- -

Molecular networks of insulin signaling and amino acid metabolism in subcutaneous adipose tissue are altered by body condition in periparturient Holstein cows

RiuNet: Repositorio Institucional de la Universidad Politécnica de Valencia

Compartir/Enviar a

Citas

Estadísticas

  • Estadisticas de Uso

Molecular networks of insulin signaling and amino acid metabolism in subcutaneous adipose tissue are altered by body condition in periparturient Holstein cows

Mostrar el registro completo del ítem

Liang, Y.; Alharthi, A.; Elolimy, A.; Bucktrout, R.; Lopreiato, V.; Cortes, I.; Xu, C.... (2020). Molecular networks of insulin signaling and amino acid metabolism in subcutaneous adipose tissue are altered by body condition in periparturient Holstein cows. Journal of Dairy Science. 103(11):10459-10476. https://doi.org/10.3168/jds.2020-18612

Por favor, use este identificador para citar o enlazar este ítem: http://hdl.handle.net/10251/165802

Ficheros en el ítem

Metadatos del ítem

Título: Molecular networks of insulin signaling and amino acid metabolism in subcutaneous adipose tissue are altered by body condition in periparturient Holstein cows
Autor: Liang, Y. Alharthi, A.S. Elolimy, A.A. Bucktrout, R. Lopreiato, V. Cortes, I. Xu, C. Fernández Martínez, Carlos Javier Trevisi, E. Loor, J.J.
Entidad UPV: Universitat Politècnica de València. Departamento de Ciencia Animal - Departament de Ciència Animal
Fecha difusión:
Resumen:
[EN] Peripartal cows mobilize not only body fat but also body protein to satisfy their energy requirements. The objective of this study was to determine the effect of prepartum BCS on blood biomarkers related to energy and ...[+]
Palabras clave: Body condition , Amino acid transporter , Urea cycle , Insulin resistance , Subcutaneous adipose tissue
Derechos de uso: Reconocimiento - No comercial - Sin obra derivada (by-nc-nd)
Fuente:
Journal of Dairy Science. (issn: 0022-0302 )
DOI: 10.3168/jds.2020-18612
Editorial:
American Dairy Science Association
Versión del editor: https://doi.org/10.3168/jds.2020-18612
Agradecimientos:
Y. Liang is a recipient of a doctoral fellowship from the China Scholarship Council (CSC, Beijing, China) to perform his PhD studies at the University of Illinois (Urbana). A. S. Alharthi received a fellowship from King ...[+]
Tipo: Artículo

References

Akter, S. H., Häussler, S., Germeroth, D., von Soosten, D., Dänicke, S., Südekum, K.-H., & Sauerwein, H. (2012). Immunohistochemical characterization of phagocytic immune cell infiltration into different adipose tissue depots of dairy cows during early lactation. Journal of Dairy Science, 95(6), 3032-3044. doi:10.3168/jds.2011-4856

Alharthi, A., Zhou, Z., Lopreiato, V., Trevisi, E., & Loor, J. J. (2018). Body condition score prior to parturition is associated with plasma and adipose tissue biomarkers of lipid metabolism and inflammation in Holstein cows. Journal of Animal Science and Biotechnology, 9(1). doi:10.1186/s40104-017-0221-1

Appuhamy, J. A. D. R. N., Knoebel, N. A., Nayananjalie, W. A. D., Escobar, J., & Hanigan, M. D. (2012). Isoleucine and Leucine Independently Regulate mTOR Signaling and Protein Synthesis in MAC-T Cells and Bovine Mammary Tissue Slices. The Journal of Nutrition, 142(3), 484-491. doi:10.3945/jn.111.152595 [+]
Akter, S. H., Häussler, S., Germeroth, D., von Soosten, D., Dänicke, S., Südekum, K.-H., & Sauerwein, H. (2012). Immunohistochemical characterization of phagocytic immune cell infiltration into different adipose tissue depots of dairy cows during early lactation. Journal of Dairy Science, 95(6), 3032-3044. doi:10.3168/jds.2011-4856

Alharthi, A., Zhou, Z., Lopreiato, V., Trevisi, E., & Loor, J. J. (2018). Body condition score prior to parturition is associated with plasma and adipose tissue biomarkers of lipid metabolism and inflammation in Holstein cows. Journal of Animal Science and Biotechnology, 9(1). doi:10.1186/s40104-017-0221-1

