Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 1.464
1.
Food Funct ; 12(19): 9432-9442, 2021 Oct 04.
Article En | MEDLINE | ID: mdl-34606535

The microRNA (miR)-432 is differentially expressed in the mammary gland of two breeds of lactating sheep with different milk production traits, and between the non-lactating and peak-lactation periods, but there have been no reports describing the molecular mechanisms involved. In this study, the effect of miR-432 on the proliferation of ovine mammary epithelial cells (OMECs) and the target genes of miR-432 were investigated. The effects of miR-432 on the expression of the target genes and the content of triglycerides in the OMECs were also analyzed. Transfection with a miR-432 mimic was found using CCK8 and Edu assays, to inhibit the viability of OMECs and reduce the number of proliferated OMECs. In contrast, a miR-432 inhibitor had the opposite effect to the miR-432 mimic, and together these results suggest that miR-432 inhibits the proliferation of OMECs. A dual luciferase assay revealed that the genes for stearoyl-CoA desaturase (SCD) and lipoprotein lipase (LPL) are targeted by miR-432. The transfection of miR-432 mimic into OMECs resulted in decreases in the expression of SCD and LPL, and three other milk fat synthesis marker genes; FABP4, LPIN1 and ACACA. The mimic also decreased the content of triglycerides. The miR-432 inhibitor had the opposite effect to the mimic on the expression of these genes and the level of triglycerides. This is the first study to reveal the biological mechanisms by which miR-432 inhibits milk fat synthesis in sheep.


Lipids/biosynthesis , Lipoprotein Lipase/genetics , Mammary Glands, Animal/metabolism , MicroRNAs/genetics , Milk/metabolism , Sheep/metabolism , Stearoyl-CoA Desaturase/genetics , Animals , Cell Proliferation , Cell Survival , Cells, Cultured , Epithelial Cells/metabolism , Female , Gene Expression Regulation , Lipoprotein Lipase/metabolism , Mammary Glands, Animal/cytology , Mammary Glands, Animal/enzymology , MicroRNAs/antagonists & inhibitors , Sheep/genetics , Stearoyl-CoA Desaturase/metabolism , Transfection , Triglycerides/metabolism
2.
Oncol Rep ; 45(5)2021 05.
Article En | MEDLINE | ID: mdl-34184749

In the present study, a rat model of breast hyperplasia was established via the administration of estradiol benzoate and progesterone. Subsequent changes associated with breast hyperplasia were then investigated by measuring the diameter and height of the nipples and by staining breast tissue with hematoxylin and eosin. The proliferation and apoptosis of hyperplastic cells in the breast tissue were then determined by analyzing the expression of proliferating cell nuclear antigen (PCNA) and cleaved­caspase­3 by immunohistochemistry and TUNEL staining. We also determined the expression of proteins associated with the phosphatase and tensin homolog (PTEN)/protein kinase B (AKT) signaling pathway by western blotting. Melatonin treatment led to a significant reduction in the degree of breast hyperplasia (P<0.05), a significant reduction in PCNA, a significant increase in the level of apoptosis (P<0.05), a significant increase in PTEN (P<0.05), and a significant reduction in AKT/p­AKT (P<0.05). Furthermore, melatonin significantly decreased the aggravation of breast hyperplasia induced by application of a PTEN inhibitor. Melatonin reduced the degree of breast hyperplasia, reduced the proliferation of hyperplastic breast tissue cells, and promoted cell apoptosis in hyperplastic tissue. These effects were achieved by the specific regulation of proteins in the PTEN/PI3K/AKT axis.


Mammary Glands, Animal/drug effects , Melatonin/pharmacology , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cell Proliferation/drug effects , Disease Models, Animal , Female , Hyperplasia/drug therapy , Hyperplasia/enzymology , Hyperplasia/pathology , Immunohistochemistry , Mammary Glands, Animal/enzymology , Mammary Glands, Animal/pathology , Proliferating Cell Nuclear Antigen/metabolism , Random Allocation , Rats , Rats, Wistar , Signal Transduction/drug effects
3.
Cells ; 10(3)2021 03 09.
Article En | MEDLINE | ID: mdl-33803196

Bovine intramammary infections are common diseases affecting dairy cattle worldwide and represent a major focus of veterinary research due to financial losses and food safety concerns. The identification of new biomarkers of intramammary infection, useful for monitoring the health of dairy cows and wellness verification, represents a key advancement having potential beneficial effects on public health. In vitro experiments using bovine peripheral blood mononuclear cells (PBMC), stimulated with the bacterial endotoxin lipopolysaccharide (LPS) enabled a flow cytometric assay in order to evaluate in vivo poly-ADP-ribose (PAR) levels. Results showed a significant increase of PAR after 1 h of treatment, which is consistent with the involvement of PARP activity in the inflammatory response. This study investigated PARP-1 activation in leukocyte subpopulations from bovine milk samples during udder infection. A flow cytometric assay was, therefore, performed to evaluate the PAR content in milk leukocyte subsets of cows with and without intramammary infection (IMI). Results showed that milk lymphocytes and macrophages isolated from cows with IMI had a significant increase of PAR content compared to uninfected samples. These results suggest mastitis as a new model for the study of the role of PARP in zoonotic inflammatory diseases, opening a new perspective to the "One Health" approach.


Cattle Diseases/blood , Cattle Diseases/microbiology , Mammary Glands, Animal/enzymology , Mammary Glands, Animal/microbiology , Poly Adenosine Diphosphate Ribose/blood , Poly(ADP-ribose) Polymerases/metabolism , Animals , Biomarkers/blood , Cattle , Enzyme Activation , Female , Flow Cytometry , Leukocytes, Mononuclear , Lipopolysaccharides , Mammary Glands, Animal/pathology , Milk/microbiology
4.
Toxicol Appl Pharmacol ; 416: 115469, 2021 04 01.
Article En | MEDLINE | ID: mdl-33640343

Heat stress-induced oxidative stress in bovine mammary epithelial cells (BMECs) threatens the normal growth and development of bovine mammary tissue, resulting in lower milk production of dairy cows. The aim of the present study is to investigate the protective effects of S-allyl cysteine (SAC), an organosulfur component extracted from aged garlic, on heat stress-induced oxidative stress and apoptosis in BMECs and to explore its underlying mechanisms. Our results showed that heat stress treatment considerably decreased cell viability, whereas SAC treatment dose-dependently restored cell viability of BMECs under heat-stress conditions. In addition, SAC protected BMECs from heat stress-induced oxidative damage by inhibiting the excessive accumulation of reactive oxygen species (ROS) and increasing the activity of antioxidant enzymes. It also inhibited heat stress-induced apoptosis by reducing the ratio of Bax/Bcl-2 and blocking proteolytic the cleavage of caspase-3 in BMECs. Interestingly, we found that the protective effect of SAC on heat stress-induced oxidative stress and apoptosis was dependent on the nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) signaling pathway. SAC promoted the Nrf2 nuclear translocation in heat stress-induced BMECs. The results were also validated by Nrf2 and Keap1 knockdown experiments further demonstrating that Nrf-2 was indeed involved in the protective effect of SAC on heat stress-induced oxidative damage and apoptosis. In summary, our results showed that SAC could protect BMECs from heat stress-induced injury by mediating the Nrf2/HO-1 signaling pathway, suggesting that SAC could be considered as a therapeutic drug for attenuating heat stress-induced mammary gland diseases.


Antioxidants/pharmacology , Cysteine/analogs & derivatives , Epithelial Cells/drug effects , Heat-Shock Response/drug effects , Heme Oxygenase-1/metabolism , Mammary Glands, Animal/drug effects , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Cattle , Cells, Cultured , Cysteine/pharmacology , Epithelial Cells/enzymology , Epithelial Cells/pathology , Female , Kelch-Like ECH-Associated Protein 1/genetics , Kelch-Like ECH-Associated Protein 1/metabolism , Mammary Glands, Animal/enzymology , Mammary Glands, Animal/pathology , Signal Transduction
5.
Physiol Genomics ; 53(2): 61-68, 2021 02 01.
Article En | MEDLINE | ID: mdl-33346690

The p21-activated kinase 1 (PAK1) gene encodes a serine/threonine kinase that is overexpressed in a subset of human breast carcinomas with poor prognosis. The laboratory rat (Rattus norvegicus) orthologous gene is located at Mammary carcinoma susceptibility 3 (Mcs3) QTL on rat chromosome 1. We used quantitative PCR to determine effects of Mcs3 genotype and 7,12-dimethylbenz(a)anthracene (DMBA) exposure on Pak1 expression. There was no effect of Mcs3 genotype; however, there was a 3.5-fold higher Pak1 level in DMBA-exposed mammary glands (MGs) than in unexposed glands (P < 0.05). Sequence variants in Pak1 exons did not alter amino acid sequence between Mcs3-susceptible and -resistant strains. Protein expression of PAK1/Pak1 in human breast carcinomas and DMBA-exposed rat mammary glands was detected using immunohistochemistry (IHC). Rat mammary glands from 12-wk-old females unexposed to DMBA were negative for Pak1, whereas 24% of carcinogen-exposed mammary glands from age-matched females stained positive for Pak1. The positive mammary glands exposed to carcinogen had no pathological signs of disease. Human breast carcinomas, used as comparative controls, had a 22% positivity rats. This was consistent with other human breast cancer studies of PAK1 expression. Similar frequencies of human/rat PAK1/Pak1 expression in female breast carcinomas and carcinogen-induced rat mammary glands, showing no visible pathogenesis of disease, suggests aberrant PAK1 expression is an early event in development of some breast cancers. Laboratory rats will be a useful experimental organism for comparative studies of Pak1-mediated mechanisms of breast carcinogenesis. Future studies of PAK1 as a diagnostic marker of early breast disease are warranted.


9,10-Dimethyl-1,2-benzanthracene/administration & dosage , Breast Neoplasms/metabolism , Carcinogenesis/chemically induced , Carcinogenesis/metabolism , Carcinogens/administration & dosage , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/enzymology , p21-Activated Kinases/metabolism , Animals , Breast Neoplasms/pathology , Disease Models, Animal , Female , Humans , Immunohistochemistry , Mammary Glands, Animal/enzymology , Rats , Rats, Wistar
6.
J Dairy Res ; 87(2): 170-174, 2020 May.
Article En | MEDLINE | ID: mdl-32482199

In this research communication we describe the DGAT1 sequence and promoter region in dairy cows and buffalo and compare the activities of DGAT1 between the two species in order to increase knowledge of the cause of milk fat variation. pGL-3 basic vectors were used to construct the reporter gene. Based on the predicted promoter region, 4 truncated plasmid vectors were constructed in cow-DGAT1 and 3 plasmid vectors in buffalo-DGAT1. Each reporter plasmid was transfected into the bovine mammary epithelial cell (BMEC), 293T cell, and CHO cells to analyze the activity using Dual-Luciferase Reporter Assay System. The results show that the region between -93 to -556 bp was essential for cow promoter activity while -84 to -590 bp was essential for buffalo promoter activity revealing these regions contain core promoter. The buffalo has higher promoter activity than cow yet it was not statistically significant. Comparison of candidate mutation K232A between cow and buffalo population revealed the presence of both the allelic population in dairy cows (lysine and alanine) however, only K (lysine) allelic amino acid was found in buffalo population. The absence of the alanine allelic population from buffalo explains the higher fat content of buffalo milk.


Buffaloes/genetics , Cattle/genetics , Diacylglycerol O-Acyltransferase/genetics , Diacylglycerol O-Acyltransferase/metabolism , Lipids/biosynthesis , Milk/metabolism , Animals , CHO Cells , Cricetulus , Epithelial Cells/enzymology , Female , HEK293 Cells , Humans , Lipids/analysis , Mammary Glands, Animal/enzymology , Milk/chemistry , Promoter Regions, Genetic/genetics , Species Specificity , Transfection
7.
J Cell Biol ; 219(3)2020 03 02.
Article En | MEDLINE | ID: mdl-32211891

Distal appendages (DAs) of the mother centriole are essential for the initial steps of ciliogenesis in G1/G0 phase of the cell cycle. DAs are released from centrosomes in mitosis by an undefined mechanism. Here, we show that specific DAs lose their centrosomal localization at the G2/M transition in a manner that relies upon Nek2 kinase activity to ensure low DA levels at mitotic centrosomes. Overexpression of active Nek2A, but not kinase-dead Nek2A, prematurely displaced DAs from the interphase centrosomes of immortalized retina pigment epithelial (RPE1) cells. This dramatic impact was also observed in mammary epithelial cells with constitutively high levels of Nek2. Conversely, Nek2 knockout led to incomplete dissociation of DAs and cilia in mitosis. As a consequence, we observed the presence of a cilia remnant that promoted the asymmetric inheritance of ciliary signaling components and supported cilium reassembly after cell division. Together, our data establish Nek2 as an important kinase that regulates DAs before mitosis.


Centrioles/enzymology , Cilia/enzymology , Epithelial Cells/enzymology , Mitosis , NIMA-Related Kinases/metabolism , Retinal Pigment Epithelium/enzymology , Animals , Binding Sites , Cell Line , Centrioles/genetics , Cilia/genetics , Female , G2 Phase Cell Cycle Checkpoints , Hematopoietic Stem Cells/enzymology , Humans , Mammary Glands, Animal/cytology , Mammary Glands, Animal/enzymology , Mice , Microtubule Proteins/genetics , Microtubule Proteins/metabolism , NIMA-Related Kinases/genetics , Protein Binding , Retinal Pigment Epithelium/cytology , Signal Transduction , Time Factors
8.
Anim Biotechnol ; 31(3): 276-281, 2020 Jun.
Article En | MEDLINE | ID: mdl-30831051

Aldehyde dehydrogenase 1 (ALDH1) and hepatocyte nuclear factor 4A (HNF4A) are the putative mammary stem cell markers. Tissue necrosis factor alpha (TNFA) is involved in inflammation-associated carcinogenesis and cell proliferation. In this study, the gene expression profile of ALDH1, HNF4A and TNFA of buffalo mammary tissue using real-time quantitative PCR (RT-qPCR). Analysis of RT-qPCR data revealed that the relative expression (log2 fold change) of ALDH1 and TNFA during mastitis (vs. lactation) was increased (P < .05) by 2.98 and 4.71, respectively. The relative expression (log2 fold change; -7.39) of stem cell marker, HNF4A was decreased (P < .05) during mastitis. Histological analysis of mammary tissue during mastitis showed thickening of stroma and occasionally hyperplasia, predominantly in prepubertal and non-lactating animals. Although, the level of expression of these genes may vary, depending upon the physiological stage of the animals, however expression of ALDH1 and TNFA was high during mastitis. A systematic study on large samples of buffalo mammary tissue with appropriate comparisons needs to be evaluated with these markers for prognosis of buffalo mammary health.


Aldehyde Dehydrogenase 1 Family/metabolism , Hepatocyte Nuclear Factor 4/metabolism , Mammary Glands, Animal , Mastitis, Bovine , Tumor Necrosis Factor-alpha/metabolism , Aldehyde Dehydrogenase 1 Family/genetics , Animals , Buffaloes/genetics , Buffaloes/metabolism , Cattle , Hepatocyte Nuclear Factor 4/genetics , Mammary Glands, Animal/chemistry , Mammary Glands, Animal/enzymology , Mammary Glands, Animal/metabolism , Mastitis, Bovine/enzymology , Mastitis, Bovine/metabolism , Tumor Necrosis Factor-alpha/genetics
9.
J Agric Food Chem ; 67(40): 11167-11178, 2019 Oct 09.
Article En | MEDLINE | ID: mdl-31542928

Milk contains a number of beneficial fatty acids including short and medium chain and unsaturated conjugated and nonconjugated fatty acids. In this study, microRNA sequencing of mammary tissue collected in early-, peak-, mid-, and late-lactation periods was performed to determine the miRNA expression profiles. miR-16a was one of the differentially expressed miRNA and was selected for in-depth functional studies pertaining to fatty acid metabolism. The mimic of miR-16a impaired fat metabolism [triacylglycerol (TAG) and cholesterol] while knock-down of miR-16a promoted fat metabolism in vitro in bovine mammary epithelial cells (BMECs). In addition, the in vitro work with BMECs also revealed that miR-16a had a negative effect on the cellular concentration of cis 9-C18:1, total C18:1, C20:1, and C22:1 and long-chain polyunsaturated fatty acids. Therefore, these data suggesting a negative effect on fatty acid metabolism extend the discovery of the key role of miR-16a in mediating adipocyte differentiation. Through a combination of bioinformatics analysis, target gene 3' UTR luciferase reporter assays, and western blotting, we identified large tumor suppressor kinase 1 (LATS1) as a target of miR-16a. Transfection of siRNA-LATS1 into BMECs led to increases in TAG, cholesterol, and cellular fatty acid concentrations, suggesting a positive role of LATS1 in mammary cell fatty acid metabolism. In summary, data suggest that miR-16a regulates biological processes associated with intracellular TAG, cholesterol, and unsaturated fatty acid synthesis through LATS1. These data provide a theoretical and experimental framework for further clarifying the regulation of lipid metabolism in mammary cells of dairy cows.


Cattle/metabolism , Epithelial Cells/enzymology , Lipid Metabolism , Mammary Glands, Animal/enzymology , MicroRNAs/metabolism , Milk/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Cattle/genetics , Cholesterol/metabolism , Epithelial Cells/metabolism , Fatty Acids/metabolism , Female , Gene Expression Regulation , Mammary Glands, Animal/metabolism , MicroRNAs/genetics , Protein Serine-Threonine Kinases/genetics , Triglycerides/metabolism
10.
Biol Reprod ; 101(2): 306-317, 2019 08 01.
Article En | MEDLINE | ID: mdl-31201420

Enhancer of zeste homolog 2 (EZH2) is a rate-limiting catalytic subunit of a histone methyltransferase, polycomb repressive complex, which silences gene activity through the repressive histone mark H3K27me3. EZH2 is critical for epigenetic effects of early estrogen treatment, and may be involved in uterine development and pathologies. We investigated EZH2 expression, regulation, and its role in uterine development/function. Uterine epithelial EZH2 expression was associated with proliferation and was high neonatally then declined by weaning. Pre-weaning uterine EZH2 expression was comparable in wild-type and estrogen receptor 1 knockout mice, showing neonatal EZH2 expression is ESR1 independent. Epithelial EZH2 was upregulated by 17ß-estradiol (E2) and inhibited by progesterone in adult uteri from ovariectomized mice. To investigate the uterine role of EZH2, we developed a EZH2 conditional knockout (Ezh2cKO) mouse using a cre recombinase driven by the progesterone receptor (Pgr) promoter that produced Ezh2cKO mice lacking EZH2 in Pgr-expressing tissues (e.g. uterus, mammary glands). In Ezh2cKO uteri, EZH2 was deleted neonatally. These uteri had reduced H3K27me3, were larger than WT, and showed adult cystic endometrial hyperplasia. Ovary-independent uterine epithelial proliferation and increased numbers of highly proliferative uterine glands were seen in adult Ezh2cKO mice. Female Ezh2cKO mice were initially subfertile, and then became infertile by 9 months. Mammary gland development in Ezh2cKO mice was inhibited. In summary, uterine EZH2 expression is developmentally and hormonally regulated, and its loss causes aberrant uterine epithelial proliferation, uterine hypertrophy, and cystic endometrial hyperplasia, indicating a critical role in uterine development and function.


Enhancer of Zeste Homolog 2 Protein/metabolism , Gene Expression Regulation, Developmental/physiology , Gene Expression Regulation, Enzymologic/physiology , Uterus/enzymology , Uterus/growth & development , Animals , Enhancer of Zeste Homolog 2 Protein/genetics , Epithelial Cells/metabolism , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogens/metabolism , Female , Histones/metabolism , Mammary Glands, Animal/enzymology , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/metabolism , Mice , Mice, Knockout , Pregnancy , Progesterone/metabolism
11.
J Agric Food Chem ; 67(32): 8950-8957, 2019 Aug 14.
Article En | MEDLINE | ID: mdl-31189310

To determine how nutritional restriction compromised milk synthesis, sows were fed 100% (control) or 76% (restricted) of the recommended feed allowance from postpartum day (PD)-1 to PD-28. In comparison to the control, more body reserves loss, increased plasma triglyceride and high-density lipoprotein cholesterol levels, and decreased plasma methionine concentrations were observed in the restricted group at PD-21. The increased plasma malondialdehyde level, decreased plasma histidine and taurine concentrations, and decreased glutathione peroxidase activity were observed at PD-28 when backfat loss further increased in the restricted group. In mammary glands, vacuolar H+-adenosine triphosphatase (v-ATPase), as the upstream of the mechanistic target of rapamycin (mTOR) signaling, showed decreased activity, while phosphorylation of mTOR, S6 kinase, and eukaryotic translation initiation factor 4E-binding protein 1 and ß-casein abundance all decreased following feed restriction. Altogether, long-term nutrition restriction could induce progressively aggravated oxidative stress and compromise mammary protein synthesis through repression of v-ATPase/mTORC1 signaling.


Mammary Glands, Animal/enzymology , Mechanistic Target of Rapamycin Complex 1/metabolism , Oxidative Stress , Protein Biosynthesis , Swine/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , Animals , Epithelial Cells/metabolism , Female , Mammary Glands, Animal/metabolism , Mechanistic Target of Rapamycin Complex 1/genetics , Milk/metabolism , Phosphorylation , Postpartum Period/metabolism , Pregnancy , Signal Transduction , Swine/genetics , Vacuolar Proton-Translocating ATPases/genetics
12.
BMC Vet Res ; 15(1): 155, 2019 May 17.
Article En | MEDLINE | ID: mdl-31101115

BACKGROUND: Sphingosine kinase 1 (SPHK1) is an enzyme that converts pro-apoptotic ceramide and sphingosine into anti-apoptotic sphingosine-1-phosphate. There is growing evidence that SPHK1 activation promotes oncogenic transformation, tumor growth, chemotherapy resistance, and metastatic spread. High SPHK1 expression has been associated with a poor prognosis in several human cancers. RESULTS: In the present study, the expression level of SPHK1 was examined in feline mammary tumor (FMT) specimens, and the IHC expression level of SPHK1 was associated with the histological grade of FMTs. IHC analysis of 88 FMT cases revealed that the expression level of SPHK1 was upregulated in 53 tumor tissues (60.2%) compared to adjacent mammary tissues. SPHK1 expression in FMTs was significantly associated with histological grade, presence of lymphovascular invasion, and estrogen receptor negativity. Treatment of primary FMT cells with SPHK1 inhibitors reduced cell viability, indicating that SPHK1 acts to promote FMT cell survival. These results indicate that SPHK1 may play an important role in FMTs and may be a therapeutic target in cats with FMT. CONCLUSIONS: SPHK1 over-expression in breast cancer tissues is associated with a poor prognosis in humans. SPHK1 over-expression in more aggressive FMTs provides support for a potential role of SPHK1 inhibitors for the treatment of FMTs. Targeting SPHK1 has potent cytotoxic effects in primary FMT cells. These findings suggest that further examination of the role SPHK1 plays in FMTs will pave the way for the investigation of SPHK1 inhibitors in future clinical applications.


Cat Diseases/pathology , Mammary Neoplasms, Animal/enzymology , Mammary Neoplasms, Animal/pathology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Animals , Blood Vessels/pathology , Cat Diseases/enzymology , Cats , Female , Gene Expression Regulation, Neoplastic , Lymphatic System/pathology , Mammary Glands, Animal/enzymology , Mammary Glands, Animal/metabolism , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/metabolism , Neoplasm Invasiveness , Phosphotransferases (Alcohol Group Acceptor)/genetics , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism
13.
J Agric Food Chem ; 67(21): 6007-6018, 2019 May 29.
Article En | MEDLINE | ID: mdl-31060359

4EBP1 is a chief downstream factor of mTORC1, and PPARγ is a key lipogenesis-related transcription factor. mTORC1 and PPARγ are associated with lipid metabolism. However, it is unknown which effector protein connects mTORC1 and PPARγ. This study investigated the interaction between 4EBP1 with PPARγ as part of the underlying mechanism by which insulin-induced lipid synthesis and secretion are regulated by mTORC1 in primary bovine mammary epithelial cells (pBMECs). Rapamycin, a specific inhibitor of mTORC1, downregulated 4EBP1 phosphorylation and the expression of PPARγ and the following lipogenic genes: lipin 1, DGAT1, ACC, and FAS. Rapamycin also decreased the levels of intracellular triacylglycerol (TAG); 10 types of fatty acid; and the accumulation of TAG, palmitic acid (PA), and stearic acid (SA) in the cell culture medium. Inactivation of mTORC1 by shRaptor or shRheb attenuated the synthesis and secretion of TAG and PA. In contrast, activation of mTORC1 by Rheb overexpression promoted 4EBP1 phosphorylation and PPARγ expression and upregulated the mRNA and protein levels of lipin 1, DGAT1, ACC, and FAS, whereas the levels of intracellular and extracellular TAG, PA, and SA also rose. Further, 4EBP1 interacted directly with PPARγ. Inactivation of mTORC1 by shRaptor prevented the nuclear location of PPARγ. These results demonstrate that mTORC1 regulates lipid synthesis and secretion by inducing the expression of lipin 1, DGAT1, ACC, and FAS, which is likely mediated by the 4EBP1/PPARγ axis. This finding constitutes a novel mechanism by which lipid synthesis and secretion are regulated in pBMECs.


Epithelial Cells/metabolism , Eukaryotic Initiation Factors/metabolism , Gene Expression Regulation , Insulin/metabolism , Lipogenesis , Mammary Glands, Animal/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , PPAR gamma/metabolism , Animals , Carbon-Carbon Lyases/genetics , Carbon-Carbon Lyases/metabolism , Cattle , Diacylglycerol O-Acyltransferase/genetics , Diacylglycerol O-Acyltransferase/metabolism , Eukaryotic Initiation Factors/genetics , Female , Mammary Glands, Animal/cytology , Mammary Glands, Animal/enzymology , Mechanistic Target of Rapamycin Complex 1/genetics , PPAR gamma/genetics , Triglycerides/metabolism
14.
Mol Biol Rep ; 46(2): 2243-2257, 2019 Apr.
Article En | MEDLINE | ID: mdl-30759297

MGP-40 is a mammary gland-specific glycoprotein which is expressed during involution and is an important marker for mammary gland apoptosis. It is an inactive chitinase-like protein belonging to Glycosyl Hydrolase family 18. The present study reports sequence characterization, tissue-specific expression analysis, production of recombinant MGP-40 and its mutant (A117D and L119E) in both E. coli and COS1 cells for their chitin-binding and chitinase activity analysis. The cDNA of buffalo MGP-40 was cloned and sequenced which corresponded to 1803 bp with an open reading frame of 1152 bp (361 aa), signal sequence of 63 bp (21 aa), 5' and 3' UTR of 144 bp and 507 bp, respectively. The 3' UTR analysis revealed potential sites for high level expression and stability during involution. The half-life of buffalo MGP-40 was found to be 11.7 h. MGP-40 was highly expressed in mammary gland followed by small intestine, spleen and mammary epithelial cells. The purified recombinant MGP-40 and its mutant expressed in E.coli were observed to bind chitin efficiently, however, no chitinase activity was observed. Further, chitinase activity was also not observed by expressing mutant recombinant MGP-40 in COS1 cells ruling out the possible role of post-translational modifications. Structure-based in-silico mutagenesis by FoldX algorithm showed a drastic decrease in overall fold stability which might be a possible reason for inability to recover its activity. Therefore, chitinase activity could not be restored in MGP-40 even after reverting back two critical residues in active site which may be due to detrimental effect of mutations on structural stability.


Buffaloes/metabolism , Chitinase-3-Like Protein 1/metabolism , Chitinase-3-Like Protein 1/physiology , Amino Acid Sequence , Animals , Apoptosis/physiology , Buffaloes/genetics , Buffaloes/physiology , COS Cells , Chitinase-3-Like Protein 1/genetics , Chitinases/genetics , Chitinases/metabolism , Chlorocebus aethiops , Cloning, Molecular/methods , DNA, Complementary/genetics , Escherichia coli/genetics , Female , Glycoproteins/genetics , Mammary Glands, Animal/enzymology , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/physiology , Open Reading Frames , Protein Sorting Signals , Recombinant Proteins/genetics
15.
J Agric Food Chem ; 67(5): 1463-1469, 2019 Feb 06.
Article En | MEDLINE | ID: mdl-30644742

trans-10, cis-12 Conjugated linoleic acid (t10c12-CLA) is a biohydrogenation intermediate in the rumen that inhibits mammary fatty acid de novo synthesis in lactating dairy goats. However, the underlying molecular pathways in milk-lipid metabolism affected by t10c12-CLA are not completely understood. The present study investigated the lipid-regulation mechanisms in goat mammary epithelial cells (GMECs) in response to t10c12-CLA. Gene-expression analysis indicated sterol-regulatory-element-binding transcription factor1 ( SREBF1) and its putative target gene stearoyl-CoA desaturase ( SCD1) were down-regulated (fold changes of 0.33 ± 0.04, P < 0.05, and 0.19 ± 0.01, P < 0.01, respectively). Concentrations of cellular palmitoleic acid (C16:1) and oleic acid (C18:1) were decreased (1.12 ± 0.05 vs 1.69 ± 0.11% and 15.70 ± 0.44 vs 24.97 ± 0.82%, respectively, P < 0.01), whereas those of linoleic acid (C18:2) were increased (5.00 ± 0.14 vs 3.81 ± 0.25%, P < 0.05); the desaturation indices of C16 and C18 were decreased in response to t10c12-CLA treatment (6.90 ± 0.05 vs 8.00 ± 0.30% and 61.41 ± 0.65 vs 67.73 ± 1.33%, respectively, P < 0.05). A luciferase-activity assay indicated that deletion of the sterol-response-element (SRE) site and the nuclear-factor (NF-Y) site in the SCD1-promoter region (-511/+65 bp) suppressed the regulatory effect of t10c12-CLA. Overexpression of SREBF1 partly counteracted the inhibitory effect of t10c12-CLA on de novo fatty acid synthesis. Overall, t10c12-CLA causes an inhibition of fatty acid synthesis and desaturation and regulates SCD1 expression by affecting the binding of SREBP1 protein to the SRE and NF-Y sites.


Goats/genetics , Linoleic Acids, Conjugated/metabolism , Mammary Glands, Animal/enzymology , Stearoyl-CoA Desaturase/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Animals , Epithelial Cells/enzymology , Epithelial Cells/metabolism , Fatty Acids, Monounsaturated/metabolism , Female , Gene Expression Regulation, Enzymologic , Goats/metabolism , Linoleic Acids, Conjugated/chemistry , Mammary Glands, Animal/metabolism , Promoter Regions, Genetic , Stearoyl-CoA Desaturase/metabolism , Sterol Regulatory Element Binding Protein 1/genetics
16.
J Cell Physiol ; 234(2): 1088-1098, 2019 02.
Article En | MEDLINE | ID: mdl-30203485

Bovine mammary epithelial cells (MAC-Ts) are a common cell line for the study of mammary epithelial inflammation; these cells are used to mechanistically elucidate molecular underpinnings that contribute to bovine mastitis. Bovine mastitis is the most prevalent form of disease in dairy cattle that culminates in annual losses of two billion dollars for the US dairy industry. Thus, there is an urgent need for improved therapeutic strategies. Histone deacetylase (HDAC) inhibitors are efficacious in rodent models of inflammation, yet their role in bovine mammary cells remain unclear. HDACs have traditionally been studied in the regulation of nucleosomal DNA, in which deacetylation of histones impact chromatin accessibility and gene expression. Using MAC-T cells stimulated with tumor necrosis factor α (TNF-α) as a model for mammary cell inflammation, we report that inhibition of HDACs1 and 2 (HDAC1/2) attenuated TNF-α-mediated inflammatory gene expression. Of note, we report that HDAC1/2-mediated inflammatory gene expression was partly regulated by c-Jun N-terminal kinase (JNK) and extracellular signal-regulated kinase (ERK) phosphorylation. Here, we report that HDAC1/2 inhibition attenuated JNK and ERK activation and thus inflammatory gene expression. These data suggest that HDACs1 and 2 regulate inflammatory gene expression via canonical (i.e., gene expression) and noncanonical (e.g., signaling dependent) mechanisms. Whereas, further studies using primary cell lines and animal models are needed. Our combined data suggest that HDAC1/2-specific inhibitors may prove efficacious for the treatment of bovine mastitis.


Epithelial Cells/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Histone Deacetylase 1/metabolism , Histone Deacetylase 2/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Mammary Glands, Animal/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Animals , Anti-Inflammatory Agents/therapeutic use , Cattle , Cell Line , Epithelial Cells/enzymology , Female , Gene Expression Regulation , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase 2/antagonists & inhibitors , Histone Deacetylase Inhibitors/therapeutic use , Mammary Glands, Animal/enzymology , Mastitis, Bovine/drug therapy , Mastitis, Bovine/enzymology , Phosphorylation , Signal Transduction
17.
J Agric Food Chem ; 66(38): 10041-10052, 2018 Sep 26.
Article En | MEDLINE | ID: mdl-30180552

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


Animals, Genetically Modified/genetics , Epithelial Cells/enzymology , Fatty Acids/metabolism , Gene Knockout Techniques/methods , Goats/genetics , Mammary Glands, Animal/enzymology , Stearoyl-CoA Desaturase/deficiency , Animals , Animals, Genetically Modified/metabolism , CRISPR-Cas Systems , Epithelial Cells/metabolism , Female , Gene Deletion , Goats/metabolism , Lipogenesis , Mammary Glands, Animal/cytology , Mammary Glands, Animal/metabolism , Milk/chemistry , Milk/metabolism , Stearoyl-CoA Desaturase/genetics , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Triglycerides/metabolism
18.
Biomed Res Int ; 2018: 1463732, 2018.
Article En | MEDLINE | ID: mdl-29862252

AKT3 gene is a constituent of the serine/threonine protein kinase family and plays a crucial role in synthesis of milk fats and cholesterol by regulating activity of the sterol regulatory element binding protein (SREBP). AKT3 is highly conserved in mammals and its expression levels during the lactation periods of cattle are markedly increased. AKT3 is highly expressed in the intestine followed by mammary gland and it is also expressed in immune cells. It is involved in the TLR pathways as effectively as proinflammatory cytokines. The aims of this study were to investigate the sequences differences between buffalo and cow. Our results showed that there were substantial differences between buffalo and cow in some exons and noteworthy differences of the gene size in different regions. We also identified the important consensus sequence motifs, variation in 2000 upstream of ATG, substantial difference in the "3'UTR" region, and miRNA association in the buffalo sequences compared with the cow. In addition, genetic analyses, such as gene structure, phylogenetic tree, position of different motifs, and functional domains, were performed to establish their correlation with other species. This may indicate that a buffalo breed has potential resistance to disease, environment changes, and airborne microorganisms and some good production and reproductive traits.


3' Untranslated Regions , Buffaloes , Cattle , Gene Expression Regulation, Enzymologic , Mammary Glands, Animal/enzymology , Mastitis, Bovine , Proto-Oncogene Proteins c-akt , Animals , Buffaloes/genetics , Buffaloes/metabolism , Cattle/genetics , Cattle/metabolism , Mastitis, Bovine/enzymology , Mastitis, Bovine/genetics , Proto-Oncogene Proteins c-akt/biosynthesis , Proto-Oncogene Proteins c-akt/genetics , Species Specificity
19.
FASEB J ; 32(6): 3361-3370, 2018 06.
Article En | MEDLINE | ID: mdl-29401621

White adipose tissue inflammation is linked with increased aromatase gene expression and estrogen production, a major risk factor for breast cancer in obese postmenopausal women. TNF-α, a proinflammatory cytokine, is a key driver of aromatase promoter I.4-mediated expression in adipose tissue. In this study, we have shown that IL-10, an anti-inflammatory cytokine, suppressed both TNF-α-stimulated human aromatase reporter-luciferase (hARO-Luc) expression in mouse bone marrow mesenchymal stromal cells and aromatase gene expression in human breast adipose stromal cells (ASCs). IL-10 blocked TNF-α-stimulated ERK1/2 activation in ASCs, suggesting an inhibitory effect through the MAPK signaling pathway. The links among obesity, IL-10, and aromatase were confirmed in ovariectomized (OVX) hARO-Luc mice, where increased adiposity was associated with upregulation of aromatase reporter activity and reduced IL-10 level in the mammary fat pad. OVX mice also exhibited changes in gut microbiota, similar to that in obese women, indicating altered immune function. In summary, our results suggest that increased adiposity, induced by the lack of ovarian hormones, results in enhanced expression of aromatase in mammary adipose tissue, mediated by reduction in local IL-10. These findings may bring new insights into the mechanisms involved in the development of postmenopausal breast cancer, as well as novel approaches for prevention.-Martínez-Chacón, G., Brown, K. A., Docanto, M. M., Kumar, H., Salminen, S., Saarinen, N., Mäkelä, S. IL-10 suppresses TNF-α-induced expression of human aromatase gene in mammary adipose tissue.


Adipose Tissue/enzymology , Aromatase/biosynthesis , Breast/enzymology , Gene Expression Regulation, Enzymologic , Interleukin-10/metabolism , MAP Kinase Signaling System , Tumor Necrosis Factor-alpha/metabolism , Animals , Female , Humans , Mammary Glands, Animal/enzymology , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism
20.
J Dairy Sci ; 101(5): 4586-4594, 2018 May.
Article En | MEDLINE | ID: mdl-29454701

Increased production of long-chain unsaturated fatty acids (LCUFA) can have a positive effect on the nutritional value of ruminant milk for human consumption. In nonruminant species, fatty acid elongase 5 (ELOVL5) is a key enzyme for endogenous synthesis of long-chain unsaturated fatty acids. However, whether ELOVL5 protein plays a role (if any) in ruminant mammary tissue remains unclear. In the present study, we assessed the mRNA abundance of ELOVL5 at 3 stages of lactation in goat mammary tissue. Results revealed that ELOVL5 had the lowest expression at peak lactation compared with the nonlactating and late-lactating periods. The ELOVL5 was overexpressed or knocked down to assess its role in goat mammary epithelial cells. Results revealed that ELOVL5 overexpression increased the expression of perilipin2 (PLIN2) and decreased diacylglycerolacyltransferase 2 (DGAT2) and fatty acid desaturase 2 (FADS2) mRNA, but had no effect on the expression of DGAT1, FADS1, and stearoyl-CoA desaturase 1 (SCD1). Overexpression of ELOVL5 decreased the concentration of C16:1n-7, whereas no significant change in C18:1n-7 and C18:1n-9 was observed. Knockdown of ELOVL5 decreased the expression of PLIN2 but had no effect on DGAT1, DGAT2, FADS1, FADS2, and SCD1 mRNA expression. Knockdown of ELOVL5 increased the concentration of C16:1n-7 and decreased that of C18:1n-7. The alterations of expression of genes related to lipid metabolism after overexpression or knockdown of ELOVL5 suggested a negative feedback regulation by the products of ELOVL5 activation. However, the content of triacylglycerol was not altered by knockdown or overexpression of ELOVL5 in goat mammary epithelial cells, which might have been due to the insufficient availability of substrate in vitro. Collectively, these are the first in vitro results highlighting an important role of ELOVL5 in the elongation of 16-carbon to 18-carbon unsaturated fatty acids in ruminant mammary cells.


Acetyltransferases/metabolism , Epithelial Cells/enzymology , Fatty Acids, Unsaturated/biosynthesis , Goats/metabolism , Mammary Glands, Animal/metabolism , Acetyltransferases/genetics , Animals , Delta-5 Fatty Acid Desaturase , Epithelial Cells/metabolism , Fatty Acid Elongases , Fatty Acids, Unsaturated/chemistry , Female , Goats/genetics , Lactation , Lipid Metabolism , Mammary Glands, Animal/enzymology , Triglycerides/metabolism
...