Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 465
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Int J Neurosci ; 131(1): 15-24, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32098538

RESUMEN

Background: Ischemic stroke is a complex pathological process, involving inflammatory reaction, energy metabolism disorder, free radical injury, cell apoptosis and other aspects. Accumulating evidences have revealed that MFG-E8 had a protective effect on multiple organ injuries. However, the comprehensive function and mechanism of MFG-E8 in ischemic brain remain largely unclear.Methods: BV-2 cells were treated with recombinant murine MFG-E8 (rmMFG-E8) or/and Colivelin TFA after exposing for 4 h with oxygen glucose deprivation (OGD). Cell viability and apoptosis were assessed by MTT assay and Flow cytometry. RT-qPCR and Western blot assays were applied to examine the expression levels of MFG-E8, apoptosis-related proteins and M1/M2 polarization markers.Results: Our results demonstrated that OGD significantly inhibited microglial viability and facilitated apoptosis. In addition, we found that OGD downregulated MFG-E8 expression, and MFG-E8 inhibited OGD-induced microglial apoptosis and promoted microglial M2 polarization. In terms of mechanism, we proved that MFG-E8 regulated OGD-induced microglial M1/M2 polarization by inhibiting p-STAT3 and SOCS3 expressions, which was reversed by STAT3 activator (Colivelin TFA). Finally, we verified MFG-E8 alleviated OGD-induced neuronal cell apoptosis by M2 polarization of BV-2 cells.Conclusions: We demonstrated that MFG-E8 reduced neuronal cell apoptosis by enhancing activation of microglia via STAT3 signaling. Therefore, we suggested that MFG-E8 might provide a novel mechanism for ischemic stroke.


Asunto(s)
Antígenos de Superficie/biosíntesis , Hipoxia de la Célula/fisiología , Glucosa/deficiencia , Microglía/metabolismo , Proteínas de la Leche/biosíntesis , Neuronas/metabolismo , Factor de Transcripción STAT3/biosíntesis , Animales , Apoptosis/fisiología , Línea Celular , Polaridad Celular/fisiología , Técnicas de Cocultivo , Ratones , Proteínas de la Leche/antagonistas & inhibidores
2.
J Dairy Sci ; 103(7): 6218-6232, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32418692

RESUMEN

Amino acids and glucose have been shown to regulate protein synthesis in the mammary gland through their effects on cellular signaling pathways. Acetate might also have an effect on protein synthesis via the AMP-activated kinase signaling pathway, because it is the main energy source for the mammary secretory cell. Thus, the objective of this experiment was to evaluate the effects of casein and energy-yielding nutrients (acetate and glucose), and their combination, on performance and mammary metabolism. Six multiparous Holstein cows, averaging 49 kg of milk/d, were used in a 6 × 6 Latin square design with 14-d periods. Cows were fed to 100% National Research Council requirements for metabolizable protein (MP) and energy (ME) for 9 d, after which they were feed-restricted for 5 d to 85% of their individual ad libitum intake and then abomasally infused with 1 of 6 treatments. Treatments were acetate (A), glucose (G), each at 5% of ad libitum ME intake, casein (C) at 15% of ad libitum MP intake, A + C, G + C, or a saline solution (negative control). Casein infused alone increased milk protein yield numerically, with 25% recovery of the infused casein in milk protein. Glucose infused alone increased milk and milk protein yield and promoted the highest efficiency of nitrogen utilization (37%), with an efficiency of MP use for milk protein of 58%. We discovered no effect of treatment on mammary plasma flow, and the increase in milk protein yield with glucose infusion was brought about by greater mammary AA clearance rate. Infusion of casein and glucose together further increased milk protein yield in an additive fashion, and 47% of the infused casein was recovered in milk protein. Acetate infused alone had no effect on milk protein yield but increased milk fat yield numerically, suggesting that the greater amount of acetate taken up by the mammary gland was used for milk fat synthesis. Infusion of acetate and casein together yielded responses similar to those of casein alone. In conclusion, glucose has a major effect on stimulating milk protein synthesis, and the mammary gland has the ability to increase its supply of nutrients to match its synthetic capacity.


Asunto(s)
Caseínas/administración & dosificación , Bovinos , Glucosa/administración & dosificación , Glándulas Mamarias Animales/metabolismo , Proteínas de la Leche/biosíntesis , Abomaso/metabolismo , Acetatos/análisis , Aminoácidos/metabolismo , Animales , Caseínas/metabolismo , Femenino , Hipersensibilidad a los Alimentos , Tracto Gastrointestinal , Glucosa/metabolismo , Lactancia/fisiología , Glándulas Mamarias Animales/efectos de los fármacos , Leche/química , Proteínas de la Leche/análisis , Biosíntesis de Proteínas
3.
J Dairy Res ; 87(2): 212-219, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32308163

RESUMEN

Ketosis is a metabolic disease of dairy cows often characterized by high concentrations of ketone bodies and fatty acids, but low milk protein and milk production. The Janus kinase 2 (JAK2)-signal transducer and activator of transcription 5 (STAT5) and the mechanistic target of rapamycin (mTOR) signaling pathways are central for the regulation of milk protein synthesis. The effect of high levels of fatty acids on these pathways and ß-casein synthesis are unknown in dairy cows with clinical ketosis. Mammary gland tissue and blood samples were collected from healthy (n = 15) and clinically-ketotic (n = 15) cows. In addition, bovine mammary epithelial cells (BMEC) were treated with fatty acids, methionine (Met) or prolactin (PRL), respectively. In vivo, the serum concentration of fatty acids was greater (P > 0.05) and the percentage of milk protein (P > 0.05) was lower in cows with clinical ketosis. The JAK2-STAT5 and mTOR signaling pathways were inhibited and the abundance of ß-casein was lower in mammary tissue of cows with clinical ketosis (P > 0.05). In vitro, high levels of fatty acids inhibited the JAK2-STAT5 and mTOR signaling pathways (P > 0.05) and further decreased the ß-casein synthesis (P > 0.05) in BMEC. Methionine or PRL treatment, as positive regulators, activated the JAK2-STAT5 and mTOR signaling pathways to increase the ß-casein synthesis. Importantly, the high concentration of fatty acids attenuated the positive effect of Met or PRL on mTOR, JAK2-STAT5 pathways and the abundance of ß-casein (P > 0.05). Overall, these data indicate that the high concentrations of fatty acids that reach the mammary cells during clinical ketosis inhibit mTOR and JAK2-STAT5 signaling pathways, and further suppress ß-casein synthesis.