Appuhamy, J. A. D. R. N., Knoebel, N. A., Nayananjalie, W. A. D., Escobar, J., & Hanigan, M. D. (2012). Isoleucine and Leucine Independently Regulate mTOR Signaling and Protein Synthesis in MAC-T Cells and Bovine Mammary Tissue Slices. The Journal of Nutrition, 142(3), 484-491. doi:10.3945/jn.111.152595

Arriarán, S., Agnelli, S., Remesar, X., Fernández-López, J.-A., & Alemany, M. (2015). The urea cycle of rat white adipose tissue. RSC Advances, 5(113), 93403-93414. doi:10.1039/c5ra16398f

Batistel, F., Arroyo, J. M., Bellingeri, A., Wang, L., Saremi, B., Parys, C., … Loor, J. J. (2017). Ethyl-cellulose rumen-protected methionine enhances performance during the periparturient period and early lactation in Holstein dairy cows. Journal of Dairy Science, 100(9), 7455-7467. doi:10.3168/jds.2017-12689

Batistel, F., Arroyo, J. M., Garces, C. I. M., Trevisi, E., Parys, C., Ballou, M. A., … Loor, J. J. (2018). Ethyl-cellulose rumen-protected methionine alleviates inflammation and oxidative stress and improves neutrophil function during the periparturient period and early lactation in Holstein dairy cows. Journal of Dairy Science, 101(1), 480-490. doi:10.3168/jds.2017-13185

Bauchart-Thevret, C., Cui, L., Wu, G., & Burrin, D. G. (2010). Arginine-induced stimulation of protein synthesis and survival in IPEC-J2 cells is mediated by mTOR but not nitric oxide. American Journal of Physiology-Endocrinology and Metabolism, 299(6), E899-E909. doi:10.1152/ajpendo.00068.2010

Bewley, J. M., & Schutz, M. M. (2008). An Interdisciplinary Review of Body Condition Scoring for Dairy Cattle. The Professional Animal Scientist, 24(6), 507-529. doi:10.15232/s1080-7446(15)30901-3

Bionaz, M., Chen, S., Khan, M. J., & Loor, J. J. (2013). Functional Role of PPARs in Ruminants: Potential Targets for Fine-Tuning Metabolism during Growth and Lactation. PPAR Research, 2013, 1-28. doi:10.1155/2013/684159

Bionaz, M., Trevisi, E., Calamari, L., Librandi, F., Ferrari, A., & Bertoni, G. (2007). Plasma Paraoxonase, Health, Inflammatory Conditions, and Liver Function in Transition Dairy Cows. Journal of Dairy Science, 90(4), 1740-1750. doi:10.3168/jds.2006-445

Burgos, S. A., Dai, M., & Cant, J. P. (2010). Nutrient availability and lactogenic hormones regulate mammary protein synthesis through the mammalian target of rapamycin signaling pathway. Journal of Dairy Science, 93(1), 153-161. doi:10.3168/jds.2009-2444

Busato, A., Faissler, D., Kupfer, U., & Blum, J. W. (2002). Body Condition Scores in Dairy Cows: Associations with Metabolic and Endocrine Changes in Healthy Dairy Cows. Journal of Veterinary Medicine Series A, 49(9), 455-460. doi:10.1046/j.1439-0442.2002.00476.x

Cai, H., Dong, L. Q., & Liu, F. (2016). Recent Advances in Adipose mTOR Signaling and Function: Therapeutic Prospects. Trends in Pharmacological Sciences, 37(4), 303-317. doi:10.1016/j.tips.2015.11.011

Caroprese, M., Albenzio, M., Marino, R., Santillo, A., & Sevi, A. (2012). Immune response and milk production of dairy cows fed graded levels of rumen-protected glutamine. Research in Veterinary Science, 93(1), 202-209. doi:10.1016/j.rvsc.2011.07.015

Closs, E. I., Boissel, J.-P., Habermeier, A., & Rotmann, A. (2006). Structure and Function of Cationic Amino Acid Transporters (CATs). Journal of Membrane Biology, 213(2), 67-77. doi:10.1007/s00232-006-0875-7

Contreras, G. A., Kabara, E., Brester, J., Neuder, L., & Kiupel, M. (2015). Macrophage infiltration in the omental and subcutaneous adipose tissues of dairy cows with displaced abomasum. Journal of Dairy Science, 98(9), 6176-6187. doi:10.3168/jds.2015-9370

Cynober, L., Boucher, J. L., & Vasson, M.-P. (1995). Arginine metabolism in mammals. The Journal of Nutritional Biochemistry, 6(8), 402-413. doi:10.1016/0955-2863(95)00066-9

