurine against diabetes: A review. Amino Acids 2018, 50, 487–502. [Google Scholar] [CrossRef]
  61. Solon, C.S.; Franci, D.; Ignacio-Souza, L.M.; Romanatto, T.; Roman, E.A.; Arruda, A.P.; Morari, J.; Torsoni, A.S.; Carneiro, E.M.; Velloso, L.A. Taurine enhances the anorexigenic effects of insulin in the hypothalamus of rats. Amino Acids 2012, 42, 2403–2410. [Google Scholar] [CrossRef]
  62. Yu, M.; Wang, Y.; Wang, Z.; Liu, Y.; Yu, Y.; Gao, X. Taurine promotes milk Synthesis via the GPR87-PI3K-SETD1A Signaling in BMECs. J. Agric. Food Chem. 2019, 67, 1927–1936. [Google Scholar] [CrossRef]
  63. Wauson, E.M.; Zaganjor, E.; Lee, A.Y.; Guerra, M.L.; Ghosh, A.B.; Bookout, A.L.; Chambers, C.P.; Jivan, A.; McGlynn, K.; Hutchison, M.R.; et al. The G protein-coupled taste receptor T1R1/T1R3 regulates mTORC1 and autophagy. Mol. Cell 2012, 47, 851–862. [Google Scholar] [CrossRef] [Green Version]
  64. Melick, C.H.; Lama-Sherpa, T.D.; Curukovic, A.; Jewell, J.L. G-Protein Coupled Receptor Signaling and Mammalian Target of Rapamycin Complex 1 Regulation. Mol. Pharmacol. 2022, 101, 181–190. [Google Scholar] [CrossRef] [PubMed]
  65. Hendawy, A.O.; Shirai, M.; Takeya, H.; Sugimura, S.; Miyanari, S.; Taniguchi, S.; Sato, K. Effects of 5-aminolevulinic acid supplementation on milk production, iron status, and immune response of dairy cows. J. Dairy Sci. 2019, 102, 11009–11015. [Google Scholar] [CrossRef] [PubMed]
  66. Yang, X.F.; Qin, J.F.; Wang, L.; Gao, K.G.; Zheng, C.T.; Huang, L.; Jiang, Z.Y. Improved Milk Glutamine Level and Growth Performance of Suckling Piglets by Glutamine Supplementation in Maternal Diet. Ann. Anim. Sci. 2018, 18, 441–452. [Google Scholar] [CrossRef] [Green Version]
  67. Wang, D.M.; Liu, H.Y.; Wang, C.; Liu, J.X.; Ferguson, J.D. Effects of rumen-protected gamma-aminobutyric acid on feed intake, performance and antioxidative status in transition cows. Livest. Sci. 2013, 153, 66–72. [Google Scholar] [CrossRef]
  68. Giallongo, F.; Harper, M.T.; Oh, J.; Lopes, J.C.; Lapierre, H.; Patton, R.A.; Parys, C.; Shinzato, I.; Hristov, A.N. Effects of rumen-protected methionine, lysine, and histidine on lactation performance of dairy cows. J. Dairy Sci. 2016, 99, 4437–4452. [Google Scholar] [CrossRef] [Green Version]
  69. Li, P.; Zhou, C.J.; Li, X.Y.; Yu, M.M.; Li, M.; Gao, X.J. CRTC2 Is a Key Mediator of Amino Acid-Induced Milk Fat Synthesis in Mammary Epithelial Cells. J. Agric. Food Chem. 2019, 67, 10513–10520. [Google Scholar] [CrossRef]
  70. Zhang, M.; Chen, D.; Zhen, Z.; Ao, J.; Yuan, X.; Gao, X. Annexin A2 positively regulates milk synthesis and proliferation of bovine mammary epithelial cells through the mTOR signaling pathway. J. Cell. Physiol. 2018, 233, 2464–2475. [Google Scholar] [CrossRef]
  71. Yu, Y.; Zhen, Z.; Qi, H.; Yuan, X.; Gao, X.; Zhang, M. U2AF65 enhances milk synthesis and growth of bovine mammary epithelial cells by positively regulating the mTOR-SREBP-1c signalling pathway. Cell Biochem. Funct. 2019, 37, 93–101. [Google Scholar] [CrossRef]
  72. Tian, W.; Wu, T.; Zhao, R.; Xu, J.; He, Y.; Wang, H. Responses of milk production of dairy cows to jugular infusions of a mixture of essential amino acids with or without exclusion leucine or arginine. Anim. Nutr. 2017, 3, 271–275. [Google Scholar] [CrossRef]
  73. Dicou, E. Biologically active, non membrane-anchored precursors: An overview. FEBS J. 2008, 275, 1960–1975. [Google Scholar] [CrossRef]
  74. Smith, D.E.; Clemencon, B.; Hediger, M.A. Proton-coupled oligopeptide transporter family SLC15: Physiological, pharmacological and pathological implications. Mol. Asp. Med. 2013, 34, 323–336. [Google Scholar] [CrossRef] [Green Version]
  75. Shennan, D.B.; Boyd, C.A. The functional and molecular entities underlying amino acid and peptide transport by the mammary gland under different physiological and pathological conditions. J. Mammary Gland. Biol. 2014, 19, 19–33. [Google Scholar] [CrossRef]
  76. Nishi, Y.; Yoh, J.; Hiejima, H.; Kojima, M. Structures and molecular forms of the ghrelin-family peptides. Peptides 2011, 32, 2175–2182. [Google Scholar] [CrossRef]
  77. Zhang, W.; Yu, G.; Huang, Y.; Xu, D.; Ren, J.; Jiang, L.; Wu, C.; Tong, D. Expression of ghrelin and GHSR-1a in mammary glands of dairy goat during the lactation and the effects of gherlin on regulation of mammary function in vitro. Mol. Cell. Endocrinol. 2013, 370, 20–31. [Google Scholar] [CrossRef]
  78. Li, S.; Liu, J.; Lv, Q.; Zhang, C.; Xu, S.; Yang, D.; Huang, B.; Zeng, Y.; Gao, Y.; Wang, W. AG and UAG induce β-casein expression via activation of ERK1/2 and AKT pathways. J. Mol. Endocrinol. 2016, 56, 213–225. [Google Scholar] [CrossRef]
  79. Lu, Z.; Xu, S. ERK1/2 MAP kinases in cell survival and apoptosis. IUBMB Life 2006, 58, 621–631. [Google Scholar] [CrossRef]
  80. Ulasov, I.V.; Borovjagin, A.V.; Timashev, P.; Cristofanili, M.; Welch, D.R. KISS1 in breast cancer progression and autophagy. Cancer Metastasis Rev. 2019, 38, 493–506. [Google Scholar] [CrossRef]
  81. Sun, J.; Liu, J.; Huang, B.; Kan, X.; Chen, G.; Wang, W.; Fu, S. Kisspeptin-10 Induces beta-Casein Synthesis via GPR54 and Its Downstream Signaling Pathways in Bovine Mammary Epithelial Cells. Int. J. Mol. Sci. 2017, 18, 2621. [Google Scholar] [CrossRef] [Green Version]
  82. Papaoiconomou, E.; Lymperi, M.; Petraki, C.; Philippou, A.; Msaouel, P.; Michalopoulou, P.; Kafiri, G.; Vassilakos, G.; Zografos, G.; Koutsilieris, M. Kiss-1/GPR54 Protein Expression in Breast Cancer. Anticancer Res. 2014, 34, 1401–1407. [Google Scholar]
  83. Goertzen, C.G.; Dragan, M.; Turley, E.; Babwah, A.V.; Bhattacharya, M. KISS1R signaling promotes invadopodia formation in human breast cancer cell via beta-arrestin2/ERK. Cell. Signal. 2016, 28, 165–176. [Google Scholar] [CrossRef]
  84. Wang, C.; Zhao, F.; Liu, J.; Liu, H. Dipeptide (Methionyl-Methionine) Transport and Its Effect on beta-Casein Synthesis in Bovine Mammary Epithelial Cells. Cell. Physiol. Biochem. 2018, 49, 479–488. [Google Scholar] [CrossRef] [PubMed]
  85. Zhou, M.M.; Wu, Y.M.; Liu, H.Y.; Zhao, K.; Liu, J.X. Effects of tripeptides and lactogenic hormones on oligopeptide transporter 2 in bovine mammary gland. J. Anim. Physiol. Anim. Nutr. 2011, 95, 781–789. [Google Scholar] [CrossRef] [PubMed]
  86. Cui, Y.; Zhang, X.; Guo, C.; Du, R.; Ailun, G.; Ao, C.; Gao, M. Identification and characterization of bovine mammary peptide transporters in response to tripeptide and lactogenic hormone treatment. Czech J. Food Sci. 2017, 62, 296–305. [Google Scholar] [CrossRef] [Green Version]
  87. Cai, B.N.; Wan, P.; Chen, H.; Chen, X.; Sun, H.L.; Pan, J.Y. Identification of octopus peptide and its promotion of beta-casein synthesis in a mouse mammary epithelial cell line. J. Food Biochem. 2020, 44, e13467. [Google Scholar] [CrossRef]
  88. Calder, P.C. Functional Roles of Fatty Acids and Their Effects on Human Health. JPEN J. Parenter. Enteral Nutr. 2015, 39, 18s–32s. [Google Scholar] [CrossRef]
  89. Eberle, D.; Hegarty, B.; Bossard, P.; Ferre, P.; Foufelle, F. SREBP transcription factors: Master regulators of lipid homeostasis. Biochimie 2004, 86, 839–848. [Google Scholar] [CrossRef]
  90. Li, N.; Zhao, F.; Wei, C.; Liang, M.; Zhang, N.; Wang, C.; Li, Q.Z.; Gao, X.J. Function of SREBP1 in the milk fat synthesis of dairy cow mammary epithelial cells. Int. J. Mol. Sci. 2014, 15, 16998–17013. [Google Scholar] [CrossRef] [Green Version]
  91. Xu, H.F.; Luo, J.; Zhao, W.S.; Yang, Y.C.; Tian, H.B.; Shi, H.B.; Bionaz, M. Overexpression of SREBP1 (sterol regulatory element binding protein 1) promotes de novo fatty acid synthesis and triacylglycerol accumulation in goat mammary epithelial cells. J. Dairy Sci. 2016, 99, 783–795. [Google Scholar] [CrossRef] [Green Version]
  92. Yao, X.; Jiang, Q.; Ding, W.; Yue, P.; Wang, J.; Zhao, K.; Zhang, H. Interleukin 4 inhibits high mobility group box-1 protein-mediated NLRP3 inflammasome formation by activating peroxisome proliferator-activated receptor-gamma in astrocytes. Biochem. Biophys. Res. Commun. 2019, 509, 624–631. [Google Scholar] [CrossRef]
  93. Ivanova, E.A.; Myasoedova, V.A.; Melnichenko, A.A.; Orekhov, A.N. Peroxisome Proliferator-Activated Receptor (PPAR) Gamma Agonists as Therapeutic Agents for Cardiovascular Disorders: Focus on Atherosclerosis. Curr. Pharm. Des. 2017, 23, 1119–1124. [Google Scholar] [CrossRef]
  94. Skat-Rordam, J.; Hojland Ipsen, D.; Lykkesfeldt, J.; Tveden-Nyborg, P. A role of peroxisome proliferator-activated receptor gamma in non-alcoholic fatty liver disease. Basic Clin. Pharmacol. Toxicol. 2019, 124, 528–537. [Google Scholar] [CrossRef]
  95. Bionaz, M.; Loor, J.J. Gene networks driving bovine milk fat synthesis during the lactation cycle. BMC Genom. 2008, 9, 366. [Google Scholar] [CrossRef] [Green Version]
  96. Chmurzynska, A. The multigene family of fatty acid-binding proteins (FABPs): Function, structure and polymorphism. J. Appl. Genet. 2006, 47, 39–48. [Google Scholar] [CrossRef]
  97. Zhao, Y.; Guo, X.; Yan, S.; Shi, B.; Sheng, R. Acetate regulates milk fat synthesis through the mammalian target of rapamycin/eukaryotic initiation factor 4E signaling pathway in bovine mammary epithelial cells. J. Dairy Sci. 2021, 104, 337–345. [Google Scholar] [CrossRef]
  98. Ali, I.; Li, C.; Li, L.; Kuang, M.; Shafiq, M.; Wang, Y.; Yang, M.; Wang, G. Effect of acetate, beta-hydroxybutyrate and their interaction on lipogenic gene expression, triglyceride contents and lipid droplet formation in dairy cow mammary epithelial cells. Vitr. Cell. Dev. Biol. Anim. 2021, 57, 66–75. [Google Scholar] [CrossRef]
  99. Vargas-Bello-Pérez, E.; Zhao, W.; Bionaz, M.; Luo, J.; Loor, J.J. Nutrigenomic effect of saturated and unsaturated long chain fatty acids on lipid-related genes in goat mammary epithelial cells: What is the role of PPARγ? J. Vet. Sci. 2019, 6, 54. [Google Scholar] [CrossRef] [Green Version]
  100. Liang, M.Y.; Hou, X.M.; Qu, B.; Zhang, N.; Li, N.; Cui, Y.J.; Li, Q.Z.; Gao, X.J. Functional analysis of FABP3 in the milk fat synthesis signaling pathway of dairy cow mammary epithelial cells. In Vitro Cell. Dev. Biol. Anim. 2014, 50, 865–873. [Google Scholar] [CrossRef]
  101. Sheng, R.; Yan, S.M.; Qi, L.Z.; Zhao, Y.L. Effect of the ratios of unsaturated fatty acids on the expressions of genes related to fat and protein in the bovine mammary epithelial cells. Vitr. Cell. Dev. Biol. Anim. 2015, 51, 381–389. [Google Scholar] [CrossRef]
  102. Liu, Q.; Wang, C.; Guo, G.; Huo, W.J.; Zhang, S.L.; Pei, C.X.; Zhang, Y.L.; Wang, H. Effects of branched-chain volatile fatty acids on lactation performance and mRNA expression of genes related to fatty acid synthesis in mammary gland of dairy cows. Animal 2018, 12, 2071–2079. [Google Scholar] [CrossRef]
  103. Liu, X.; Shen, J.; Zong, J.; Liu, J.; Jin, Y. Beta-Sitosterol Promotes Milk Protein and Fat Syntheses-Related Genes in Bovine Mammary Epithelial Cells. Animals 2021, 11, 3238. [Google Scholar] [CrossRef]
  104. Rico, J.E.; Allen, M.S.; Lock, A.L. Compared with stearic acid, palmitic acid increased the yield of milk fat and improved feed efficiency across production level of cows. J. Dairy Sci. 2014, 97, 1057–1066. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Ghasemi, E.; Golabadi, D.; Piadeh, A. Effect of supplementing palmitic acid and altering the dietary ratio of n-6:n-3 fatty acids in low-fibre diets on production responses of dairy cows. Br. J. Nutr. 2021, 126, 355–365. [Google Scholar] [CrossRef] [PubMed]
  106. Casamassima, D.; Nardoia, M.; Palazzo, M.; Vizzarri, F.; D’Alessandro, A.G.; Corino, C. Effect of dietary extruded linseed, verbascoside and vitamin E supplements on yield and quality of milk in Lacaune ewes. J. Dairy Res. 2014, 81, 485–493. [Google Scholar] [CrossRef] [PubMed]
  107. Pezzi, P.; Giammarco, M.; Vignola, G.; Brogna, N. Effects of extruded linseed dietary supplementation on milk yield, milk quality and lipid metabolism of dairy cows. Ital. J. Anim. Sci. 2016, 6, 333–335. [Google Scholar] [CrossRef]
  108. Musco, N.; Tudisco, R.; Esposito, G.; Iommelli, P.; Totakul, P.; D’Aniello, B.; Lombardi, P.; Amato, R.; Wanapat, M.; Infascelli, F. Effects of Linseed Supplementation on Milk Production, Composition, Odd- and Branched-Chain Fatty Acids, and on Serum Biochemistry in Cilentana Grazing Goats. Animals 2022, 12, 783. [Google Scholar] [CrossRef]
  109. Kholif, A.E.; Morsy, T.A.; Abd El Tawab, A.M.; Anele, U.Y.; Galyean, M.L. Effect of Supplementing Diets of Anglo-Nubian Goats with Soybean and Flaxseed Oils on Lactational Performance. J. Agric. Food Chem. 2016, 64, 6163–6170. [Google Scholar] [CrossRef]
  110. Dai, X.J.; Wang, C.; Zhu, Q. Milk performance of dairy cows supplemented with rapeseed oil, peanut oil and sunflower seed oil. Czech J. Anim. Sci. 2011, 56, 181–191. [Google Scholar] [CrossRef] [Green Version]
  111. Barfourooshi, H.J.; Towhidi, A.; Sadeghipanah, H.; Zhandi, M.; Zeinoaldini, S.; Dirandeh, E.; Akers, R.M. Effect of Dietary Fish Oil on Mammary Gland Development and Milk Production of Holstein Cow. Ann. Anim. Sci. 2018, 18, 973–990. [Google Scholar] [CrossRef] [Green Version]
  112. Kupczynski, R.; Szoltysik, M.; Janeczek, W.; Chrzanowska, J.; Kinal, S.; Kroliczewska, B. Effect of dietary fish oil on milk yield, fatty acids content and serum metabolic profile in dairy cows. J. Anim. Physiol. Anim. Nutr. 2011, 95, 512–522. [Google Scholar] [CrossRef]
  113. Zisis, F.; Kyriakaki, P.; Satolias, F.F.; Mavrommatis, A.; Simitzis, P.E.; Pappas, A.C.; Surai, P.F.; Tsiplakou, E. The Effect of Dietary Inclusion of Microalgae Schizochytrium spp. on Ewes’ Milk Quality and Oxidative Status. Foods 2022, 11, 2950. [Google Scholar] [CrossRef]
  114. Pajor, F.; Egerszegi, I.; Szucs, A.; Poti, P.; Bodnar, A. Effect of Marine Algae Supplementation on Somatic Cell Count, Prevalence of Udder Pathogens, and Fatty Acid Profile of Dairy Goats’ Milk. Animals 2021, 11, 1097. [Google Scholar] [CrossRef]
  115. Marques, J.A.; Del Valle, T.A.; Ghizzi, L.G.; Zilio, E.M.C.; Gheller, L.S.; Nunes, A.T.; Silva, T.B.P.; Dias, M.; Grigoletto, N.T.S.; Koontz, A.F.; et al. Increasing dietary levels of docosahexaenoic acid-rich microalgae: Ruminal fermentation, animal performance, and milk fatty acid profile of mid-lactating dairy cows. J. Dairy Sci. 2019, 102, 5054–5065. [Google Scholar] [CrossRef]
  116. Habel, J.; Sundrum, A. Mismatch of Glucose Allocation between Different Life Functions in the Transition Period of Dairy Cows. Animals 2020, 10, 1028. [Google Scholar] [CrossRef]
  117. Dienel, G.A. Brain Glucose Metabolism: Integration of Energetics with Function. Physiol. Rev. 2019, 99, 949–1045. [Google Scholar] [CrossRef]
  118. Zhao, F.Q.; Keating, A.F. Expression and regulation of glucose transporters in the bovine mammary gland. J. Dairy Sci. 2007, 90 (Suppl. 1), E76–E86. [Google Scholar] [CrossRef] [Green Version]
  119. Xu, J.; Ji, J.; Yan, X.H. Cross-talk between AMPK and mTOR in regulating energy balance. Crit. Rev. Food Sci. Nutr. 2012, 52, 373–381. [Google Scholar] [CrossRef]
  120. Kim, J.; Yang, G.; Kim, Y.; Kim, J.; Ha, J. AMPK activators: Mechanisms of action and physiological activities. Exp. Mol. Med. 2016, 48, 224. [Google Scholar] [CrossRef] [Green Version]