Asunto(s)
Caseínas/biosíntesis , Enfermedades de los Bovinos/metabolismo , Ácidos Grasos/farmacología , Cetosis/veterinaria , Glándulas Mamarias Animales/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Bovinos , Células Cultivadas , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Ácidos Grasos/sangre , Femenino , Janus Quinasa 2/metabolismo , Cetosis/metabolismo , Metionina/farmacología , Proteínas de la Leche/biosíntesis , Prolactina/farmacología , Factor de Transcripción STAT5/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
4.
Molecules ; 25(6)2020 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-32178391

RESUMEN

The high temperatures used in the production of milk may induce modifications in proteins structure. Due to occurrence of the Maillard reaction, lactose binds lysine residues in proteins, affecting the nutritional value. Milk is also an important source of allergenic proteins (i.e., caseins, ß-lactoglobulin and α-lactalbumin). Thus, this modification may also affect the allergenicity of these proteins. Focusing on milk whey proteins, a screening on different Ultra High Temperatures (UHT) and pasteurized milk samples was performed to identify lactosylation sites, in particular in protein known epitopes, and to verify the correlation between lactosylation and the harshness of the treatment. Whey proteins were extracted from milk samples after caseins precipitations at pH 4.6 and, after chymotryptic and tryptic in solution digestion, peptides were analysed by UPLC-MS and LTQ-Orbitrap. Results show the presence of lactosylated lysine residues in several known epitopes. Then, a ß-lactoglobulin epitope was selected and synthesized by solid phase synthesis followed by in solution lactosylation, obtaining high reaction yields and purities. The synthesis of lactosylated allergenic epitopes, described here for the first time, is a useful tool for further studies on the technological impacts on food allergenicity.


Asunto(s)
Epítopos/genética , Lactoglobulinas/biosíntesis , Proteínas de la Leche/biosíntesis , Proteína de Suero de Leche/biosíntesis , Animales , Caseínas/química , Caseínas/genética , Bovinos , Cromatografía Liquida , Epítopos/inmunología , Calor , Lactalbúmina/química , Lactalbúmina/genética , Lactoglobulinas/química , Lactoglobulinas/genética , Lactoglobulinas/inmunología , Lactosa/química , Reacción de Maillard , Leche/química , Proteínas de la Leche/química , Proteínas de la Leche/genética , Proteínas de la Leche/inmunología , Espectrometría de Masa por Ionización de Electrospray , Espectrometría de Masas en Tándem , Proteína de Suero de Leche/química , Proteína de Suero de Leche/genética , Proteína de Suero de Leche/inmunología
5.
Physiol Genomics ; 51(8): 400-409, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31298615

RESUMEN

Inadequate dry matter intake only partially accounts for the decrease in milk protein synthesis during heat stress (HS) in dairy cows. Our hypothesis is that reduced milk protein synthesis during HS in dairy cows is also caused by biological changes within the mammary gland. The objective of this study was to assess the hypothesis via RNA-Seq analysis of mammary tissue. Herein, four dairy cows were used in a crossover design where HS was induced for 9 days in environmental chambers. There was a 30-day washout between periods. Mammary tissue was collected via biopsy at the end of each environmental period (HS or pair-fed and thermal neutral) for transcriptomic analysis. RNA-Seq analysis revealed HS affected >2,777 genes (false discovery rate-adjusted P value < 0.05) in mammary tissue. Expression of main milk protein-encoding genes and several key genes related to regulation of protein synthesis and amino acid and glucose transport were downregulated by HS. Bioinformatics analysis revealed an overall decrease of mammary tissue metabolic activity by HS (especially carbohydrate and lipid metabolism) and an increase in immune activation and inflammation. Network analysis revealed a major role of TNF, IFNG, S100A8, S100A9, and IGF-1 in inducing/controlling the inflammatory response, with a central role of NF-κB in the process of immunoactivation. The same analysis indicated an overall inhibition of PPARγ. Collectively, these data suggest HS directly controls milk protein synthesis via reducing the transcription of metabolic-related genes and increasing inflammation-related genes.


Asunto(s)
Respuesta al Choque Térmico/fisiología , Glándulas Mamarias Animales/metabolismo , Proteínas de la Leche/biosíntesis , Transcriptoma , Animales , Metabolismo de los Hidratos de Carbono/genética , Bovinos , Estudios Cruzados , Femenino , Inflamación/genética , Metabolismo de los Lípidos/genética , Glándulas Mamarias Animales/inmunología , FN-kappa B/genética , PPAR gamma/genética , RNA-Seq
6.
J Cell Physiol ; 234(5): 6077-6090, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30187485

RESUMEN

Tudor staphylococcal nuclease (Tudor-SN) is a multifunctional protein involved in a variety of cellular processes and plays a critical role in the regulation of gene expression. Recently, Tudor-SN was found to be upregulated in mammary epithelial cells during lactation in response to prolactin, which further to regulate milk protein synthesis. However, the detailed regulatory mechanism of Tudor-SN to milk protein still remains to be elucidated. In our study, we observed that the levels of Tudor-SN and phosphor-Tudor-SN (Thr103) were both enhanced upon prolactin stimulation. Immunofluorescence assays demonstrated that prolactin treatment facilitated the nuclear transport of Tudor-SN. Further study revealed that the phosphorylation of Tudor-SN was depended on activated JNK. Coimmunoprecipitation assays disclosed that Tudor-SN might be phosphorylated directly by JNK. Using gene mutation assays, we further discovered that mutation of Thr to Ala at site of 103 prevented the nuclear transport of Tudor-SN. Thus, these results suggested the essential mechanism of the activated Tudor-SN in milk protein regulation in response to prolactin, which may provide some new sights into improve milk protein production.