Dann, H. M., Morin, D. E., Bollero, G. A., Murphy, M. R., & Drackley, J. K. (2005). Prepartum Intake, Postpartum Induction of Ketosis, and Periparturient Disorders Affect the Metabolic Status of Dairy Cows. Journal of Dairy Science, 88(9), 3249-3264. doi:10.3168/jds.s0022-0302(05)73008-3

De Koster, J., Hostens, M., Van Eetvelde, M., Hermans, K., Moerman, S., Bogaert, H., … Opsomer, G. (2015). Insulin response of the glucose and fatty acid metabolism in dry dairy cows across a range of body condition scores. Journal of Dairy Science, 98(7), 4580-4592. doi:10.3168/jds.2015-9341

De Koster, J., Strieder-Barboza, C., de Souza, J., Lock, A. L., & Contreras, G. A. (2018). Short communication: Effects of body fat mobilization on macrophage infiltration in adipose tissue of early lactation dairy cows. Journal of Dairy Science, 101(8), 7608-7613. doi:10.3168/jds.2017-14318

De Koster, J., Urh, C., Hostens, M., Van den Broeck, W., Sauerwein, H., & Opsomer, G. (2017). Relationship between serum adiponectin concentration, body condition score, and peripheral tissue insulin response of dairy cows during the dry period. Domestic Animal Endocrinology, 59, 100-104. doi:10.1016/j.domaniend.2016.12.004

De Koster, J., Van den Broeck, W., Hulpio, L., Claeys, E., Van Eetvelde, M., Hermans, K., … Opsomer, G. (2016). Influence of adipocyte size and adipose depot on the in vitro lipolytic activity and insulin sensitivity of adipose tissue in dairy cows at the end of the dry period. Journal of Dairy Science, 99(3), 2319-2328. doi:10.3168/jds.2015-10440

Depreester, E., De Koster, J., Van Poucke, M., Hostens, M., Van den Broeck, W., Peelman, L., … Opsomer, G. (2018). Influence of adipocyte size and adipose depot on the number of adipose tissue macrophages and the expression of adipokines in dairy cows at the end of pregnancy. Journal of Dairy Science, 101(7), 6542-6555. doi:10.3168/jds.2017-13777

Durante, W. (2013). Role of Arginase in Vessel Wall Remodeling. Frontiers in Immunology, 4. doi:10.3389/fimmu.2013.00111

Edmonson, A. J., Lean, I. J., Weaver, L. D., Farver, T., & Webster, G. (1989). A Body Condition Scoring Chart for Holstein Dairy Cows. Journal of Dairy Science, 72(1), 68-78. doi:10.3168/jds.s0022-0302(89)79081-0

Frayn, K. N., Khan, K., Coppack, S. W., & Elia, M. (1991). Amino acid metabolism in human subcutaneous adipose tissue in vivo. Clinical Science, 80(5), 471-474. doi:10.1042/cs0800471

Ghaffari, M. H., Sadri, H., Schuh, K., Dusel, G., Frieten, D., Koch, C., … Sauerwein, H. (2019). Biogenic amines: Concentrations in serum and skeletal muscle from late pregnancy until early lactation in dairy cows with high versus normal body condition score. Journal of Dairy Science, 102(7), 6571-6586. doi:10.3168/jds.2018-16034

Ghaffari, M. H., Schuh, K., Dusel, G., Frieten, D., Koch, C., Prehn, C., … Sadri, H. (2019). Mammalian target of rapamycin signaling and ubiquitin-proteasome–related gene expression in skeletal muscle of dairy cows with high or normal body condition score around calving. Journal of Dairy Science, 102(12), 11544-11560. doi:10.3168/jds.2019-17130

Ghaffari, M. H., Jahanbekam, A., Sadri, H., Schuh, K., Dusel, G., Prehn, C., … Sauerwein, H. (2019). Metabolomics meets machine learning: Longitudinal metabolite profiling in serum of normal versus overconditioned cows and pathway analysis. Journal of Dairy Science, 102(12), 11561-11585. doi:10.3168/jds.2019-17114

Gonzalez, E., & McGraw, T. E. (2009). Insulin-modulated Akt subcellular localization determines Akt isoform-specific signaling. Proceedings of the National Academy of Sciences, 106(17), 7004-7009. doi:10.1073/pnas.0901933106

González, F. D., Muiño, R., Pereira, V., Campos, R., & Benedito, J. L. (2011). Relationship among blood indicators of lipomobilization and hepatic function during early lactation in high-yielding dairy cows. Journal of Veterinary Science, 12(3), 251. doi:10.4142/jvs.2011.12.3.251

Häussler, S., Germeroth, D., Laubenthal, L., Ruda, L. F., Rehage, J., Dänicke, S., & Sauerwein, H. (2017). Short Communication: Immunohistochemical localization of the immune cell marker CD68 in bovine adipose tissue: impact of tissue alterations and excessive fat accumulation in dairy cows. Veterinary Immunology and Immunopathology, 183, 45-48. doi:10.1016/j.vetimm.2016.12.005

Horie, T., Fukasawa, K., Iezaki, T., Park, G., Onishi, Y., Ozaki, K., … Hinoi, E. (2017). Hypoxic Stress Upregulates the Expression of Slc38a1 in Brown Adipocytes via Hypoxia-Inducible Factor-1α. Pharmacology, 101(1-2), 64-71. doi:10.1159/000480405

Hosogai, N., Fukuhara, A., Oshima, K., Miyata, Y., Tanaka, S., Segawa, K., … Shimomura, I. (2007). Adipose Tissue Hypoxia in Obesity and Its Impact on Adipocytokine Dysregulation. Diabetes, 56(4), 901-911. doi:10.2337/db06-0911

Hu, H., Moon, J., Chung, J. H., Kim, O. Y., Yu, R., & Shin, M.-J. (2015). Arginase inhibition ameliorates adipose tissue inflammation in mice with diet-induced obesity. Biochemical and Biophysical Research Communications, 464(3), 840-847. doi:10.1016/j.bbrc.2015.07.048

Ibrahim, M. M. (2010). Subcutaneous and visceral adipose tissue: structural and functional differences. Obesity Reviews, 11(1), 11-18. doi:10.1111/j.1467-789x.2009.00623.x

Jafari, A., Emmanuel, D. G. V., Christopherson, R. J., Thompson, J. R., Murdoch, G. K., Woodward, J., … Ametaj, B. N. (2006). Parenteral Administration of Glutamine Modulates Acute Phase Response in Postparturient Dairy Cows. Journal of Dairy Science, 89(12), 4660-4668. doi:10.3168/jds.s0022-0302(06)72516-4

Jager, J., Grémeaux, T., Cormont, M., Le Marchand-Brustel, Y., & Tanti, J.-F. (2007). Interleukin-1β-Induced Insulin Resistance in Adipocytes through Down-Regulation of Insulin Receptor Substrate-1 Expression. Endocrinology, 148(1), 241-251. doi:10.1210/en.2006-0692

Jamali Emam Gheise, N., Riasi, A., Zare Shahneh, A., Celi, P., & Ghoreishi, S. M. (2017). Effect of pre-calving body condition score and previous lactation on BCS change, blood metabolites, oxidative stress and milk production in Holstein dairy cows. Italian Journal of Animal Science, 16(3), 474-483. doi:10.1080/1828051x.2017.1290507

Janovick, N. A., Boisclair, Y. R., & Drackley, J. K. (2011). Prepartum dietary energy intake affects metabolism and health during the periparturient period in primiparous and multiparous Holstein cows. Journal of Dairy Science, 94(3), 1385-1400. doi:10.3168/jds.2010-3303

Javed, K., & Fairweather, S. J. (2019). Amino acid transporters in the regulation of insulin secretion and signalling. Biochemical Society Transactions, 47(2), 571-590. doi:10.1042/bst20180250

Ji, P., Drackley, J. K., Khan, M. J., & Loor, J. J. (2014). Inflammation- and lipid metabolism-related gene network expression in visceral and subcutaneous adipose depots of Holstein cows. Journal of Dairy Science, 97(6), 3441-3448. doi:10.3168/jds.2013-7296

Ji, P., Drackley, J. K., Khan, M. J., & Loor, J. J. (2014). Overfeeding energy upregulates peroxisome proliferator-activated receptor (PPAR)γ-controlled adipogenic and lipolytic gene networks but does not affect proinflammatory markers in visceral and subcutaneous adipose depots of Holstein cows. Journal of Dairy Science, 97(6), 3431-3440. doi:10.3168/jds.2013-7295

Ji, P., Osorio, J. S., Drackley, J. K., & Loor, J. J. (2012). Overfeeding a moderate energy diet prepartum does not impair bovine subcutaneous adipose tissue insulin signal transduction and induces marked changes in peripartal gene network expression. Journal of Dairy Science, 95(8), 4333-4351. doi:10.3168/jds.2011-5079

Kanda, H. (2006). MCP-1 contributes to macrophage infiltration into adipose tissue, insulin resistance, and hepatic steatosis in obesity. Journal of Clinical Investigation, 116(6), 1494-1505. doi:10.1172/jci26498

Kenéz, Á., Ruda, L., Dänicke, S., & Huber, K. (2019). Insulin signaling and insulin response in subcutaneous and retroperitoneal adipose tissue in Holstein cows during the periparturient period. Journal of Dairy Science, 102(12), 11718-11729. doi:10.3168/jds.2019-16873

Laplante, M., & Sabatini, D. M. (2009). mTOR signaling at a glance. Journal of Cell Science, 122(20), 3589-3594. doi:10.1242/jcs.051011

Laubenthal, L., Ruda, L., Sultana, N., Winkler, J., Rehage, J., Meyer, U., … Häussler, S. (2017). Effect of increasing body condition on oxidative stress and mitochondrial biogenesis in subcutaneous adipose tissue depot of nonlactating dairy cows. Journal of Dairy Science, 100(6), 4976-4986. doi:10.3168/jds.2016-12356

Le Floc’h, N., Melchior, D., & Obled, C. (2004). Modifications of protein and amino acid metabolism during inflammation and immune system activation. Livestock Production Science, 87(1), 37-45. doi:10.1016/j.livprodsci.2003.09.005

Li, Y., Wei, H., Li, F., Chen, S., Duan, Y., Guo, Q., … Yin, Y. (2016). Supplementation of branched-chain amino acids in protein-restricted diets modulates the expression levels of amino acid transporters and energy metabolism associated regulators in the adipose tissue of growing pigs. Animal Nutrition, 2(1), 24-32. doi:10.1016/j.aninu.2016.01.003

Liang, Y., Alharthi, A. S., Bucktrout, R., Elolimy, A. A., Lopreiato, V., Martinez-Cortés, I., … Loor, J. J. (2020). Body condition alters glutathione and nuclear factor erythroid 2-like 2 (NFE2L2)–related antioxidant network abundance in subcutaneous adipose tissue of periparturient Holstein cows. Journal of Dairy Science, 103(7), 6439-6453. doi:10.3168/jds.2019-17813

Liang, Y., Batistel, F., Parys, C., & Loor, J. J. (2019). Methionine supply during the periparturient period enhances insulin signaling, amino acid transporters, and mechanistic target of rapamycin pathway proteins in adipose tissue of Holstein cows. Journal of Dairy Science, 102(5), 4403-4414. doi:10.3168/jds.2018-15738

Liao, W., Nguyen, M. T. A., Yoshizaki, T., Favelyukis, S., Patsouris, D., Imamura, T., … Olefsky, J. M. (2007). Suppression of PPAR-γ attenuates insulin-stimulated glucose uptake by affecting both GLUT1 and GLUT4 in 3T3-L1 adipocytes. American Journal of Physiology-Endocrinology and Metabolism, 293(1), E219-E227. doi:10.1152/ajpendo.00695.2006

Locher, L. F., Meyer, N., Weber, E.-M., Rehage, J., Meyer, U., Dänicke, S., & Huber, K. (2011). Hormone-sensitive lipase protein expression and extent of phosphorylation in subcutaneous and retroperitoneal adipose tissues in the periparturient dairy cow. Journal of Dairy Science, 94(9), 4514-4523. doi:10.3168/jds.2011-4145

Ma, Y. F., Zhao, L., Coleman, D. N., Gao, M., & Loor, J. J. (2019). Tea polyphenols protect bovine mammary epithelial cells from hydrogen peroxide-induced oxidative damage in vitro by activating NFE2L2/HMOX1 pathways. Journal of Dairy Science, 102(2), 1658-1670. doi:10.3168/jds.2018-15047

Mackenzie, B., & Erickson, J. D. (2004). Sodium-coupled neutral amino acid (System N/A) transporters of the SLC38 gene family. Pfl�gers Archiv European Journal of Physiology, 447(5), 784-795. doi:10.1007/s00424-003-1117-9

Mann, S., Nydam, D. V., Abuelo, A., Leal Yepes, F. A., Overton, T. R., & Wakshlag, J. J. (2016). Insulin signaling, inflammation, and lipolysis in subcutaneous adipose tissue of transition dairy cows either overfed energy during the prepartum period or fed a controlled-energy diet. Journal of Dairy Science, 99(8), 6737-6752. doi:10.3168/jds.2016-10969

Megahed, A. A., Hiew, M. W. H., Ragland, D., & Constable, P. D. (2019). Changes in skeletal muscle thickness and echogenicity and plasma creatinine concentration as indicators of protein and intramuscular fat mobilization in periparturient dairy cows. Journal of Dairy Science, 102(6), 5550-5565. doi:10.3168/jds.2018-15063

Menchini, R. J., & Chaudhry, F. A. (2019). Multifaceted regulation of the system A transporter Slc38a2 suggests nanoscale regulation of amino acid metabolism and cellular signaling. Neuropharmacology, 161, 107789. doi:10.1016/j.neuropharm.2019.107789

Minuti, A., Bionaz, M., Lopreiato, V., Janovick, N. A., Rodriguez-Zas, S. L., Drackley, J. K., & Loor, J. J. (2020). Prepartum dietary energy intake alters adipose tissue transcriptome profiles during the periparturient period in Holstein dairy cows. Journal of Animal Science and Biotechnology, 11(1). doi:10.1186/s40104-019-0409-7

Moisá, S. J., Ji, P., Drackley, J. K., Rodriguez-Zas, S. L., & Loor, J. J. (2017). Transcriptional changes in mesenteric and subcutaneous adipose tissue from Holstein cows in response to plane of dietary energy. Journal of Animal Science and Biotechnology, 8(1). doi:10.1186/s40104-017-0215-z

Moyes, K. M., Drackley, J. K., Morin, D. E., & Loor, J. J. (2010). Greater expression of TLR2, TLR4, and IL6 due to negative energy balance is associated with lower expression of HLA-DRA and HLA-A in bovine blood neutrophils after intramammary mastitis challenge with Streptococcus uberis. Functional & Integrative Genomics, 10(1), 53-61. doi:10.1007/s10142-009-0154-7

Mukesh, M., Bionaz, M., Graugnard, D. E., Drackley, J. K., & Loor, J. J. (2010). Adipose tissue depots of Holstein cows are immune responsive: Inflammatory gene expression in vitro. Domestic Animal Endocrinology, 38(3), 168-178. doi:10.1016/j.domaniend.2009.10.001

Newgard, C. B. (2017). Metabolomics and Metabolic Diseases: Where Do We Stand? Cell Metabolism, 25(1), 43-56. doi:10.1016/j.cmet.2016.09.018

Newgard, C. B., An, J., Bain, J. R., Muehlbauer, M. J., Stevens, R. D., Lien, L. F., … Svetkey, L. P. (2009). A Branched-Chain Amino Acid-Related Metabolic Signature that Differentiates Obese and Lean Humans and Contributes to Insulin Resistance. Cell Metabolism, 9(4), 311-326. doi:10.1016/j.cmet.2009.02.002

Newman, A. W., Miller, A., Leal Yepes, F. A., Bitsko, E., Nydam, D., & Mann, S. (2019). The effect of the transition period and postpartum body weight loss on macrophage infiltrates in bovine subcutaneous adipose tissue. Journal of Dairy Science, 102(2), 1693-1701. doi:10.3168/jds.2018-15362

Newsholme, P., Procopio, J., Lima, M. M. R., Pithon-Curi, T. C., & Curi, R. (2003). Glutamine and glutamate?their central role in cell metabolism and function. Cell Biochemistry and Function, 21(1), 1-9. doi:10.1002/cbf.1003

Osorio, J. S., Ji, P., Drackley, J. K., Luchini, D., & Loor, J. J. (2014). Smartamine M and MetaSmart supplementation during the peripartal period alter hepatic expression of gene networks in 1-carbon metabolism, inflammation, oxidative stress, and the growth hormone–insulin-like growth factor 1 axis pathways. Journal of Dairy Science, 97(12), 7451-7464. doi:10.3168/jds.2014-8680

Palmieri, E. M., Spera, I., Menga, A., Infantino, V., Iacobazzi, V., & Castegna, A. (2014). Glutamine synthetase desensitizes differentiated adipocytes to proinflammatory stimuli by raising intracellular glutamine levels. FEBS Letters, 588(24), 4807-4814. doi:10.1016/j.febslet.2014.11.015

Petrus, P., Lecoutre, S., Dollet, L., Wiel, C., Sulen, A., Gao, H., … Rydén, M. (2020). Glutamine Links Obesity to Inflammation in Human White Adipose Tissue. Cell Metabolism, 31(2), 375-390.e11. doi:10.1016/j.cmet.2019.11.019

Pires, J. A. A., Delavaud, C., Faulconnier, Y., Pomiès, D., & Chilliard, Y. (2013). Effects of body condition score at calving on indicators of fat and protein mobilization of periparturient Holstein-Friesian cows. Journal of Dairy Science, 96(10), 6423-6439. doi:10.3168/jds.2013-6801

Rico, J. E., Myers, W. A., Laub, D. J., Davis, A. N., Zeng, Q., & McFadden, J. W. (2018). Hot topic: Ceramide inhibits insulin sensitivity in primary bovine adipocytes. Journal of Dairy Science, 101(4), 3428-3432. doi:10.3168/jds.2017-13983

Rico, J. E., Bandaru, V. V. R., Dorskind, J. M., Haughey, N. J., & McFadden, J. W. (2015). Plasma ceramides are elevated in overweight Holstein dairy cows experiencing greater lipolysis and insulin resistance during the transition from late pregnancy to early lactation. Journal of Dairy Science, 98(11), 7757-7770. doi:10.3168/jds.2015-9519

Rius, A. G., Appuhamy, J. A. D. R. N., Cyriac, J., Kirovski, D., Becvar, O., Escobar, J., … Hanigan, M. D. (2010). Regulation of protein synthesis in mammary glands of lactating dairy cows by starch and amino acids. Journal of Dairy Science, 93(7), 3114-3127. doi:10.3168/jds.2009-2743

Roche, J. R., Kay, J. K., Friggens, N. C., Loor, J. J., & Berry, D. P. (2013). Assessing and Managing Body Condition Score for the Prevention of Metabolic Disease in Dairy Cows. Veterinary Clinics of North America: Food Animal Practice, 29(2), 323-336. doi:10.1016/j.cvfa.2013.03.003

Ruan, H., & Dong, L. Q. (2016). Adiponectin signaling and function in insulin target tissues. Journal of Molecular Cell Biology, 8(2), 101-109. doi:10.1093/jmcb/mjw014

Saltiel, A. R., & Kahn, C. R. (2001). Insulin signalling and the regulation of glucose and lipid metabolism. Nature, 414(6865), 799-806. doi:10.1038/414799a

Saremi, B., Al-Dawood, A., Winand, S., Müller, U., Pappritz, J., von Soosten, D., … Sauerwein, H. (2012). Bovine haptoglobin as an adipokine: Serum concentrations and tissue expression in dairy cows receiving a conjugated linoleic acids supplement throughout lactation. Veterinary Immunology and Immunopathology, 146(3-4), 201-211. doi:10.1016/j.vetimm.2012.03.011

Schuh, K., Sadri, H., Häussler, S., Webb, L. A., Urh, C., Wagner, M., … Sauerwein, H. (2019). Comparison of performance and metabolism from late pregnancy to early lactation in dairy cows with elevated v. normal body condition at dry-off. Animal, 13(7), 1478-1488. doi:10.1017/s1751731118003385

Sundberg, B. E., Wååg, E., Jacobsson, J. A., Stephansson, O., Rumaks, J., Svirskis, S., … Fredriksson, R. (2008). The Evolutionary History and Tissue Mapping of Amino Acid Transporters Belonging to Solute Carrier Families SLC32, SLC36, and SLC38. Journal of Molecular Neuroscience, 35(2), 179-193. doi:10.1007/s12031-008-9046-x

Takagi, M., Yonezawa, T., Haga, S., Shingu, H., Kobayashi, Y., Takahashi, T., … Katoh, K. (2008). Changes of activity and mRNA expression of urea cycle enzymes in the liver of developing Holstein calves1. Journal of Animal Science, 86(7), 1526-1532. doi:10.2527/jas.2007-0799

Trayhurn, P. (2013). Hypoxia and Adipose Tissue Function and Dysfunction in Obesity. Physiological Reviews, 93(1), 1-21. doi:10.1152/physrev.00017.2012

Trevisi, E., Amadori, M., Cogrossi, S., Razzuoli, E., & Bertoni, G. (2012). Metabolic stress and inflammatory response in high-yielding, periparturient dairy cows. Research in Veterinary Science, 93(2), 695-704. doi:10.1016/j.rvsc.2011.11.008

Vailati-Riboni, M., Farina, G., Batistel, F., Heiser, A., Mitchell, M. D., Crookenden, M. A., … Loor, J. J. (2017). Far-off and close-up dry matter intake modulate indicators of immunometabolic adaptations to lactation in subcutaneous adipose tissue of pasture-based transition dairy cows. Journal of Dairy Science, 100(3), 2334-2350. doi:10.3168/jds.2016-11790

Vailati-Riboni, M., Kanwal, M., Bulgari, O., Meier, S., Priest, N. V., Burke, C. R., … Loor, J. J. (2016). Body condition score and plane of nutrition prepartum affect adipose tissue transcriptome regulators of metabolism and inflammation in grazing dairy cows during the transition period. Journal of Dairy Science, 99(1), 758-770. doi:10.3168/jds.2015-10046

Vailati Riboni, M., Meier, S., Priest, N. V., Burke, C. R., Kay, J. K., McDougall, S., … Loor, J. J. (2015). Adipose and liver gene expression profiles in response to treatment with a nonsteroidal antiinflammatory drug after calving in grazing dairy cows. Journal of Dairy Science, 98(5), 3079-3085. doi:10.3168/jds.2014-8579

Van der Vos, K. E., & Coffer, P. J. (2012). Glutamine metabolism links growth factor signaling to the regulation of autophagy. Autophagy, 8(12), 1862-1864. doi:10.4161/auto.22152

Wang, D., Wan, X., Peng, J., Xiong, Q., Niu, H., Li, H., … Jiang, S. (2017). The effects of reduced dietary protein level on amino acid transporters and mTOR signaling pathway in pigs. Biochemical and Biophysical Research Communications, 485(2), 319-327. doi:10.1016/j.bbrc.2017.02.084

Wang, X., Frank, J. W., Little, D. R., Dunlap, K. A., Satterfield, M. C., Burghardt, R. C., … Bazer, F. W. (2014). Functional role of arginine during the peri‐implantation period of pregnancy. I. Consequences of loss of function of arginine transporter SLC7A1 mRNA in ovine conceptus trophectoderm. The FASEB Journal, 28(7), 2852-2863. doi:10.1096/fj.13-248757

Webb, L. A., Sadri, H., Schuh, K., Egert, S., Stehle, P., Meyer, I., … Sauerwein, H. (2020). Branched-chain amino acids: Abundance of their transporters and metabolizing enzymes in adipose tissue, skeletal muscle, and liver of dairy cows at high or normal body condition. Journal of Dairy Science, 103(3), 2847-2863. doi:10.3168/jds.2019-17147

Webb, L. A., Sadri, H., von Soosten, D., Dänicke, S., Egert, S., Stehle, P., & Sauerwein, H. (2019). Changes in tissue abundance and activity of enzymes related to branched-chain amino acid catabolism in dairy cows during early lactation. Journal of Dairy Science, 102(4), 3556-3568. doi:10.3168/jds.2018-14463

Whiteman, E. L., Cho, H., & Birnbaum, M. J. (2002). Role of Akt/protein kinase B in metabolism. Trends in Endocrinology & Metabolism, 13(10), 444-451. doi:10.1016/s1043-2760(02)00662-8

Xue, M.-Y., Sun, H.-Z., Wu, X.-H., Liu, J.-X., & Guan, L. L. (2020). Multi-omics reveals that the rumen microbiome and its metabolome together with the host metabolome contribute to individualized dairy cow performance. Microbiome, 8(1). doi:10.1186/s40168-020-00819-8

Zachut, M., Honig, H., Striem, S., Zick, Y., Boura-Halfon, S., & Moallem, U. (2013). Periparturient dairy cows do not exhibit hepatic insulin resistance, yet adipose-specific insulin resistance occurs in cows prone to high weight loss. Journal of Dairy Science, 96(9), 5656-5669. doi:10.3168/jds.2012-6142

Zhang, F., Li, D., Wu, Q., Sun, J., Guan, W., Hou, Y., … Wang, J. (2019). Prepartum body conditions affect insulin signaling pathways in postpartum adipose tissues in transition dairy cows. Journal of Animal Science and Biotechnology, 10(1). doi:10.1186/s40104-019-0347-4

Zhou, Z., Bulgari, O., Vailati-Riboni, M., Trevisi, E., Ballou, M. A., Cardoso, F. C., … Loor, J. J. (2016). Rumen-protected methionine compared with rumen-protected choline improves immunometabolic status in dairy cows during the peripartal period. Journal of Dairy Science, 99(11), 8956-8969. doi:10.3168/jds.2016-10986

[-]

recommendations

 

Este ítem aparece en la(s) siguiente(s) colección(ones)

Mostrar el registro completo del ítem