  121. Tokunaga, C.; Yoshino, K.; Yonezawa, K. mTOR integrates amino acid- and energy-sensing pathways. Biochem. Biophys. Res. Commun. 2004, 313, 443–446. [Google Scholar] [CrossRef]
  122. Cai, J.; Wang, D.; Zhao, F.Q.; Liang, S.; Liu, J. AMPK-mTOR pathway is involved in glucose-modulated amino acid sensing and utilization in the mammary glands of lactating goats. J. Anim. Sci. Biotechnol. 2020, 11, 32. [Google Scholar] [CrossRef]
  123. Gomes de Paiva, P.; Ferreira de Jesus, E.; Del Valle, T.A.; Ferreira de Almeida, G.; Costa, A.G.B.V.B.; Consentini, C.E.C.; Zanferari, F.; Takiya, C.S.; Bueno, I.C.d.S.; Rennó, F.P. Effects of chitosan on ruminal fermentation, nutrient digestibility, and milk yield and composition of dairy cows. Anim. Prod. Sci. 2017, 57, 10939–10952. [Google Scholar] [CrossRef] [Green Version]
  124. Akers, R.M. Major advances associated with hormone and growth factor regulation of mammary growth and lactation in dairy cows. J. Dairy Sci. 2006, 89, 1222–1234. [Google Scholar] [CrossRef] [PubMed]
  125. 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. 2022, 33, 43–52. [Google Scholar] [CrossRef] [PubMed]
  126. Hadaya, O.; Bransi-Nicola, R.; Shalev, Y.; Azaizeh, H.; Roth, Z.; Muklada, H.; Deutch, T.; Landau, S.Y.; Argov-Argaman, N. Pistacia lentiscus extract enhances mammary epithelial cells’ productivity by modulating their oxidative status. Sci. Rep. 2020, 10, 20985. [Google Scholar] [CrossRef] [PubMed]
  127. Shiu, R.P.; Friesen, H.G. Mechanism of action of prolactin in the control of mammary gland function. Annu. Rev. Physiol. 1980, 42, 83–96. [Google Scholar] [CrossRef]
  128. Fendrick, J.L.; Raafat, A.M.; Haslam, S.Z. Mammary gland growth and development from the postnatal period to postmenopause: Ovarian steroid receptor ontogeny and regulation in the mouse. J. Mammary Gland. Biol. 1998, 3, 7–22. [Google Scholar] [CrossRef]
  129. Zhou, J.; Jiang, M.; Shi, Y.; Song, S.; Hou, X.; Lin, Y. Prolactin regulates LAT1 expression via STAT5 (signal transducer and activator of transcription 5) signaling in mammary epithelial cells of dairy cows. J. Dairy Sci. 2020, 103, 6627–6634. [Google Scholar] [CrossRef]
  130. Sokolov, A.M.; Holmberg, J.C.; Feliciano, D.M. The amino acid transporter Slc7a5 regulates the mTOR pathway and is required for granule cell development. Hum. Mol. Genet. 2020, 29, 3003–3013. [Google Scholar] [CrossRef]
  131. Zhang, S.; Qi, H.; Wen, X.P.; Li, P.; Gao, X.J.; Ao, J.X. The phosphorylation of Tudor-SN mediated by JNK is involved in the regulation of milk protein synthesis induced by prolactin in BMECs. J. Cell. Physiol. 2019, 234, 6077–6090. [Google Scholar] [CrossRef]
  132. Raynal, P.; Pollard, H.B. Annexins: The problem of assessing the biological role for a gene family of multifunctional calcium- and phospholipid-binding proteins. Biochim. Biophys. Acta 1994, 1197, 63–93. [Google Scholar] [CrossRef]
  133. Pauloin, A.; Chanat, E. Prolactin and epidermal growth factor stimulate adipophilin synthesis in HC11 mouse mammary epithelial cells via the PI3-kinase/Akt/mTOR pathway. Biochim. Biophys. Acta 2012, 1823, 987–996. [Google Scholar] [CrossRef] [Green Version]
  134. Cheng, J.; Zhang, Y.; Ge, Y.; Li, W.; Cao, Y.; Qu, Y.; Liu, S.; Guo, Y.; Fu, S.; Liu, J. Sodium butyrate promotes milk fat synthesis in bovine mammary epithelial cells via GPR41 and its downstream signalling pathways. Life Sci. 2020, 259, 118375. [Google Scholar] [CrossRef]
  135. Nilsson, N.E.; Kotarsky, K.; Owman, C.; Olde, B. Identification of a free fatty acid receptor, FFA2R, expressed on leukocytes and activated by short-chain fatty acids. Biochem. Biophys. Res. Commun. 2003, 303, 1047–1052. [Google Scholar] [CrossRef]
  136. Brown, A.J.; Goldsworthy, S.M.; Barnes, A.A.; Eilert, M.M.; Tcheang, L.; Daniels, D.; Muir, A.I.; Wigglesworth, M.J.; Kinghorn, I.; Fraser, N.J.; et al. The Orphan G protein-coupled receptors GPR41 and GPR43 are activated by propionate and other short chain carboxylic acids. J. Biol. Chem. 2003, 278, 11312–11319. [Google Scholar] [CrossRef] [Green Version]
  137. Zhong, W.; Shen, J.; Liao, X.; Liu, X.; Zhang, J.; Zhou, C.; Jin, Y. Camellia (Camellia oleifera Abel.) seed oil promotes milk fat and protein synthesis-related gene expression in bovine mammary epithelial cells. Food Sci. Nutr. 2020, 8, 419–427. [Google Scholar] [CrossRef] [Green Version]
  138. Liao, X.D.; Zhou, C.H.; Zhang, J.; Shen, J.L.; Wang, Y.J.; Jin, Y.C.; Li, S.L. Effect of all-trans retinoic acid on casein and fatty acid synthesis in MAC-T cells. Asian-Australas. J. Anim. Sci. 2020, 33, 1012–1022. [Google Scholar] [CrossRef]
  139. Khan, M.Z.; Liu, L.; Zhang, Z.; Khan, A.; Wang, D.; Mi, S.; Usman, T.; Liu, G.; Guo, G.; Li, X.; et al. Folic acid supplementation regulates milk production variables, metabolic associated genes and pathways in perinatal Holsteins. J. Anim. Physiol. Anim. Nutr. 2020, 104, 483–492. [Google Scholar] [CrossRef]
  140. Huang, Y.; Oikonomou, G.; Hu, J.; Li, Y.; Du, X.; Du, Y.; Liu, Y.; Zhang, P.; Wang, P.; Yu, H.; et al. Effect of feeding grape seed Proanthocyanidin extract on production performance, metabolic and anti-oxidative status of dairy cattle. Arq. Bras. Med. Vet. Zootec. 2019, 71, 1207–1216. [Google Scholar] [CrossRef] [Green Version]
  141. Wang, C.; Wang, C.; Liu, J.; Liu, H. Proteomic analysis of the effects of lutein on mammary gland metabolism in dairy cows. J. Dairy Res. 2018, 85, 152–156. [Google Scholar] [CrossRef]
  142. de Morentin, P.B.M.; Martinez-Sanchez, N.; Roa, J.; Ferno, J.; Nogueiras, R.; Tena-Sempere, M.; Dieguez, C.; Lopez, M. Hypothalamic mTOR: The Rookie Energy Sensor. Curr. Mol. Med. 2014, 14, 3–21. [Google Scholar] [CrossRef]
Figure 1. The regulation of milk synthesis by methionine, leucine, valine and lysine. The amino acids were transport across membrance by different transportors including LAT1, LAT2, SNAT2, ASCT2, and ATB0,+, respectively. GPRC6A: G protein-coupled receptor class C group 6 member A, is a basic amino acid receptor. The mammalian target of rapamycin complex 1 (mTORC1) is the most important regulator of these pathways. The activation of mTORC1 increases milk protein synthesis and regulates milk fat synthesis.
Figure 1. The regulation of milk synthesis by methionine, leucine, valine and lysine. The amino acids were transport across membrance by different transportors including LAT1, LAT2, SNAT2, ASCT2, and ATB0,+, respectively. GPRC6A: G protein-coupled receptor class C group 6 member A, is a basic amino acid receptor. The mammalian target of rapamycin complex 1 (mTORC1) is the most important regulator of these pathways. The activation of mTORC1 increases milk protein synthesis and regulates milk fat synthesis.
Animals 13 00419 g001
Figure 2. Network of lactation-related proteins under nutritional supplementation.
Figure 2. Network of lactation-related proteins under nutritional supplementation.
Animals 13 00419 g002
Table 2. Effects of peptides on milk production and its potential regulatory pathways.
Table 2. Effects of peptides on milk production and its potential regulatory pathways.
ItemsTreatmentsFunctionsPotential Signaling PathwaysReferences
Ghrelin (+)Mature Guanzhong Saanen dairy goats and mammary epithelial cellsβ-Casein synthesis ↑——[77]
Ghrelin (+)Primary bovine mammary epithelial cellsβ-Casein synthesis ↑ERK1/2 and AKT signaling pathways[78]
Kisspeptin-10 (+)Bovine mammary epithelial cells (100 nmol/L)β-Casein Synthesis ↑CSN2 via GPR54 and its downstream signaling pathways mTOR, ERK1/2, STAT5 and AKT[81]
Methionyl-methionine dipeptide (+)Bovine mammary epithelial cells (80 µg/mL)PepT2 expression ↑
β- Casein synthesis ↑
AK2-STAT5 and mTOR signaling pathways[84]
Threonyl-phenylalanyl-phenylalanine (+)Bovine mammary epithelial cells (5, 10 and 15%)PepT2 mRNA abundance↑——[85]
Threonyl-phenylalanyl-phenylalanine (+)Bovine mammary epithelial cells (add lactogenic hormone treatment)PepT mRNA abundance↑——[86]
Octopus peptide (+)Mouse mammary epithelial cell lineβ- Casein synthesis ↑
Cell proliferation ↑
——[87]
↑: increase; ↓: decrease; ——: reported but not fully confirmed.
Table 4. Effects of carbohydrates on milk production and its potential regulatory pathways.
Table 4. Effects of carbohydrates on milk production and its potential regulatory pathways.
ItemsTreatmentsFunctionsPotential Signaling PathwaysReferences
Glucose (+)lactating dairy goats (60 g/d)Amino acid ↑AMPK-mTOR signaling pathways[122]
Glucose (+)Bovine mammary epithelial cellsβ-casein ↑
Cell proliferation ↑
AMPK/mTOR signaling pathways[55]
Chitosan (+)Holstein cows (225 mg/kg bodyweight)Milk yield ↑
Fat-corrected milk, protein and lactose production ↑
——[123]
↑: increase; ↓: decrease; ——: reported but not fully confirmed.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Pan, F.; Li, P.; Hao, G.; Liu, Y.; Wang, T.; Liu, B. Enhancing Milk Production by Nutrient Supplements: Strategies and Regulatory Pathways. Animals 2023, 13, 419. https://doi.org/10.3390/ani13030419

AMA Style

Pan F, Li P, Hao G, Liu Y, Wang T, Liu B. Enhancing Milk Production by Nutrient Supplements: Strategies and Regulatory Pathways. Animals. 2023; 13(3):419. https://doi.org/10.3390/ani13030419

Chicago/Turabian Style

Pan, Fengguang, Peizhi Li, Guijie Hao, Yinuo Liu, Tian Wang, and Boqun Liu. 2023. "Enhancing Milk Production by Nutrient Supplements: Strategies and Regulatory Pathways" Animals 13, no. 3: 419. https://doi.org/10.3390/ani13030419

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop