Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 24(18)2023 Sep 20.
Article in English | MEDLINE | ID: mdl-37762652

ABSTRACT

The mammary gland undergoes intensive remodeling during the lactation cycle, and the involution process of mammary gland contains extensive epithelial cells involved in the process of autophagy. Our studies of mice mammary glands suggest that miR-30a-3p expression was low during involution compared with its high expression in the mammary glands of lactating mice. Then, we revealed that miR-30a-3p negatively regulated autophagy by autophagy related 12 (Atg12) in mouse mammary gland epithelial cells (MMECs). Restoring ATG12, knocking down autophagy related 5 (Atg5), starvation, and Rapamycin were used to further confirm this conclusion. Overexpression of miR-30a-3p inhibited autophagy and altered mammary structure in the involution of the mammary glands of mice, which was indicative of alteration in mammary remodeling. Taken together, these results elucidated the molecular mechanisms of miR-30a-3p as a key induction mediator of autophagy by targeting Atg12 within the transition period between lactation and involution in mammary glands.


Subject(s)
Autophagy , Mammary Glands, Animal , MicroRNAs , Animals , Female , Mice , Autophagy/genetics , Autophagy-Related Protein 5 , Epithelial Cells , Lactation/genetics , MicroRNAs/genetics , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/metabolism
2.
In Vitro Cell Dev Biol Anim ; 56(3): 243-252, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32078727

ABSTRACT

The prolactin/STAT5 and AKT1/mTOR pathways play a key role in milk protein transcription and translation, respectively. However, the correlation between them in bovine mammary epithelial cells remains unclear. Here, mRNA and protein expression levels of AKT1, STAT5, and mTOR and the phosphorylation of these proteins were determined. Cell proliferation and viability were examined using the CASY-TT assay. Purified bovine mammary epithelial cells were cultured in differentiation media for different periods. The basic differentiation medium contained a lactogenic hormone cocktail of insulin (5 µg/mL), hydrocortisone (1 µg/mL), and prolactin (5 µg/mL). The cells cultured in this medium grew slowly and expressed higher levels of p-STAT5, p-AKT1, and p-mTOR (activated form) than those of control cells. Although the phosphorylation ratio was not increased, transcription and translation of these proteins were upregulated by the addition of insulin-like growth factor-1 or growth hormone, which further increased ß-casein mRNA expression. Furthermore, the three proteins were upregulated or downregulated synchronously, even after RNA interference silencing of either Stat5 or Akt1. These findings indicate that a few hormones and other factors play lactogenic and galactogenic roles by promoting two key lactogenic signaling associated with milk protein expression. We provide evidence of prolactin/STAT5 and AKT1/mTOR synchronization, establishing a direct correlation between transcription regulation and translation regulation of milk protein in bovine mammary epithelial cells.


Subject(s)
Epithelial Cells/metabolism , Mammary Glands, Animal/cytology , Prolactin/metabolism , Proto-Oncogene Proteins c-akt/metabolism , STAT5 Transcription Factor/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Cattle , Cell Proliferation , Cell Survival , Cells, Cultured , Female , Gene Knockdown Techniques , Signal Transduction
3.
J Cell Physiol ; 234(7): 10101-10110, 2019 07.
Article in English | MEDLINE | ID: mdl-30422318

ABSTRACT

MicroRNAs (miRNAs) are closely related with the posttranscriptional regulation of gene expression. Our past study showed that let-7g-5p decreased during pregnancy and lactation in mouse mammary gland, what suggested the prospective regulating role of let-7g-5p in mammary epithelial cells. In this study, to unravel the role of let-7g-5p, we found PRKCA (PKC-alpha, PKC-α) might serve as potential targets of let-7g-5p by bioinformatics. Then let-7g-5p was knocked down by an antisense oligonucleotide in mouse primary mammary epithelial cells. As predicted, PRKCA gene expression and mammary epithelial cells growth were increased by anti-let-7g-5p regulation in vitro. By using reporter constructs, it showed that the let-7g-5p could direct binding with 3'-the untranslated regions (3'-UTR) of PRKCA mRNA. Furthermore, ectopic overexpression of let-7g-5p in vivo reduced PRKCA and ß-casein protein expression as well as inhibited mammary gland growth. These results suggested that let-7g-5p could inhibit the mammary epithelial cells differentiation and ß-casein protein synthesis and expression through suppression of PRKCA on the cycle of mammary cell differentiation and development and that let-7g-5p may be a novel important regulated target in mammary cells function.


Subject(s)
Cell Differentiation/genetics , Gene Expression Regulation/physiology , Mammary Glands, Animal/cytology , MicroRNAs/metabolism , Protein Kinase C-alpha/biosynthesis , Animals , Female , Mammary Glands, Animal/metabolism , Mice , Mice, Inbred BALB C , MicroRNAs/genetics , Protein Kinase C-alpha/genetics
4.
Sci Rep ; 8(1): 6250, 2018 04 19.
Article in English | MEDLINE | ID: mdl-29674689

ABSTRACT

miRNAs play an important role in the processes of cell differentiation, biological development, and physiology. Here we investigated the molecular mechanisms regulating milk secretion and quality in dairy cows via transcriptome analyses of mammary gland tissues from dairy cows during the high-protein/high-fat, low-protein/low-fat or dry periods. To characterize the important roles of miRNAs and mRNAs in milk quality and to elucidate their regulatory networks in relation to milk secretion and quality, an integrated analysis was performed. A total of 25 core miRNAs were found to be differentially expressed (DE) during lactation compared to non-lactation, and these miRNAs were involved in epithelial cell terminal differentiation and mammary gland development. In addition, comprehensive analysis of mRNA and miRNA expression between high-protein/high-fat group and low-protein/low-fat groups indicated that, 38 miRNAs and 944 mRNAs were differentially expressed between them. Furthermore, 38 DE miRNAs putatively negatively regulated 253 DE mRNAs. The putative genes (253 DE mRNAs) were enriched in lipid biosynthetic process and amino acid transmembrane transporter activity. Moreover, putative DE genes were significantly enriched in fatty acid (FA) metabolism, biosynthesis of amino acids, synthesis and degradation of ketone bodies and biosynthesis of unsaturated FAs. Our results suggest that DE miRNAs might play roles as regulators of milk quality and milk secretion during mammary gland differentiation.


Subject(s)
Gene Expression Profiling , MicroRNAs/physiology , Milk/standards , Animals , Cell Differentiation , Dairy Products , Epithelial Cells/cytology , Fatty Acids/biosynthesis , Fatty Acids/metabolism , Lactation/metabolism , Lipogenesis , Milk Proteins/analysis
5.
BMC Vet Res ; 13(1): 350, 2017 Nov 25.
Article in English | MEDLINE | ID: mdl-29178948

ABSTRACT

BACKGROUND: MicroRNAs have important roles in many biological processes. However, the role of miR-139 in healthy mammary gland remains unclear. The objective of this study was to investigate the effects of miR-139 on lactation in dairy cows. RESULTS: Here, we found that miR-139 was down-regulated in mid-lactation dairy cow mammary tissues compared with mid-pregnancy tissues. Then, we prioritized two of potential target genes of miR-139 in cow, growth hormone receptor (GHR) and type I insulin-like growth factor receptor (IGF1R) for further functional studies based on their roles in lactation processes. Dual luciferase reporter assays validated direct binding of miR-139 to the 3'- untranslated region (UTR) of GHR and IGF1R. Moreover, over-expression or silencing of miR-139 affected mRNA levels of GHR and IGF1R in cultured bovine mammary epithelial cells (BMECs). Furthermore, over-expression of miR-139 decreased protein levels of ß-casein, proliferation in mammary epithelial cell, and the protein levels of IGF1R and key members of the GHR or IGF1R pathways as well, whereas silencing miR-139 produced the opposite result. Among these signal molecules, signal transducer and activator of transcription-5 (STAT5), protein kinase B (also known as AKT1), mammalian target of rapamycin (mTOR), and p70-S6 Kinase (p70S6K) are involed in ß-casein synthesis, and Cyclin D1 is involved in cell proliferation. In addition, silencing GHR decreased protein levels of ß-casein, IGF1R, and key members of the IGF1R pathway, whereas co-silencing miR-139 and GHR rescued the expression of GHR and reversed GHR silencing effects. CONCLUSIONS: Our results demonstrate that GHR and IGF1R are target genes of miR-139 in dairy cow. MiR-139 suppresses ß-casein synthesis and proliferation in BMECs by targeting the GHR and IGF1R signaling pathways.


Subject(s)
Caseins/biosynthesis , Mammary Glands, Animal/metabolism , MicroRNAs/genetics , Animals , Cattle , Cell Proliferation/genetics , Cells, Cultured , Epithelial Cells/metabolism , Female , Gene Silencing , Lactation/genetics , Lactation/metabolism , MicroRNAs/metabolism , Pregnancy/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Somatomedin/genetics , Receptors, Somatomedin/metabolism , Receptors, Somatotropin/genetics , Receptors, Somatotropin/metabolism , Signal Transduction
6.
J Dairy Sci ; 100(5): 4014-4024, 2017 May.
Article in English | MEDLINE | ID: mdl-28284693

ABSTRACT

Adequate lipid synthesis by the mammary gland during lactation is essential for the survival of mammalian offspring. Cell death-inducing DNA fragmentation factor-α-like effector C (CIDEC) is a lipid droplet-associated protein and functions to promote lipid accumulation and inhibit lipolysis in mice and human adipocytes. However, the function of CIDEC in regulation of milk lipid synthesis in dairy cow mammary gland remains largely unknown. In this study, 6 multiparous Holstein cows (parity = 3) in early lactation were allocated to high-fat milk (milk yield 33.9 ± 2.1 kg/d, milk fat >3.5%, n = 3) and low-fat milk (milk yield 33.7 ± 0.5 kg/d, milk fat <3.5%, n = 3) groups according to their milk fat content. Lactating cows were slaughtered at 90 d in milk and mammary tissues were collected to detect CIDEC localization. Immunofluorescence staining of sections of lactating mammary glands with high- and low-fat milk showed that CIDEC was expressed in the cytoplasm of epithelial cells and localized to lipid droplets. Lipid droplets and CIDEC protein were also detected in isolated lactating mammary epithelial cells of dairy cows. Immunostaining of CIDEC in isolated mammary epithelial cells also confirmed its presence in the nucleus. The knockdown of CIDEC in cultured bovine mammary epithelial cells decreased milk lipid content and reduced expression of genes associated with mammary de novo fatty acid synthesis, short- and long-chain intracellular fatty acid activation, triacylglycerol synthesis, and transcription regulation. These genes included those for acetyl-CoA carboxylase (ACC, -60%), fatty acid synthase (FASN, -65%), acyl-CoA synthetase short-chain family member 2 (ACSS2, -50%), acyl-CoA synthetase long-chain family member 1 (ACSL1, -30%), diacylglycerol acyltransferase 1 (DGAT1, -60%), sterol regulatory element-binding protein 1 (SREBP1, -45%), and SREBP cleavage activating protein (SCAP, -66%). Conversely, in cells overexpressing CIDEC, triacylglycerol content was increased, and transcription of those genes involved in milk lipid synthesis was coordinately upregulated. These results suggest that CIDEC plays an important role in regulating milk lipid synthesis in dairy cow mammary gland via a mechanism involving gene expression, which provides further insight into the mechanisms regulating mammary lipogenesis in ruminants.


Subject(s)
Lactation , Milk/chemistry , Animals , Cattle , Cell Death , DNA Fragmentation , Fatty Acids , Female , Lipids , Mammary Glands, Animal/metabolism
7.
In Vitro Cell Dev Biol Anim ; 52(10): 1044-1059, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27287918

ABSTRACT

Peroxisome proliferator-activated receptor gamma (PPARγ) participates in lipogenesis in rats, goats, and humans. However, the exact mechanism of PPARγ regulation on milk fat synthesis in dairy cow mammary epithelial cells (DCMECs) remains largely unexplored. The aim of this study was to investigate the role of PPARγ regarding milk fat synthesis in DCMECs and to ascertain whether milk fat precursor acetic acid and palmitic acid could interact with PPARγ signaling to regulate milk fat synthesis. For this study, we examined the effects of PPARγ overexpression and gene silencing on cell growth, triacylglycerol synthesis, and the messenger RNA (mRNA) and protein expression levels of genes involved in milk fat synthesis in DCMECs. In addition, we investigated the influences of acetic acid and palmitic acid on the mRNA and protein levels of milk lipogenic genes and triacylglycerol synthesis in DCMECs transfected with PPARγ small interfering RNA (siRNA) and PPARγ expression vector. The results showed that when PPARγ was silenced, cell viability, proliferation, and triacylglycerol secretion were obviously reduced. Gene silencing of PPARγ significantly downregulated the expression levels of milk fat synthesis-related genes in DCMECs. PPARγ overexpression improved cell viability, proliferation, and triacylglycerol secretion. The expression levels of milk lipogenic genes were significantly increased when PPARγ was overexpressed. Acetic acid and palmitic acid could markedly improve triacylglycerol synthesis and upregulate the expression levels of PPARγ and other lipogenic genes in DCMECs. These results suggest that PPARγ is a positive regulator of milk fat synthesis in DCMECs and that acetic acid and palmitic acid could partly regulate milk fat synthesis in DCMECs via PPARγ signaling.


Subject(s)
Dairying , Epithelial Cells/metabolism , Lipids/biosynthesis , Mammary Glands, Animal/cytology , Milk/chemistry , PPAR gamma/metabolism , Acetic Acid/pharmacology , Animals , Cattle , Cell Proliferation/drug effects , Cells, Cultured , Epithelial Cells/cytology , Epithelial Cells/drug effects , Female , Gene Expression Regulation/drug effects , Gene Knockdown Techniques , Gene Silencing/drug effects , Lipogenesis/drug effects , Lipogenesis/genetics , PPAR gamma/genetics , Palmitic Acid/pharmacology , Triglycerides/metabolism
8.
J Dairy Sci ; 99(7): 5828-5836, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27132107

ABSTRACT

Milk protein is an important component of milk and a nutritional source for human consumption. To better understand the molecular events underlying synthesis of milk proteins, the global gene expression patterns in mammary glands of dairy cow with high-quality milk (>3% milk protein; >3.5% milk fat) and low-quality milk (<3% milk protein; <3.5% milk fat) were examined via digital gene expression study. A total of 139 upregulated and 66 downregulated genes were detected in the mammary tissues of lactating cows with high-quality milk compared with the tissues of cows with low-quality milk. A pathway enrichment study of these genes revealed that the top 5 pathways that were differentially affected in the tissues of cows with high- versus low-quality milk involved metabolic pathways, cancer, cytokine-cytokine receptor interactions, regulation of the actin cytoskeleton, and insulin signaling. We also found that the G protein-coupled receptor kinase 2 (GRK2) was one of the most highly upregulated genes in lactating mammary tissue with low-quality milk compared with tissue with high-quality milk. The knockdown of GRK2 in cultured bovine mammary epithelial cells enhanced CSN2 expression and activated signaling molecules related to translation, including protein kinase B, mammalian target of rapamycin, and p70 ribosomal protein S6 kinase 1 (S6K1), whereas overexpression of GRK2 had the opposite effects. However, expression of genes involved in the mitogen-activated protein kinase pathway was positively regulated by GRK2. Therefore, GRK2 seems to act as a negative mediator of milk-protein synthesis via the protein kinase B-mammalian target of rapamycin signaling axis. Furthermore, GRK2 may negatively control milk-protein synthesis by activating the mitogen-activated protein kinase pathway in dairy cow mammary epithelial cells.


Subject(s)
Lactation , Mammary Glands, Animal/metabolism , Animals , Cattle , Epithelial Cells/metabolism , Female , Humans , Milk/metabolism , Milk Proteins/metabolism
9.
BMC Vet Res ; 12: 81, 2016 May 26.
Article in English | MEDLINE | ID: mdl-27229304

ABSTRACT

BACKGROUND: Lactose, as the primary osmotic component in milk, is the major determinant of milk volume. Glucose is the primary precursor of lactose. However, the effect of glucose on lactose synthesis in dairy cow mammary glands and the mechanism governing this process are poorly understood. RESULTS: Here we showed that glucose has the ability to induce lactose synthesis in dairy cow mammary epithelial cells, as well as increase cell viability and proliferation. A concentration of 12 mM glucose was the optimum concentration to induce cell growth and lactose synthesis in cultured dairy cow mammary epithelial cells. In vitro, 12 mM glucose enhanced lactose content, along with the expression of genes involved in glucose transportation and the lactose biosynthesis pathway, including GLUT1, SLC35A2, SLC35B1, HK2, ß4GalT-I, and AKT1. In addition, we found that AKT1 knockdown inhibited cell growth and lactose synthesis as well as expression of GLUT1, SLC35A2, SLC35B1, HK2, and ß4GalT-I. CONCLUSIONS: Glucose induces cell growth and lactose synthesis in dairy cow mammary epithelial cells. Protein kinase B alpha acts as a regulator of metabolism in dairy cow mammary gland to mediate the effects of glucose on lactose synthesis.


Subject(s)
Glucose/pharmacology , Lactose/biosynthesis , Mammary Glands, Animal/drug effects , Animals , Cattle , Cells, Cultured , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Female , Gene Knockdown Techniques , Mammary Glands, Animal/cytology , Mammary Glands, Animal/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Real-Time Polymerase Chain Reaction/veterinary
10.
Exp Ther Med ; 11(4): 1279-1287, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27073437

ABSTRACT

Our previous study demonstrated that 14-3-3γ overexpression was able to inhibit the production of lipopolysaccharide (LPS)-induced cytokines in dairy cow mammary epithelial cells (DCMECs) by inhibiting the activation of nuclear factor-κB (NF-κB) signaling pathways. However, the association between 14-3-3γ overexpression and milk fat synthesis in LPS-induced DCMECs remains unclear. Therefore, the present study investigated the effect of 14-3-3γ on cell viability and milk fat synthesis in LPS-induced DCMECs. The results of the MTT assay and lactate dehydrogenase activity assay demonstrated that 14-3-3γ overexpression was able to attenuate LPS-induced cytotoxicity in DCMECs, and increase the viability of the cells. In addition, the results of reverse transcription-quantitative polymerase chain reaction suggested that mRNA expression levels of genes associated with milk fat synthesis, including sterol regulatory element binding protein (SREBP1), peroxisome proliferator-activated receptor-γ (PPARG), cluster of differentiation 36, acetyl-coA carboxylase (ACC), fatty acid synthase (FAS) and fatty acid binding protein-3, were significantly upregulated in cells overexpressing the 14-3-3γ protein. In addition, as compared with the LPS-treated group, the activities of FAS and ACC were significantly increased. Furthermore, western blotting demonstrated that 14-3-3γ overexpression enhanced the protein expression levels of phosphorylated SREBP1 and PPARG. These results suggested that high levels of 14-3-3γ protein were able to attenuate LPS-induced cell damage and promote milk fat synthesis in LPS-induced DCMECs by increasing the cell viability and upregulating the expression levels of transcription factors associated with milk fat synthesis.

11.
J Dairy Sci ; 99(5): 3858-3868, 2016 May.
Article in English | MEDLINE | ID: mdl-26947307

ABSTRACT

Spleen tyrosine kinase (SYK) is a nonreceptor tyrosine kinase that has been considered a hematopoietic cell-specific signal transducer involved in cell proliferation and differentiation. However, the role of SYK in normal mammary gland is still poorly understood. Here we show that SYK is expressed in mammary glands of dairy cows. Expression of SYK was higher in dry period mammary tissues than in lactating mammary tissues. Knockdown and overexpression of SYK affected dairy cow mammary epithelial cell proliferation as well as the expression of signal molecules involved in proliferation, including protein kinase B (PKB, also known as AKT1), p42/44 mitogen-activated protein kinase (MAPK), and signal transducer and activator of transcription 5 (STAT5). Dual-luciferase reporter assay showed that SYK increased the transcriptional activity of the AKT1 promoter, and cis-elements within the AKT1 promoter region from -439 to -84 bp mediated this regulation. These results suggest that SYK affects mammary epithelial cell proliferation by activating AKT1 at the transcriptional level in mammary glands of dairy cows, which is important for the mammary remodeling process in dry cows as well as for increasing persistency of lactation in lactating cows.


Subject(s)
Lactation , Mammary Glands, Animal/metabolism , Animals , Cattle , Cell Proliferation , Epithelial Cells/metabolism , Female , Syk Kinase
12.
Int J Mol Sci ; 16(7): 16622-41, 2015 Jul 22.
Article in English | MEDLINE | ID: mdl-26204835

ABSTRACT

As a protective factor for lipopolysaccharide (LPS)-induced injury, 14-3-3γ has been the subject of recent research. Nevertheless, whether 14-3-3γ can regulate lactation in dairy cow mammary epithelial cells (DCMECs) induced by LPS remains unknown. Here, the anti-inflammatory effect and lactation regulating ability of 14-3-3γ in LPS-induced DCMECs are investigated for the first time, and the molecular mechanisms responsible for their effects are explored. The results of qRT-PCR showed that 14-3-3γ overexpression significantly inhibited the mRNA expression of tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), interleukin-1ß (IL-1ß) and inducible nitric oxide synthase (iNOS). Enzyme-linked immunosorbent assay (ELISA) analysis revealed that 14-3-3γ overexpression also suppressed the production of TNF-α and IL-6 in cell culture supernatants. Meanwhile, CASY-TT Analyser System showed that 14-3-3γ overexpression clearly increased the viability and proliferation of cells. The results of kit methods and western blot analysis showed that 14-3-3γ overexpression promoted the secretion of triglycerides and lactose and the synthesis of ß-casein. Furthermore, the expression of genes relevant to nuclear factor-κB (NF-κB) and mitogen-activated protein kinase (MAPKs) and lactation-associated proteins were assessed by western blot, and the results suggested that 14-3-3γ overexpression inactivated the NF-κB and MAPK signaling pathways by down-regulating extracellular signal regulated protein kinase (ERK), p38 mitogen-activated protein kinase (p38MAPK) and inhibitor of NF-κB (IκB) phosphorylation levels, as well as by inhibiting NF-κB translocation. Meanwhile, 14-3-3γ overexpression enhanced the expression levels of ß-casein, mammalian target of rapamycin (mTOR), ribosomal protein S6 kinase 1 (S6K1), serine/threonine protein kinase Akt 1 (AKT1), sterol regulatory element binding protein 1 (SREBP1) and peroxisome proliferator-activated receptor gamma (PPARγ). These results suggest that 14-3-3γ was able to attenuate the LPS-induced inflammatory responses and promote proliferation and lactation in LPS-induced DCMECs by inhibiting the activation of the NF-κB and MAPK signaling pathways and up-regulating mTOR signaling pathways to protect against LPS-induced injury.


Subject(s)
14-3-3 Proteins/metabolism , Caseins/metabolism , Epithelial Cells/metabolism , MAP Kinase Signaling System , Mammary Glands, Animal/metabolism , NF-kappa B/metabolism , TOR Serine-Threonine Kinases/metabolism , 14-3-3 Proteins/genetics , Animals , Caseins/genetics , Cattle , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Interleukins/genetics , Interleukins/metabolism , Lactose/metabolism , Lipopolysaccharides/pharmacology , Mammary Glands, Animal/cytology , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
13.
In Vitro Cell Dev Biol Anim ; 51(7): 697-704, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26183263

ABSTRACT

14-3-3 proteins are an acidic protein family that is highly conserved and widely distributed in eukaryotic cells. Recent studies have found that 14-3-3 proteins play critical roles in cell signal transductions, cell growth and differentiation, and protein synthesis. 14-3-3γ is an important member of 14-3-3 protein family. In our previous study, we found that 14-3-3γ was upregulated by estrogen in dairy cow mammary epithelial cell (DCMEC), but the function and mechanism of 14-3-3γ is not known. In this experiment, we first cultured and purified the primary DCMEC and found 14-3-3γ located both in the cytoplasm and nucleus by using immunofluorescence assay. Methionine, lysine, estrogen, and prolactin could upregulate the expression of 14-3-3γ, stimulate the secretion of ß-casein and triglyceride, and raise the cell viability of DCMEC. We constructed a stable 14-3-3γ overexpression cell line of DCMEC and found that the expressions of mTOR and p-mTOR, the secretion of triglyceride and ß-casein (CSN2), and the cell viability of DCMEC were all upregulated. We also observed the effects of 14-3-3γ gene silencing and gained consistent results with 14-3-3γ overexpression. These findings reveal that 14-3-3γ affects the mTOR pathway and regulates lactogenesis in DCMECs.


Subject(s)
14-3-3 Proteins/metabolism , Mammary Glands, Animal/cytology , TOR Serine-Threonine Kinases/metabolism , 14-3-3 Proteins/genetics , Animals , Caseins/metabolism , Cattle , Cell Survival , Cells, Cultured , Dairying , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Female , Lactation , Lysine/metabolism , Lysine/pharmacology , Mammary Glands, Animal/metabolism , Methionine/metabolism , Methionine/pharmacology , Prolactin/metabolism , Prolactin/pharmacology , Signal Transduction , Triglycerides/metabolism
14.
PLoS One ; 10(3): e0118284, 2015.
Article in English | MEDLINE | ID: mdl-25738494

ABSTRACT

Mammary gland development is controlled by several genes. Although miRNAs have been implicated in mammary gland function, the mechanism by which miR-486 regulates mammary gland development and lactation remains unclear. We investigated miR-486 expression in cow mammary gland using qRT-PCR and ISH and show that miR-486 expression was higher during the high-quality lactation period. We found that miR-486 targets phosphoinositide signaling in the cow mammary gland by directly downregulating PTEN gene expression and by altering the expression of downstream genes that are important for the function of the mammary gland, such as AKT, mTOR. We analyzed the effect of ß-casein, lactose and triglyceride secretion in bovine mammary gland epithelial cells (BMECs) transfected by an inhibitor and by mimics of miR-486. Our results identify miR-486 as a downstream regulator of PTEN that is required for the development of the cow mammary gland.


Subject(s)
Lactation/genetics , Mammary Glands, Animal/metabolism , MicroRNAs/genetics , PTEN Phosphohydrolase/metabolism , Animals , Cattle , Female , Lactation/metabolism , Mammary Glands, Animal/physiology , MicroRNAs/metabolism , PTEN Phosphohydrolase/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
15.
J Cell Physiol ; 230(9): 2152-63, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25656908

ABSTRACT

Milk is important for human nutrition, and enhanced milk quality has become a major selection criterion for the genetic improvement of livestock. Epigenetic modifications have been shown to be involved in mammary gland development; but the mechanisms underlying their effects remain unknown. MicroRNAs are involved in the regulation of milk synthesis and in mammary gland development. Our study is the first to investigate the roles of miR-29s and epigenetic regulation in dairy cow mammary epithelial cells (DCMECs). Our results show that miR-29s regulate the DNA methylation level by inversely targeting both DNMT3A and DNMT3B in DCMECs. The inhibition of miR-29s caused global DNA hypermethylation and increased the methylation levels of the promoters of important lactation-related genes, including casein alpha s1 (CSN1S1), E74-like factor 5 (ElF5), peroxisome proliferator-activated receptor gamma (PPARγ), sterol regulatory element binding protein-1 (SREBP1), and glucose transporter 1 (GLUT1). The inhibition of miR-29s reduced the secretion of lactoprotein, triglycerides (TG) and lactose by DCMECs. Moreover, the treatment of DCMECs with 5-aza-2'-deoxycytidine (5-Aza-dC) decreased the methylation levels of the miR-29b promoter and increased the expression of miR-29b. The link between miR-29s and DNMT3A/3B enhances our understanding of the roles of miRNAs in mammary gland function, and our data will inform more experimentally oriented studies to identify new mechanisms of regulating lactation. We present new insights regarding the epigenetic regulation of lactation performance. Improved understanding of the molecular basis of lactation will aid in the development of strategies for optimizing milk quality in dairy cows and modifying the lactation performance of offspring.


Subject(s)
Epigenesis, Genetic , Mammary Glands, Animal/metabolism , MicroRNAs/metabolism , Milk/metabolism , Animals , Cattle , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation/genetics , DNA Methyltransferase 3A , Female , Gene Expression Regulation , Humans , Lactation , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics
16.
In Vitro Cell Dev Biol Anim ; 51(6): 586-94, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25608868

ABSTRACT

Thyroid hormone responsive spot 14 (THRSP, Spot14, S14) is a nuclear protein that regulates milk fat synthesis. To investigate the role of THRSP in lipogenesis in the dairy cow mammary gland, first, we examined the association between milk fat concentration and THRSP expression in the mammary gland. We found that the dairy cow mammary glands that produced milk with high fat had high THRSP mRNA and protein levels. Additionally, the study described the consequences of overexpression or depletion of THRSP on lipogenesis in cultured bovine mammary epithelial cells (BMECs). We found that BMECs with overexpressed THRSP increased triacylglycerol levels and enhanced the expression of FAS, PPARγ, and SREBP1, compared with the control BMECs. Depletion of THRSP produced the opposite effects. Overall, increased mammary expression of THRSP can be a marker of high fat. In addition, our results provide evidence that THRSP may regulate expression of PPARγ and SREBP1 and can regulate milk fat synthesis by directly affecting the activity of some classical lipogenic enzymes.


Subject(s)
Epithelial Cells/metabolism , Lipogenesis , Mammary Glands, Animal/cytology , Nuclear Proteins/metabolism , Adiposity , Animals , Cattle , Cell Separation , Cells, Cultured , Epithelial Cells/cytology , Female , Milk/metabolism , PPAR gamma/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Triglycerides/metabolism
17.
In Vitro Cell Dev Biol Anim ; 51(3): 287-99, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25319126

ABSTRACT

In dairy cows, the extracellular microenvironment varies significantly from the virgin state to lactation. The function of integrin α6ß4 is dependent on cell type and extracellular microenvironment, and the precise expression profile of α6ß4 and its effects on mammary development remain to be determined. In the present study, real-time PCR and immunohistochemistry were used to analyze the expression and localization of integrin α6ß4 in Holstein dairy cow mammary glands. The effects of integrin α6ß4 on the proliferation induced by mammogenic mitogens were identified by blocking integrin function in purified dairy cow mammary epithelial cells (DCMECs). The results showed that the localization of ß4 subunit and its exclusive partner the α6 subunit were not consistent but were co-localized in basal luminal cells and myoepithelial cells, appearing to prefer the basal surface of the plasma membrane. Moreover, α6 and ß4 subunit messenger RNA (mRNA) levels changed throughout the stages of dairy cow mammary development, reflected well by protein levels, and remained higher in the virgin and pregnancy states, with duct/alveolus morphogenesis and active cell proliferation, than during lactation, when growth arrest is essential for mammary epithelial cell differentiation. Finally, the upregulation of integrin expression by both mammogenic growth hormone and insulin-like growth factor-1 and the inhibited growth of DCMECs by function-blocking integrin antibodies confirmed that integrin α6ß4 was indeed involved in dairy cow mammary development.


Subject(s)
Dairying , Integrin alpha6beta4/genetics , Mammary Glands, Animal/growth & development , Mitogens/pharmacology , Up-Regulation/genetics , Animals , Blotting, Western , Cattle , Cell Proliferation/drug effects , Cell Separation , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental/drug effects , Growth Hormone/pharmacology , Insulin-Like Growth Factor I/pharmacology , Integrin alpha6beta4/metabolism , Laminin/metabolism , Mammary Glands, Animal/cytology , Mammary Glands, Animal/drug effects , Pregnancy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Recombinant Proteins/pharmacology , Up-Regulation/drug effects
18.
Arch Biochem Biophys ; 564: 142-55, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25281768

ABSTRACT

14-3-3γ, an isoform of the 14-3-3 protein family, was proved to be a positive regulator of mTOR pathway. Here, we analyzed the function of 14-3-3γ in protein synthesis using bovine mammary epithelial cells (BMECs). We found that 14-3-3γ interacted with eIF1AX and RPS7 by 14-3-3γ coimmunoprecipitation (CoIP) and matrix-assisted laser desorption/ionization-time-of-flight/time-of-flight (MALDI-TOF/TOF) peptide mass fingerprinting analysis. These interactions of 14-3-3γ with eIF1AX and RPS7 were further confirmed by colocalization and fluorescence resonance energy transfer (FRET) analysis. We also found that methionine could promote protein synthesis and trigger the protein expression levels of 14-3-3γ, eIF1AX and RPS7. Analysis of overexpression and inhibition of 14-3-3γ confirmed that it positively affected the protein expression levels of eIF1AX, RPS7, Stat5 and mTOR pathway to promote protein synthesis and cell proliferation in BMECs. We further showed that overexpression of eIF1AX and RPS7 also triggered protein translation and cell proliferation. From these results, we conclude that molecular network including eIF1AX, RPS7, and 14-3-3γ regulates protein translation and cell proliferation in BMECs.


Subject(s)
14-3-3 Proteins/metabolism , Cell Proliferation/physiology , Epithelial Cells/metabolism , Eukaryotic Initiation Factor-1/metabolism , Mammary Glands, Animal/metabolism , Protein Biosynthesis/physiology , Ribosomal Protein S6/metabolism , 14-3-3 Proteins/genetics , Animals , Cattle , Cells, Cultured , Epithelial Cells/cytology , Eukaryotic Initiation Factor-1/genetics , Female , Gene Expression Regulation/physiology , Humans , Mammary Glands, Animal/cytology , Ribosomal Protein S6/genetics , Surface Plasmon Resonance
19.
Int J Mol Sci ; 15(9): 16998-7013, 2014 Sep 23.
Article in English | MEDLINE | ID: mdl-25250914

ABSTRACT

Sterol regulatory element-binding proteins (SREBPs) belong to a family of nuclear transcription factors. The question of which is the most important positive regulator in milk fat synthesis in dairy cow mammary epithelial cells (DCMECs) between SREBPs or other nuclear transcription factors, such as peroxisome proliferator-activated receptor γ (PPARγ), remains a controversial one. Recent studies have found that mTORC1 (the mammalian target of rapamycin C1) regulates SREBP1 to promote fat synthesis. Thus far, however, the interaction between the SREBP1 and mTOR (the mammalian target of rapamycin) pathways in the regulation of milk fat synthesis remains poorly understood. This study aimed to identify the function of SREBP1 in milk fat synthesis and to characterize the relationship between SREBP1 and mTOR in DCMECs. The effects of SREBP1 overexpression and gene silencing on milk fat synthesis and the effects of stearic acid and serum on SREBP1 expression in the upregulation of milk fat synthesis were investigated in DCMECs using immunostaining, Western blotting, real-time quantitative PCR, lipid droplet staining, and detection kits for triglyceride content. SREBP1 was found to be a positive regulator of milk fat synthesis and was shown to be regulated by stearic acid and serum. These findings indicate that SREBP1 is the key positive regulator in milk fat synthesis.


Subject(s)
Cattle/metabolism , Lipids/biosynthesis , Mammary Glands, Animal/metabolism , Milk/metabolism , Sterol Regulatory Element Binding Protein 1/physiology , Animals , Cells, Cultured , Culture Media, Serum-Free/pharmacology , Epithelial Cells/metabolism , Fatty Acid-Binding Proteins/biosynthesis , Fatty Acid-Binding Proteins/genetics , Female , Milk Proteins/biosynthesis , Milk Proteins/genetics , RNA Interference , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Small Interfering/pharmacology , Recombinant Proteins/metabolism , Serum , Stearic Acids/pharmacology , Sterol Regulatory Element Binding Protein 1/antagonists & inhibitors , Sterol Regulatory Element Binding Protein 1/genetics , TOR Serine-Threonine Kinases/physiology , Transfection , Triglycerides/biosynthesis
20.
Molecules ; 19(7): 9435-52, 2014 Jul 03.
Article in English | MEDLINE | ID: mdl-24995926

ABSTRACT

Glycogen synthase kinase 3 (GSK3) is a serine/threonine kinase, whose activity is inhibited by AKT phosphorylation. This inhibitory phosphorylation of GSK3ß can in turn play a regulatory role through phosphorylation of several proteins (such as mTOR, elF2B) to promote protein synthesis. mTOR is a key regulator in protein synthesis and cell proliferation, and recent studies have shown that both GSK3ß and mTORC1 can regulate SREBP1 to promote fat synthesis. Thus far, however, the cross talk between GSK3ß and the mTOR pathway in the regulation of milk synthesis and associated cell proliferation is not well understood. In this study the interrelationship between GSK3ß and the mTOR/S6K1 signaling pathway leading to milk synthesis and proliferation of dairy cow mammary epithelial cells (DCMECs) was analyzed using techniques including GSK3ß overexpression by transfection, GSK3ß inhibition, mTOR inhibition and methionine stimulation. The analyses revealed that GSK3ß represses the mTOR/S6K1 pathway leading to milk synthesis and cell proliferation of DCMECs, whereas GSK3ß phosphorylation enhances this pathway. Conversely, the activated mTOR/S6K1 signaling pathway downregulates GSK3ß expression but enhances GSK3ß phosphorylation to increase milk synthesis and cell proliferation, whereas inhibition of mTOR leads to upregulation of GSK3ß and repression of GSK3ß phosphorylation, which in turn decreases milk synthesis, and cell proliferation. These findings indicate that GSK3ß and phosphorylated GSK3ß regulate milk synthesis and proliferation of DCMECs via the mTOR/S6K1 signaling pathway. These findings provide new insight into the mechanisms of milk synthesis.


Subject(s)
Epithelial Cells/enzymology , Glycogen Synthase Kinase 3/physiology , Mammary Glands, Animal/cytology , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Cattle , Cell Proliferation , Cells, Cultured , Epithelial Cells/drug effects , Female , Lactation , Lithium Chloride/pharmacology , Methionine/pharmacology , Phosphorylation , Protein Processing, Post-Translational , Signal Transduction , Sirolimus/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...