Asunto(s)
Células Epiteliales/metabolismo , Lactancia/metabolismo , Nucleasa Microcócica/metabolismo , Proteínas de la Leche/biosíntesis , Prolactina/metabolismo , Animales , Bovinos , Femenino , MAP Quinasa Quinasa 4/metabolismo , Glándulas Mamarias Animales/metabolismo , Fosforilación , Biosíntesis de Proteínas/fisiología , Activación Transcripcional
7.
J Cell Physiol ; 234(2): 1522-1533, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30221364

RESUMEN

MiR-24-3p, a broadly conserved, small, noncoding RNA, is abundantly expressed in mammary tissue. However, its regulatory role in this tissue remains poorly understood. It was predicted that miR-24-3p targets the 3' untranslated region (3'-UTR) of multiple endocrine neoplasia type 1 (MEN1), an important regulatory factor in mammary tissue. The objective of this study was to investigate the function of miR-24-3p in mammary cells. Using a luciferase assay in mammary epithelial cells (MAC-T), miR-24-3p was confirmed to target the 3'-UTR of MEN1. Furthermore, miR-24-3p negatively regulated the expression of the MEN1 gene and its encoded protein, menin. miR-24-3p enhanced proliferation of MAC-T by promoting G1/S phase progression. MiR-24-3p also regulated the expression of key factors involved in phosphatidylinositol-3-kinase/protein kinase B/mammalian target of rapamycin and Janus kinase/signal transducer and activators of transcription signaling pathways, therefore controlling milk protein synthesis in epithelial cells. Thus, miR-24-3p appears to act on MAC-T by targeting MEN1. The expression of miR-24-3p was controlled by MEN1/menin, indicating a negative feedback loop between miR-24-3p and MEN1/menin. The negatively inhibited expression pattern of miR-24-3p and MEN1 was active in mammary tissues at different lactation stages. The feedback mechanism is a new concept to further understand the lactation cycle of mammary glands and can possibly to be manipulated to improve milk yield and quality.


Asunto(s)
Proliferación Celular , Células Epiteliales/metabolismo , Glándulas Mamarias Animales/metabolismo , MicroARNs/metabolismo , Proteínas de la Leche/biosíntesis , Proteínas Proto-Oncogénicas/metabolismo , Regiones no Traducidas 3' , Animales , Sitios de Unión , Bovinos , Línea Celular , Industria Lechera , Femenino , Glándulas Mamarias Animales/citología , MicroARNs/genética , Proteínas Proto-Oncogénicas/genética , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Regulación hacia Arriba
8.
J Cell Physiol ; 234(3): 2973-2983, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30171693

RESUMEN

Amino acids are required for the mammalian target of rapamycin (mTOR) signaling pathway and milk synthesis in bovine mammary epithelial cells (BMECs). However, the mechanism through which amino acids activate this pathway is largely unknown. Here we show that glycyl-tRNA synthetase (GlyRS) mediates amino acid-induced activation of the mTOR-S6K1/4EBP1 pathway, and milk protein and fat synthesis in BMECs. Among 19 aminoacyl-tRNA synthetases, only the mRNA expression of GlyRS and Leucyl-tRNA synthetase (LeuRS) were significantly increased by several amino acids including Met and Leu. We then observed that GlyRS knockdown abolished the stimulation of Met on milk protein and fat synthesis in BMECs, whereas GlyRS overexpression led to more significantly increased milk synthesis in cells treated with Met. By western blotting and qualitative real time-polymerase chain reaction analysis (qRT-PCR) analysis, we next revealed that GlyRS is required for amino acid-induced activation of the mTOR-S6K1/4EBP1 pathway. Thus, this study establishes that GlyRS mediates amino acid-induced activation of the mTOR pathway, thereby regulating milk protein and fat synthesis.


Asunto(s)
Células Epiteliales/metabolismo , Glicina-ARNt Ligasa/genética , Glándulas Mamarias Animales/metabolismo , Leche/metabolismo , Aminoácidos/genética , Aminoácidos/metabolismo , Animales , Bovinos , Femenino , Leucina/metabolismo , Glándulas Mamarias Animales/crecimiento & desarrollo , Metionina/metabolismo , Proteínas de la Leche/biosíntesis , Proteínas Quinasas S6 Ribosómicas 70-kDa , Transducción de Señal , Serina-Treonina Quinasas TOR/genética
9.
Genome ; 62(7): 489-501, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31071269

RESUMEN

A better understanding of the biology of lactation, both in terms of gene expression and the identification of candidate genes for the production of milk and its components, is made possible by recent advances in RNA seq technology. The purpose of this study was to understand the synthesis of milk components and the molecular pathways involved, as well as to identify candidate genes for milk production traits within whole mammary transcriptomic datasets. We performed a meta-analysis of publically available RNA seq transcriptome datasets of mammary tissue/milk somatic cells. In total, 11 562 genes were commonly identified from all RNA seq based mammary gland transcriptomes. Functional annotation of commonly expressed genes revealed the molecular processes that contribute to the synthesis of fats, proteins, and lactose in mammary secretory cells and the molecular pathways responsible for milk synthesis. In addition, we identified several candidate genes responsible for milk production traits and constructed a gene regulatory network for RNA seq data. In conclusion, this study provides a basic understanding of the lactation biology of cows at the gene expression level.


Asunto(s)
Bovinos/genética , Lactancia/genética , Glándulas Mamarias Animales , Transcriptoma , Animales , Femenino , Redes Reguladoras de Genes , Lactosa/biosíntesis , Proteínas de la Leche/biosíntesis , Análisis de Secuencia de ARN
10.
J Dairy Sci ; 102(1): 742-753, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30447981

RESUMEN

An annual pattern of milk composition has been well recognized in dairy cattle, with the highest milk fat and protein concentration observed during the winter and lowest occurring in the summer; however, rhythms of milk yield and composition have not been well quantified. Cosinor rhythmometry is commonly used to model repeating daily and annual rhythms and allows determination of the amplitude (peak to mean), acrophase (time at peak), and period (time between peaks) of the rhythm. The objective of this study was to use cosinor rhythmometry to characterize the annual rhythms of milk yield and milk fat and protein concentration and yield using both national milk market and cow-level data. First, 10 yr of monthly average milk butterfat and protein concentration for each Federal Milk Marketing Order were obtained from the US Department of Agriculture Agricultural Marketing Service database. Fat and protein concentration fit a cosine function with a 12-mo period in all milk markets. We noted an interaction between milk marketing order and milk fat and protein concentration. The acrophase (time at peak) of the fat concentration rhythm ranged from December 4 to January 19 in all regions, whereas the rhythm of protein concentration peaked between December 27 and January 6. The amplitude (peak to mean) of the annual rhythm ranged from 0.07 to 0.14 percentage points for milk fat and from 0.08 to 0.12 percentage points for milk protein. The amplitude of the milk fat rhythm generally was lower in southern markets and higher in northern markets. Second, the annual rhythm of milk yield and milk fat and protein yield and concentration were analyzed in monthly test day data from 1,684 cows from 11 tiestall herds in Pennsylvania. Fat and protein concentration fit an annual rhythm in all herds, whereas milk and milk fat and protein yield only fit rhythms in 8 of the 11 herds. On average, milk yield peaked in April, fat and protein yield peaked in February, fat concentration peaked in January, and protein concentration peaked in December. Amplitudes of milk, fat, and protein yield averaged 0.82 kg, 55.3 g, and 30.4 g, respectively. Milk fat and protein concentration had average amplitudes of 0.12 and 0.07, respectively, similar to the results of the milk market data. Generally, milk yield and milk components fit annual rhythm regardless of parity or diacylglycerol O-acyltransferase 1 (DGAT1) K232A polymorphism, with only cows of the low-frequency AA genotype (5.2% of total cows) failing to fit rhythm of milk yield. In conclusion, the yearly rhythms of milk yield and fat and protein concentration and yield consistently occur regardless of region, herd, parity, or DGAT1 genotype and supports generation by a conserved endogenous annual rhythm.


Asunto(s)
Bovinos/fisiología , Grasas/análisis , Lactancia/fisiología , Proteínas de la Leche/análisis , Leche/química , Estaciones del Año , Animales , Bovinos/genética , Industria Lechera , Diacilglicerol O-Acetiltransferasa/genética , Femenino , Genotipo , Lactancia/genética , Proteínas de la Leche/biosíntesis , Paridad , Pennsylvania , Periodicidad , Polimorfismo Genético , Embarazo , Estados Unidos
11.
J Dairy Sci ; 102(5): 4094-4104, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30827543

RESUMEN

To investigate the possible pathways of Met deficiency to depress milk protein synthesis, 4 lactating goats fitted with jugular vein, mammary vein, and carotid artery catheters and transonic blood flow detectors on the external pudic artery were used in a 4 × 4 Latin square experiment. Goats were fasted for 24 h followed by a 9-h intravenous infusion of an AA mixture plus glucose. Milk yield was recorded and samples were taken in h 2 to 8 of the infusion period, and mammary biopsy was performed in the last hour. Treatments were graded removal of Met from the infused AA mixture to achieve Met content in the infusate of 100 (complete), 60, 30, or 0% of that in casein. Graded Met removal decreased yield of milk, milk protein, and lactose linearly and tended to decrease yield of milk fat linearly. Milk protein yield decreased to 82, 78, and 69% that of complete mixture infusion, respectively, when the 60, 30, and 0% Met infusate was infused. Circulating Met decreased linearly with graded Met removal. Arterial and venous Met decreased to 36 and 23% that of complete mixture infusion, respectively, when all Met was removed out of the mixture. Concomitant with the decreased circulating concentration was a similar increase in mammary Met affinity as reflected by the linearly increased mammary Met clearance rate. The increased affinity plus the linearly increased mammary blood flow totally offset the negative effect of decreased circulating Met concentration on mammary Met uptake. The overall result was similar mammary Met uptakes across treatments ranging from 285.9 to 339.5 µmol/h. Mammary uptakes of the other AA measured were generally not affected by treatments except for a linearly decreased Thr uptake and a trend of linearly increased Glu uptake. Consistent with the behavior of an AA mainly catabolized in the liver and mainly used for protein synthesis in peripheral tissues, mammary uptake to milk output ratios of Met measured in the present study ranged from 1.25 to 1.49 and was not affected by treatments. For the other AA measured, the ratio of Thr was linearly decreased and that of Glu was linearly increased by graded Met removal. Graded Met removal linearly elevated circulating urea N and glucose concentrations, indicating enhanced whole-body catabolism of AA and hepatic gluconeogenesis. Treatments had no significant effects on circulating insulin, growth hormone, and the other hormones and metabolites measured. Phosphorylation status of eIF4E binding protein 1 tended to decrease linearly and that of p70S6k was linearly decreased by graded Met removal, indicating depressed signal in the intracellular mechanistic target of rapamycin complex 1 (mTORC1) signaling pathway. In conclusion, results of the present study indicated that the mTORC1 pathway and whole-body AA catabolism rather than mammary uptake appeared the drivers for changes in milk protein synthesis in response to varying Met supply.


Asunto(s)
Aminoácidos/farmacología , Cabras/metabolismo , Glándulas Mamarias Animales/metabolismo , Metionina/farmacología , Administración Intravenosa , Aminoácidos/administración & dosificación , Aminoácidos/metabolismo , Animales , Caseínas/análisis , Femenino , Glucosa/metabolismo , Insulina/metabolismo , Lactancia , Lactosa/análisis , Metionina/administración & dosificación , Leche/química , Proteínas de la Leche/biosíntesis , Urea/análisis
12.
J Cell Physiol ; 233(3): 2464-2475, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28771726

RESUMEN

Annexin A2 (AnxA2) has been shown to play multiple roles in growth, development, and metabolism, but the functions of AnxA2 and the signaling pathways associated with AnxA2 are still not fully understood. In this study, we aim to reveal whether and how AnxA2 could be involved in milk synthesis and proliferation of bovine mammary epithelial cells (BMECs). Using gene function study approaches, we found that AnxA2 positively regulates PIP3 level, phosphorylation of mTOR, and protein levels of SREBP-1c and Cyclin D1 leading to milk synthesis and cell proliferation. We further observed that both AnxA2-36 kD phosphorylated form and AnxA2-33 kD protein could be induced from AnxA2-36 kD protein in BMECs under methionine, leucine, estrogen or prolactin stimulation. These above results strongly demonstrate that AnxA2 functions as a critical regulator for amino acid or hormone-induced milk synthesis and cell proliferation via the PI3K-mTOR-SREBP-1c/Cyclin D1 signaling pathway.


Asunto(s)
Anexina A2/metabolismo , Proliferación Celular , Células Epiteliales/enzimología , Glándulas Mamarias Humanas/enzimología , Proteínas de la Leche/biosíntesis , Serina-Treonina Quinasas TOR/metabolismo , Animales , Anexina A2/genética , Bovinos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Ciclina D1/metabolismo , Células Epiteliales/efectos de los fármacos , Estrógenos/farmacología , Femenino , Humanos , Leucina/farmacología , Glándulas Mamarias Humanas/efectos de los fármacos , Metionina/farmacología , Fosfatidilinositol 3-Quinasa/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Fosforilación , Progesterona/farmacología , Interferencia de ARN , Transducción de Señal , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Transfección
13.
Ann Neurol ; 82(5): 813-827, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29059709

RESUMEN

OBJECTIVE: Mutations in ABCD1 cause the neurodegenerative disease, adrenoleukodystrophy, which manifests as the spinal cord axonopathy adrenomyeloneuropathy (AMN) in nearly all males surviving into adulthood. Microglial dysfunction has long been implicated in pathogenesis of brain disease, but its role in the spinal cord is unclear. METHODS: We assessed spinal cord microglia in humans and mice with AMN and investigated the role of ABCD1 in microglial activity toward neuronal phagocytosis in cell culture. Because mutations in ABCD1 lead to incorporation of very-long-chain fatty acids into phospholipids, we separately examined the effects of lysophosphatidylcholine (LPC) upon microglia. RESULTS: Within the spinal cord of humans and mice with AMN, upregulation of several phagocytosis-related markers, such as MFGE8 and TREM2, precedes complement activation and synapse loss. Unexpectedly, this occurs in the absence of overt inflammation. LPC C26:0 added to ABCD1-deficient microglia in culture further enhances MFGE8 expression, aggravates phagocytosis, and leads to neuronal injury. Furthermore, exposure to a MFGE8-blocking antibody reduces phagocytic activity. INTERPRETATION: Spinal cord microglia lacking ABCD1 are primed for phagocytosis, affecting neurons within an altered metabolic milieu. Blocking phagocytosis or specific phagocytic receptors may alleviate synapse loss and axonal degeneration. Ann Neurol 2017;82:813-827.


Asunto(s)
Miembro 1 de la Subfamilia D de Transportador de Casetes de Unión al ATP/fisiología , Adrenoleucodistrofia/fisiopatología , Microglía/fisiología , Miembro 1 de la Subfamilia D de Transportador de Casetes de Unión al ATP/genética , Animales , Anticuerpos/inmunología , Antígenos de Superficie/biosíntesis , Antígenos de Superficie/inmunología , Estudios de Casos y Controles , Células Cultivadas , Técnicas de Cocultivo , Expresión Génica/efectos de los fármacos , Humanos , Lisofosfatidilcolinas/farmacología , Glicoproteínas de Membrana/biosíntesis , Ratones Noqueados , Microglía/efectos de los fármacos , Proteínas de la Leche/biosíntesis , Proteínas de la Leche/inmunología , Neuronas/fisiología , Fagocitosis/efectos de los fármacos , Fagocitosis/fisiología , Cultivo Primario de Células , Receptores Inmunológicos/biosíntesis , Médula Espinal/fisiología
14.
J Nutr ; 148(9): 1426-1433, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-30184226

RESUMEN

Background: l-lysine (Lys) is a critical dietary nutrient for mammary gland development and milk production. However, the specific pathways of Lys utilization and how milk protein synthesis is affected in bovine mammary epithelial cells (BMECs) are poorly understood. Objective: We aimed to investigate the effects of Lys on milk protein synthesis and the mechanism of Lys uptake and catabolism in BMECs. Methods: BMECs were cultured in 0, 0.5, 1.0, 1.5, 2.0, 5.0, and 10.0 mmol Lys/L to detect cell viability, or cultured in 0-2.0 mmol Lys/L with l-[ring-3H5] phenylalanine to study the effect of Lys on protein turnover, or cultured in Krebs buffer with [U-14C] l-Lys to quantify Lys metabolism. In some experiments, BMECs were cultured in a conditioned medium alone or including 1.0 mmol Lys/L and 2-amino-endo-bicyclo [2.2.1] heptane-2-carboxylic acid (BCH) for 24 h to analyze the expression of amino acid transporter B (0+) (ATB0,+), mammalian target of rapamycin (mTOR), and Janus kinase 2 (JAK2)-signal transducer and activator of transcription 5 (STAT5) pathways. Results: Including 1.0 mmol Lys/L in cultures increased cell viability by 17-47% and protein synthesis by 7-23%, whereas protein degradation was inhibited by 4-64% compared with BMECs cultured with 0, 0.5, or 2.0 mmol Lys/L (all P ≤ 0.05). Studies that used [U-14C] l-Lys showed that most Lys was incorporated into proteins (90%), whereas the remainder was either oxidized into CO2 (4%) or used as a substrate for aspartate (3%) and histidine synthesis (3%). Furthermore, Lys significantly increased expression of ATB0,+ (71% mRNA and 44% protein), STAT5 (27% mRNA and 21% phosphorylated proteins), and mTOR (51% mRNA and 22% phosphorylated proteins) compared with cells without Lys. Conclusions: Lys promoted protein synthesis, mostly through enhancing uptake by ATB0,+ and the mTOR and JAK2-STAT5 pathways. Understanding the utilization of Lys in BMECs provides insights into the role of amino acid nutrition in bovine milk production.


Asunto(s)
Sistemas de Transporte de Aminoácidos/genética , Bovinos , Lisina/farmacología , Glándulas Mamarias Animales/metabolismo , Proteínas de la Leche/biosíntesis , Serina-Treonina Quinasas TOR/metabolismo , Animales , Caseínas/biosíntesis , Células Cultivadas , Relación Dosis-Respuesta a Droga , Células Epiteliales/metabolismo , Femenino , Expresión Génica/efectos de los fármacos , Lisina/administración & dosificación , Lisina/metabolismo , Proteínas de la Leche/efectos de los fármacos , Proteínas de la Leche/metabolismo , ARN Mensajero/análisis , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/efectos de los fármacos
15.
Amino Acids ; 50(5): 621-628, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29435722

RESUMEN

Milk is an important food for mammalian neonates, but its insufficient production is a nutritional problem for humans and other animals. Recent studies indicate that dietary supplementation with L-arginine (Arg) increases milk production in mammals, including sows, rabbits, and cows. However, the underlying molecular mechanisms remain largely unknown. The present study was conducted with porcine mammary epithelial cells (PMECs) to test the hypothesis that Arg enhances milk protein synthesis via activation of the mechanistic target of rapamycin (mTOR) cell signaling. PMECs were cultured for 4 days in Arg-free basal medium supplemented with 10, 50, 200, or 500 µmol/L Arg. Rates of protein synthesis and degradation in cells were determined with the use of L-[ring-2,4-3H]phenylalanine. Cell medium was analyzed for ß-casein and α-lactalbumin, whereas cells were used for quantifying total and phosphorylated levels of mTOR, ribosomal protein S6 kinase (p70S6K), 4E-binding protein 1 (4EBP1), ubiquitin, and proteasome. Addition of 50-500 µmol/L Arg to culture medium increased (P < 0.05) the proliferation of PMECs and the synthesis of proteins (including ß-casein and α-lactalbumin), while reducing the rates of proteolysis, in a dose-dependent manner. The phosphorylated levels of mTOR, p70S6K and 4EBP1 were elevated (P < 0.05), but the abundances of ubiquitin and proteasome were lower (P < 0.05), in PMECs supplemented with 200-500 µmol/L Arg, compared with 10-50 µmol/L Arg. These results provide a biochemical basis for the use of Arg to enhance milk production by sows and have important implications for improving lactation in other mammals (including humans and cows).


Asunto(s)
Arginina/farmacología , Glándulas Mamarias Animales/metabolismo , Proteínas de la Leche/biosíntesis , Biosíntesis de Proteínas/efectos de los fármacos , Animales , Proteínas Portadoras/metabolismo , Células Cultivadas , Femenino , Glándulas Mamarias Animales/citología , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis/efectos de los fármacos , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Porcinos , Serina-Treonina Quinasas TOR/metabolismo , Ubiquitina/metabolismo
16.
J Dairy Sci ; 101(12): 11150-11158, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30268611

RESUMEN

This research assessed the gene expression patterns related to the synthesis of milk in yak, which is characterized by high fat and protein content but low yield. The yak (Bos grunniens) is one of the most crucial domestic animals in Tibetan life; however, the genetic and molecular factors underlying yak milk protein synthesis remain understudied. Yak mammary biopsies harvested during late-pregnancy (d -15) through the end of subsequent lactation (d 1, 15, 30, 60, 180, and 240) were used to evaluate gene expression via real-time quantitative PCR. The expression pattern of 41 genes encompassing multiple pathways integral to milk protein synthesis including insulin, mammalian target of rapamycin (mTOR), 5' AMP-activated protein kinase, Jak2-Stat5 signaling, and the expression of glucose and AA transporters was evaluated. Our results confirmed that most upregulated genes increased from d -15 and peaked at d 30 or 60 and then remained relatively highly expressed. Specifically, there was an increased expression of mTOR-related amino acid transporters (SLC1A5, SLC7A5, and SLC36A1), glucose transporters (SLC2A1, SLC2A3, and SLC2A8), Jak2-Stat5 pathway (ELF5), and insulin signaling pathway components (IRS1, PDPK1, and AKT1). For activation of proteins synthesis, MTOR was significantly increased only at d 1. Among inhibitors of mTOR signaling, TSC1 and PRKAA2 were significantly upregulated during lactation. The RPL23 was downregulated among ribosomal components. In conclusion, a critical role for AA and glucose transporters and insulin signaling through mTOR for regulation of yak milk protein synthesis was revealed in this study of the yak mammary gland.


Asunto(s)
Bovinos/genética , Bovinos/metabolismo , Proteínas de la Leche/biosíntesis , Leche/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Femenino , Glucosa/metabolismo , Proteínas Facilitadoras del Transporte de la Glucosa/genética , Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Lactancia , Glándulas Mamarias Animales/metabolismo , Embarazo , Biosíntesis de Proteínas , Transducción de Señal , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Transcriptoma
17.
J Dairy Sci ; 101(2): 1687-1696, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29224866

RESUMEN

The mammary gland requires the uptake of AA for milk protein synthesis during lactation. The L-type amino acid transporter 1 (LAT1, encoded by SLC7A5), found in many different types of mammalian cells, is indispensable as a transporter of essential AA to maintain cell growth and protein synthesis. However, the function of LAT1 in regulating milk protein synthesis in the mammary gland of the dairy cow remains largely unknown. For the current study, we characterized the relationship between LAT1 expression and milk protein synthesis in lactating dairy cows and investigated whether the mammalian target of rapamycin complex 1 (mTORC1) signaling controls the expression of LAT1 in their mammary glands. We found that LAT1 and the heavy chain of its chaperone, 4F2, were expressed in mammary tissues of lactating cows, with the expression levels of LAT1 and the 4F2 heavy chain being significantly greater in lactating mammary tissues with high-milk protein content (milk yield, 33.8 ± 2.1 kg/d; milk protein concentration >3%, wt/vol,; n = 3) than in tissues from cows with low-milk protein content (milk yield, 33.7 ± 0.5 kg/d; milk protein concentration <3%, wt/vol; n = 3). Immunofluorescence staining of sectioned mammary tissues from cows with high and low milk protein content showed that LAT1 was located on the whole plasma membrane of alveolar epithelial cells, suggesting that LAT1 provides essential AA to the mammary gland. In cultured mammary epithelial cells from the dairy cows with high-milk protein content, knockdown of LAT1 expression decreased cell viability and ß-casein expression; in contrast, overexpression of LAT1 had the opposite effect. Inhibition of mTORC1 by rapamycin attenuated the phosphorylation of molecules related to mTORC1 signaling and caused a marked decrease in LAT1 expression in the cultured cells; expression of ß-casein also decreased significantly. These results suggest that LAT1 is involved in milk protein synthesis in the mammary glands of lactating dairy cows and that the mTORC1 signaling pathway might be a control point for regulation of LAT1 expression, which could ultimately be used to alter milk protein synthesis.


Asunto(s)
Sistemas de Transporte de Aminoácidos/metabolismo , Bovinos/metabolismo , Glándulas Mamarias Animales/metabolismo , Proteínas de la Leche/biosíntesis , Leche/metabolismo , Biosíntesis de Proteínas , Sistemas de Transporte de Aminoácidos/genética , Animales , Bovinos/genética , Supervivencia Celular/efectos de los fármacos , Femenino , Cadena Pesada de la Proteína-1 Reguladora de Fusión/genética , Cadena Pesada de la Proteína-1 Reguladora de Fusión/metabolismo , Lactancia , Leche/química , Fosforilación
18.
Circulation ; 133(9): 826-39, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26819373

RESUMEN

BACKGROUND: In infarcted heart, improper clearance of dying cells by activated neighboring phagocytes may precipitate the transition to heart failure. We analyzed the coordinated role of 2 major mediators of efferocytosis, the myeloid-epithelial-reproductive protein tyrosine kinase (Mertk) and the milk fat globule epidermal growth factor (Mfge8), in directing cardiac remodeling by skewing the inflammatory response after myocardial infarction. METHODS AND RESULTS: We generated double-deficient mice for Mertk and Mfge8 (Mertk(-/-)/Mfge8(-/-)) and challenged them with acute coronary ligature. Compared with wild-type, Mertk-deficient (Mertk(-/-)), or Mfge8-deficient (Mfge8(-/-)) animals, Mertk(-/-)/Mfge8(-/-) mice displayed greater alteration in cardiac function and remodeling. Mertk and Mfge8 were expressed mainly by cardiac Ly6C(High and Low) monocytes and macrophages. In parallel, Mertk(-/-)/Mfge8(-/-) bone marrow chimeras manifested increased accumulation of apoptotic cells, enhanced fibrotic area, and larger infarct size, as well as reduced angiogenesis. We found that the abrogation of efferocytosis affected neither the ability of circulating monocytes to infiltrate cardiac tissue nor the number of resident Ly6C(High) and Ly6C(How) monocytes/macrophages populating the infarcted milieu. In contrast, combined Mertk and Mfge8 deficiency in Ly6C(High)/Ly6C(Low) monocytes/macrophages either obtained from in vitro differentiation of bone marrow cells or isolated from infarcted hearts altered their capacity of efferocytosis and subsequently blunted vascular endothelial growth factor A (VEGFA) release. Using LysMCre(+)/VEGFA(fl/fl) mice, we further identified an important role for myeloid-derived VEGFA in improving cardiac function and angiogenesis. CONCLUSIONS: After myocardial infarction, Mertk- and Mfge8-expressing monocyte/macrophages synergistically engage the clearance of injured cardiomyocytes, favoring the secretion of VEGFA to locally repair the dysfunctional heart.


Asunto(s)
Antígenos de Superficie/biosíntesis , Proteínas de la Leche/biosíntesis , Infarto del Miocardio/metabolismo , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Factor A de Crecimiento Endotelial Vascular/metabolismo , Remodelación Ventricular/fisiología , Animales , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Infarto del Miocardio/patología , Fagocitosis/fisiología , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Tirosina Quinasas Receptoras/deficiencia , Tirosina Quinasa c-Mer
19.
Tumour Biol ; 39(6): 1010428317707881, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28653875

RESUMEN

Several studies have revealed that MFG-E8 (milk fat globule-epidermal growth factor 8) is related to tumour development and progression. However, the relationship between MFG-E8 expression and metastasis in colorectal cancer patients and the role of MFG-E8 in colorectal cancer invasion and progression remain unknown. In this study, we performed immunohistochemistry and quantitative real-time polymerase chain reaction to assess MFG-E8 expression in colorectal cancer and adjacent non-cancerous tissues. Colorectal cancer RNAseq data from The Cancer Genome Atlas project were downloaded and MFG-E8 expression was analysed. Gene set enrichment analysis was performed for gene ontology and pathway analysis associated with MFG-E8 expression. For in vitro studies, we used lentivirus-mediated MFG-E8 RNA interference and commercialized recombinant human MFG-E8 to investigate its role in colorectal cancer cell growth, migration and invasion. It seems that MFG-E8 was overexpressed in advanced colorectal cancer tissues compared with early-stage colorectal cancer tissues and adjacent non-cancerous tissues. Correlation analysis revealed that MFG-E8 expression was significantly related to plasma membrane invasion, lymph node metastasis, distant metastasis and tumour-node-metastasis stage. Survival analysis revealed that high MFG-E8 expression predicted a poorer prognosis than low MFG-E8 expression group both in our colorectal cancer cohort and The Cancer Genome Atlas colorectal cancer cohort. In vitro study suggested that MFG-E8 knockdown can suppress the growth of colorectal cancer cells without affecting the expression of the proliferation-related gene Ki67. MFG-E8 knockdown also suppressed colorectal cancer cell migration and invasion, a change accompanied by MMP-2 and MMP-9 downregulation. Moreover, MFG-E8 knockdown induced a shift from mesenchymal makers to epithelial makers, while pretreatment with rhMFG-E8 had the opposite effect. The effect of MFG-E8 on colorectal cancer cell migration, invasion and epithelial-to-mesenchymal was partially dependent on the PI3K/AKT signalling pathway. These findings provide a better understanding of the molecular mechanism underlying colorectal cancer progression and suggest a predictive role for MFG-E8 in colorectal cancer metastasis and prognosis.


Asunto(s)
Antígenos de Superficie/biosíntesis , Neoplasias Colorrectales/genética , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/genética , Proteínas de la Leche/biosíntesis , Adulto , Anciano , Antígenos de Superficie/genética , Movimiento Celular/genética , Neoplasias Colorrectales/patología , Progresión de la Enfermedad , Transición Epitelial-Mesenquimal/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Proteínas de la Leche/genética , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Metástasis de la Neoplasia , Proteínas Proto-Oncogénicas c-akt/genética , Transducción de Señal/genética
20.
Eur J Nutr ; 56(8): 2467-2474, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27539583

RESUMEN

PURPOSE: The mechanism of dietary amino acids in regulating milk protein synthesis at the translational level is not well understood. Numerous studies have shown that the amino acid signal is transferred through the mammalian target of rapamycin (mTOR) pathway; however, the extracellular amino acid-sensing mechanism that activates mTOR complex 1 is unknown. We tested the hypotheses that the T1R1/T1R3 heterodimer functions as a direct sensor of the fed state and amino acid availability preceding the mTOR pathway and affects milk protein synthesis in mammary epithelial cells. METHODS: The expression of T1R1 was repressed by T1R1 siRNA in mouse mammary epithelial cells model (HC11). Western blot was used to analyze activity of the mTOR pathway and ß-casein expression, and quantitative real-time RT-PCR was used to analyze the change in mRNA abundance of amino acid transporters. RESULTS: The transcripts and proteins of T1R1 and T1R3 were detected in HC11 cells and mouse mammary gland tissue. siRNA silencing of T1R1 repressed ß-casein synthesis in HC11 cells both with and without essential amino acids present in the culture medium. The phosphorylation of mTOR, S6K, and 4EBP1 in T1R1 knockdown HC11 cells declined to 25, 50, and 30 %, indicating T1R1 knockdown repressed the activity of the mTOR pathway. T1R1 knockdown increased the mRNAs coding three important amino acid transporters (SLC1A5 and SLC3A2/SLC7A5). Activation of the mTOR pathway was partially repressed by T1R1 siRNA or SLC7A5/SLC3A2 inhibitor (BCH, 10 mM), and the combination of these two treatments further repressed the activity of this pathway. CONCLUSION: T1R1/T1R3 serves as sensor of extracellular amino acids in mouse mammary epithelial cells and involved in milk protein synthesis regulation.


Asunto(s)
Aminoácidos/farmacología , Células Epiteliales/metabolismo , Proteínas de la Leche/biosíntesis , Receptores Acoplados a Proteínas G/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Sistema de Transporte de Aminoácidos ASC/antagonistas & inhibidores , Sistema de Transporte de Aminoácidos ASC/genética , Sistema de Transporte de Aminoácidos ASC/metabolismo , Aminoácidos Cíclicos/farmacología , Animales , Caseínas/genética , Caseínas/metabolismo , Línea Celular , Femenino , Cadena Pesada de la Proteína-1 Reguladora de Fusión/antagonistas & inhibidores , Cadena Pesada de la Proteína-1 Reguladora de Fusión/genética , Cadena Pesada de la Proteína-1 Reguladora de Fusión/metabolismo , Silenciador del Gen , Transportador de Aminoácidos Neutros Grandes 1/genética , Transportador de Aminoácidos Neutros Grandes 1/metabolismo , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/efectos de los fármacos , Glándulas Mamarias Animales/metabolismo , Ratones , Antígenos de Histocompatibilidad Menor/genética , Antígenos de Histocompatibilidad Menor/metabolismo , Fosforilación , Biosíntesis de Proteínas , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Receptores Acoplados a Proteínas G/genética , Serina-Treonina Quinasas TOR/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA