56 research outputs found

    Methyl donor supply to heat stress-challenged polymorphonuclear leukocytes from lactating Holstein cows enhances 1-carbon metabolism, immune response, and cytoprotective gene network abundance

    Full text link
    [EN] Mechanisms controlling immune function of dairy cows are dysregulated during heat stress (HS). Methyl donor supply-methionine (Met) and choline (Chop-positively modulates innate immune function, particularly antioxidant systems of polymorphonuclear leukocytes (PMN). The objective of this study was to investigate the effect of Met and Chol supply in vitro on mRNA abundance of genes related to 1-carbon metabolism, inflammation, and immune function in short-term cultures of PMN isolated from mid-lactating Holstein cows in response to heat challenge. Blood PMN were isolated from 5 Holstein cows (153 +/- 5 d postpartum, 34.63 +/- 2.73 kg/d of milk production; mean +/- SD). The PMN were incubated for 2 h at thermal-neutral (37 degrees C; TN) or heat stress (42 degrees C; HS) temperatures with 3 levels of Chol (0, 400, or 800 mu g/mL) or 3 ratios of Lys:Met (Met; 3.6:1, 2.9:1, or 2.4:1). Supernatant concentrations of IL-1 beta, IL-6, and tumor necrosis factor-alpha were measured via bovine-specific ELISA. Fold-changes in mRNA abundance were calculated separately for Chol and Met treatments to obtain the fold-change response at 42 degrees C (HS) relative to 37 degrees C (TN). Data were subjected to ANOVA using PROC MIXED in SAS (SAS Institute Inc., Cary, NC). Orthogonal contrasts were used to determine the linear or quadratic effect of Met and Chol for mRNA fold-change and supernatant cytokine concentrations. Compared with PMN receiving 0 mu g of Chol/mL, heat-stressed PMN supplemented with Chol at 400 or 800 mu g/mL had greater fold-change in abundance of CBS, CSAD, GSS, GSR, and GPX1. Among genes associated with inflammation and immune function, fold-change in abundance of TLR2, TLR4, IRAK1, IL1B, and IL10 increased with 400 and 800 mu g of Chol/mL compared with PMN receiving 0 mu g of Chol/mL. Fold-change in abundance of SAHH decreased linearly at increasing levels of Met supply. A linear effect was detected for MPO, NFKB1, and SOD1 due to greater fold-change in abundance when Met was increased to reach Lys: Met ratios of 2.9:1 and 2.4:1. Although increasing Chol supply upregulated BAX, BCL2, and HSP70, increased Met supply only upregulated BAX. Under HS conditions, enhancing PMN supply of Chol to 400 mu g/mL effectively increased fold-change in abundance of genes involved in antioxidant production (conferring cellular processes protection from free radicals and reactive oxygen species), inflammatory signaling, and innate immunity. Although similar outcomes were obtained with Met supply at Lys:Met ratios of 2.9:1 and 2.4:1, the response was less pronounced. Both Chol and Met supply enhanced the cytoprotective characteristics of PMN through upregulation of heat shock proteins. Overall, the modulatory effects detected in the present experiment highlight an opportunity to use Met and particularly Chol supplementation during thermal stress.M. Vailati-Riboni was supported in part by Hatch funds under project ILLU-538-914, National Institute of Food and Agriculture (Washington, DC). The authors declare no conflict of interest.Lopreiato, V.; Vailati-Riboni, M.; Parys, C.; Fernández Martínez, CJ.; Minuti, A.; Loor, J. (2020). Methyl donor supply to heat stress-challenged polymorphonuclear leukocytes from lactating Holstein cows enhances 1-carbon metabolism, immune response, and cytoprotective gene network abundance. Journal of Dairy Science. 103(11):10477-10493. https://doi.org/10.3168/jds.2020-18638S104771049310311Abdelmegeid, M. K., Vailati-Riboni, M., Alharthi, A., Batistel, F., & Loor, J. J. (2017). Supplemental methionine, choline, or taurine alter in vitro gene network expression of polymorphonuclear leukocytes from neonatal Holstein calves. Journal of Dairy Science, 100(4), 3155-3165. doi:10.3168/jds.2016-12025Armentano, L. E., Bertics, S. J., & Ducharme, G. A. (1997). Response of Lactating Cows to Methionine or Methionine Plus Lysine Added to High Protein Diets Based on Alfalfa and Heated Soybeans. Journal of Dairy Science, 80(6), 1194-1199. doi:10.3168/jds.s0022-0302(97)76047-8Banerjee, R., Evande, R., Kabil, Ö., Ojha, S., & Taoka, S. (2003). Reaction mechanism and regulation of cystathionine β-synthase. Biochimica et Biophysica Acta (BBA) - Proteins and Proteomics, 1647(1-2), 30-35. doi:10.1016/s1570-9639(03)00044-xBatistel, 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.2017-12689Baumgard, L. H., & Rhoads, R. P. (2013). Effects of Heat Stress on Postabsorptive Metabolism and Energetics. Annual Review of Animal Biosciences, 1(1), 311-337. doi:10.1146/annurev-animal-031412-103644Bernabucci, U., Biffani, S., Buggiotti, L., Vitali, A., Lacetera, N., & Nardone, A. (2014). The effects of heat stress in Italian Holstein dairy cattle. Journal of Dairy Science, 97(1), 471-486. doi:10.3168/jds.2013-6611Bernabucci, U., Lacetera, N., Baumgard, L. H., Rhoads, R. P., Ronchi, B., & Nardone, A. (2010). Metabolic and hormonal acclimation to heat stress in domesticated ruminants. Animal, 4(7), 1167-1183. doi:10.1017/s175173111000090xBoldyrev, A., Bryushkova, E., Mashkina, A., & Vladychenskaya, E. (2013). Why Is Homocysteine Toxic for the Nervous and Immune Systems? Current Aging Science, 6(1), 29-36. doi:10.2174/18746098112059990007Catozzi, C., Ávila, G., Zamarian, V., Pravettoni, D., Sala, G., Ceciliani, F., … Lecchi, C. (2020). In-vitro effect of heat stress on bovine monocytes lifespan and polarization. Immunobiology, 225(2), 151888. doi:10.1016/j.imbio.2019.11.023Chinenov, Y., Gupte, R., & Rogatsky, I. (2013). Nuclear receptors in inflammation control: Repression by GR and beyond. Molecular and Cellular Endocrinology, 380(1-2), 55-64. doi:10.1016/j.mce.2013.04.006Chorąży, M., Kontny, E., Marcinkiewicz, J., & Maśliński, W. (2002). Amino Acids, 23(4), 407-413. doi:10.1007/s00726-002-0204-0Coleman, D. N., Lopreiato, V., Alharthi, A., & Loor, J. J. (2020). Amino acids and the regulation of oxidative stress and immune function in dairy cattle. Journal of Animal Science, 98(Supplement_1), S175-S193. doi:10.1093/jas/skaa138Collier, R. J., Stiening, C. M., Pollard, B. C., VanBaale, M. J., Baumgard, L. H., Gentry, P. C., & Coussens, P. M. (2006). Use of gene expression microarrays for evaluating environmental stress tolerance at the cellular level in cattle1. Journal of Animal Science, 84(suppl_13), E1-E13. doi:10.2527/2006.8413_supple1xCouper, K. N., Blount, D. G., & Riley, E. M. (2008). IL-10: The Master Regulator of Immunity to Infection. The Journal of Immunology, 180(9), 5771-5777. doi:10.4049/jimmunol.180.9.5771Del Vesco, A. P., Gasparino, E., Grieser, D. de O., Zancanela, V., Soares, M. A. M., & de Oliveira Neto, A. R. (2015). Effects of methionine supplementation on the expression of oxidative stress-related genes in acute heat stress-exposed broilers. British Journal of Nutrition, 113(4), 549-559. doi:10.1017/s0007114514003535Ekremoğlu, M., Türközkan, N., Erdamar, H., Kurt, Y., & Yaman, H. (2006). Protective effect of taurine on respiratory burst activity of polymorphonuclear leukocytes in endotoxemia. Amino Acids, 32(3), 413-417. doi:10.1007/s00726-006-0382-2El-Benna, J., Hurtado-Nedelec, M., Marzaioli, V., Marie, J.-C., Gougerot-Pocidalo, M.-A., & Dang, P. M.-C. (2016). Priming of the neutrophil respiratory burst: role in host defense and inflammation. Immunological Reviews, 273(1), 180-193. doi:10.1111/imr.12447Esposito, G., Irons, P. C., Webb, E. C., & Chapwanya, A. (2014). Interactions between negative energy balance, metabolic diseases, uterine health and immune response in transition dairy cows. Animal Reproduction Science, 144(3-4), 60-71. doi:10.1016/j.anireprosci.2013.11.007Fear, J. M., & Hansen, P. J. (2011). Developmental Changes in Expression of Genes Involved in Regulation of Apoptosis in the Bovine Preimplantation Embryo1. Biology of Reproduction, 84(1), 43-51. doi:10.1095/biolreprod.110.086249Gao, S. T., Guo, J., Quan, S. Y., Nan, X. M., Fernandez, M. V. S., Baumgard, L. H., & Bu, D. P. (2017). The effects of heat stress on protein metabolism in lactating Holstein cows. Journal of Dairy Science, 100(6), 5040-5049. doi:10.3168/jds.2016-11913Han, Z.-Y., Mu, T., & Yang, Z. (2014). Methionine protects against hyperthermia-induced cell injury in cultured bovine mammary epithelial cells. Cell Stress and Chaperones, 20(1), 109-120. doi:10.1007/s12192-014-0530-7Heiser, A., LeBlanc, S. J., & McDougall, S. (2018). Pegbovigrastim treatment affects gene expression in neutrophils of pasture-fed, periparturient cows. Journal of Dairy Science, 101(9), 8194-8207. doi:10.3168/jds.2017-14129Horowitz, M. (2001). Heat acclimation: phenotypic plasticity and cues to the underlying molecular mechanisms. Journal of Thermal Biology, 26(4-5), 357-363. doi:10.1016/s0306-4565(01)00044-4Hunter-Lavin, C., Davies, E. L., Bacelar, M. M. F. V. G., Marshall, M. J., Andrew, S. M., & Williams, J. H. H. (2004). Hsp70 release from peripheral blood mononuclear cells. Biochemical and Biophysical Research Communications, 324(2), 511-517. doi:10.1016/j.bbrc.2004.09.075Ingvartsen, K. L., & Moyes, K. (2013). Nutrition, immune function and health of dairy cattle. Animal, 7, 112-122. doi:10.1017/s175173111200170xJoshi, B. C., Joshi, H. B., McDowell, R. E., & Sadhu, D. P. (1968). Composition of Skin Secretions from Three Indian Breeds of Cattle Under Thermal Stress. Journal of Dairy Science, 51(6), 917-920. doi:10.3168/jds.s0022-0302(68)87105-xKobayashi, S. D., & DeLeo, F. R. (2009). Role of neutrophils in innate immunity: a systems biology‐level approach. Wiley Interdisciplinary Reviews: Systems Biology and Medicine, 1(3), 309-333. doi:10.1002/wsbm.32Kumar, H., Kawai, T., & Akira, S. (2011). Pathogen Recognition by the Innate Immune System. International Reviews of Immunology, 30(1), 16-34. doi:10.3109/08830185.2010.529976Lacetera, N., Bernabucci, U., Basiricò, L., Morera, P., & Nardone, A. (2009). Heat shock impairs DNA synthesis and down-regulates gene expression for leptin and Ob-Rb receptor in concanavalin A-stimulated bovine peripheral blood mononuclear cells. Veterinary Immunology and Immunopathology, 127(1-2), 190-194. doi:10.1016/j.vetimm.2008.09.020Lacetera, N., Bernabucci, U., Scalia, D., Basiricò, L., Morera, P., & Nardone, A. (2006). Heat Stress Elicits Different Responses in Peripheral Blood Mononuclear Cells from Brown Swiss and Holstein Cows. Journal of Dairy Science, 89(12), 4606-4612. doi:10.3168/jds.s0022-0302(06)72510-3Lecchi, C., Rota, N., Vitali, A., Ceciliani, F., & Lacetera, N. (2016). In vitro assessment of the effects of temperature on phagocytosis, reactive oxygen species production and apoptosis in bovine polymorphonuclear cells. Veterinary Immunology and Immunopathology, 182, 89-94. doi:10.1016/j.vetimm.2016.10.007Loos, H., Roos, D., Weening, R., & Houwerzijl, J. (1976). Familial deficiency of glutathione reductase in human blood cells. Blood, 48(1), 53-62. doi:10.1182/blood.v48.1.53.53Lopreiato, V., Vailati-Riboni, M., Bellingeri, A., Khan, I., Farina, G., Parys, C., & Loor, J. J. (2019). Inflammation and oxidative stress transcription profiles due to in vitro supply of methionine with or without choline in unstimulated blood polymorphonuclear leukocytes from lactating Holstein cows. Journal of Dairy Science, 102(11), 10395-10410. doi:10.3168/jds.2019-16413Lubos, E., Loscalzo, J., & Handy, D. E. (2011). Glutathione Peroxidase-1 in Health and Disease: From Molecular Mechanisms to Therapeutic Opportunities. Antioxidants & Redox Signaling, 15(7), 1957-1997. doi:10.1089/ars.2010.3586Lushchak, V. I. (2012). Glutathione Homeostasis and Functions: Potential Targets for Medical Interventions. Journal of Amino Acids, 2012, 1-26. doi:10.1155/2012/736837McGuire, M. A., Beede, D. K., DeLorenzo, M. A., Wilcox, C. J., Huntington, G. B., Reynolds, C. K., & Collier, R. J. (1989). Effects of Thermal Stress and Level of Feed Intake on Portal Plasma Flow and Net Fluxes of Metabolites in Lactating Holstein Cows2,3. Journal of Animal Science, 67(4), 1050-1060. doi:10.2527/jas1989.6741050xMin, L., Zheng, N., Zhao, S., Cheng, J., Yang, Y., Zhang, Y., … Wang, J. (2016). Long-term heat stress induces the inflammatory response in dairy cows revealed by plasma proteome analysis. Biochemical and Biophysical Research Communications, 471(2), 296-302. doi:10.1016/j.bbrc.2016.01.185Moyes, 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/s10142-009-0154-7Moyes, K. M., Graugnard, D. E., Khan, M. J., Mukesh, M., & Loor, J. J. (2014). Postpartal immunometabolic gene network expression and function in blood neutrophils are altered in response to prepartal energy intake and postpartal intramammary inflammatory challenge. Journal of Dairy Science, 97(4), 2165-2177. doi:10.3168/jds.2013-7433Nakamura, M. (2000). Preconditioning decreases Bax expression, PMN accumulation and apoptosis in reperfused rat heart. Cardiovascular Research, 45(3), 661-670. doi:10.1016/s0008-6363(99)00393-4Oeckinghaus, A., & Ghosh, S. (2009). The NF- B Family of Transcription Factors and Its Regulation. Cold Spring Harbor Perspectives in Biology, 1(4), a000034-a000034. doi:10.1101/cshperspect.a000034Osorio, 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.2014-8680Salama, A. A. K., Duque, M., Wang, L., Shahzad, K., Olivera, M., & Loor, J. J. (2019). Enhanced supply of methionine or arginine alters mechanistic target of rapamycin signaling proteins, messenger RNA, and microRNA abundance in heat-stressed bovine mammary epithelial cells in vitro. Journal of Dairy Science, 102(3), 2469-2480. doi:10.3168/jds.2018-15219Schell, M. T., Spitzer, A. L., Johnson, J. A., Lee, D., & Harris, H. W. (2005). Heat Shock Inhibits NF-kB Activation in a Dose- and Time-Dependent Manner. Journal of Surgical Research, 129(1), 90-93. doi:10.1016/j.jss.2005.05.025Silanikove, N. (2000). Effects of heat stress on the welfare of extensively managed domestic ruminants. Livestock Production Science, 67(1-2), 1-18. doi:10.1016/s0301-6226(00)00162-7Stankiewicz, A. R., Lachapelle, G., Foo, C. P. Z., Radicioni, S. M., & Mosser, D. D. (2005). Hsp70 Inhibits Heat-induced Apoptosis Upstream of Mitochondria by Preventing Bax Translocation. Journal of Biological Chemistry, 280(46), 38729-38739. doi:10.1074/jbc.m509497200Steel, G. J., Fullerton, D. M., Tyson, J. R., & Stirling, C. J. (2004). Coordinated Activation of Hsp70 Chaperones. Science, 303(5654), 98-101. doi:10.1126/science.1092287Sun, D., Chen, D., Du, B., & Pan, J. (2005). Heat Shock Response Inhibits NF-κB Activation and Cytokine Production in Murine Kupffer Cells. Journal of Surgical Research, 129(1), 114-121. doi:10.1016/j.jss.2005.05.028Taraktsoglou, M., Szalabska, U., Magee, D. A., Browne, J. A., Sweeney, T., Gormley, E., & MacHugh, D. E. (2011). Transcriptional profiling of immune genes in bovine monocyte-derived macrophages exposed to bacterial antigens. Veterinary Immunology and Immunopathology, 140(1-2), 130-139. doi:10.1016/j.vetimm.2010.12.002Trevisi, E., Jahan, N., Bertoni, G., Ferrari, A., & Minuti, A. (2015). Pro-Inflammatory Cytokine Profile in Dairy Cows: Consequences for New Lactation. Italian Journal of Animal Science, 14(3), 3862. doi:10.4081/ijas.2015.3862Tsan, M.-F., & Gao, B. (2004). Cytokine function of heat shock proteins. American Journal of Physiology-Cell Physiology, 286(4), C739-C744. doi:10.1152/ajpcell.00364.2003Vailati-Riboni, M., Zhou, Z., Jacometo, C. B., Minuti, A., Trevisi, E., Luchini, D. N., & Loor, J. J. (2017). Supplementation with rumen-protected methionine or choline during the transition period influences whole-blood immune response in periparturient dairy cows. Journal of Dairy Science, 100(5), 3958-3968. doi:10.3168/jds.2016-11812Yan, J., Meng, X., Wancket, L. M., Lintner, K., Nelin, L. D., Chen, B., … Liu, Y. (2012). Glutathione Reductase Facilitates Host Defense by Sustaining Phagocytic Oxidative Burst and Promoting the Development of Neutrophil Extracellular Traps. The Journal of Immunology, 188(5), 2316-2327. doi:10.4049/jimmunol.1102683Zhou, 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.2016-10986Zhou, Z., Ferdous, F., Montagner, P., Luchini, D. N., Corrêa, M. N., & Loor, J. J. (2018). Methionine and choline supply during the peripartal period alter polymorphonuclear leukocyte immune response and immunometabolic gene expression in Holstein cows. Journal of Dairy Science, 101(11), 10374-10382. doi:10.3168/jds.2018-14972Zhou, Z., Vailati-Riboni, M., Trevisi, E., Drackley, J. K., Luchini, D. N., & Loor, J. J. (2016). Better postpartal performance in dairy cows supplemented with rumen-protected methionine compared with choline during the peripartal period. Journal of Dairy Science, 99(11), 8716-8732. doi:10.3168/jds.2015-1052

    Better postpartal performance in dairy cows supplemented with rumen-protected methionine compared with choline during the peripartal period

    Get PDF
    The onset of lactation in dairy cows is characterized by high output of methylated compounds in milk when sources of methyl group are in short supply. Methionine and choline (CHOL) are key methyl donors and their availability during this time may be limiting for milk production, hepatic lipid metabolism, and immune function. Supplementing rumen-protected Met and CHOL may improve overall performance and health of transition cows. The objective of this study was to evaluate the effect of supplemental rumen-protected Met and CHOL on performance and health of transition cows. Eighty-one multiparous Holstein cows were used in a randomized, complete, unbalanced block design with 2×2 factorial arrangement of Met (Smartamine M, Adisseo NA, Alpharetta, GA) and CHOL (ReaShure, Balchem Inc., New Hampton, NY) inclusion (with or without). Treatments (20 to 21 cows each) were control (CON), CON+Met (SMA), CON+CHOL (REA), and CON+Met+CHOL (MIX). From -50 to -21d before expected calving, all cows received the same diet (1.40Mcal of NEL/kg of DM) with no Met or CHOL. From -21d to calving, cows received the same close-up diet (1.52Mcal of NEL/kg of DM) and were assigned randomly to treatments (CON, SMA, REA, or MIX) supplied as top dresses. From calving to 30 DIM, cows were fed the same postpartal diet (1.71Mcal of NEL/kg of DM) and continued to receive the same treatments through 30 DIM. The Met supplementation was adjusted daily at 0.08% DM of diet and REA was supplemented at 60g/d. Incidence of clinical ketosis and retained placenta tended to be lower in Met-supplemented cows. Supplementation of Met (SMA, MIX) led to greater DMI compared with other treatments (CON, REA) in both close-up (14.3 vs. 13.2kg/d, SEM 0.3) and first 30d postpartum (19.2 vs. 17.2kg/d, SEM 0.6). Cows supplemented with Met (SMA, MIX) had greater yields of milk (44.2 vs. 40.4kg/d, SEM 1.2), ECM (44.6 vs. 40.5kg/d, SEM 1.0), and FCM (44.6 vs. 40.8kg/d, SEM 1.0) compared with other (CON, REA) treatments. Milk fat content did not differ in response to Met or CHOL. However, milk protein content was greater in Met-supplemented (3.32% vs. 3.14%, SEM 0.04%) but not CHOL-supplemented (3.27 vs. 3.19%, SEM 0.04%) cows. Supplemental CHOL led to greater blood glucose and insulin concentrations with lower glucose:insulin ratio. No Met or CHOL effects were detected for blood fatty acids or BHB, but a Met × time effect was observed for fatty acids due to higher concentrations on d 20. Results from the present study indicate that peripartal supplementation of rumen-protected Met but not CHOL has positive effects on cow performance

    Inflammation and oxidative stress transcription profiles due to in vitro supply of methionine with or without choline in unstimulated blood polymorphonuclear leukocytes from lactating Holstein cows.

    Get PDF
    Neutrophils are the most important polymorphonuclear leukocytes (PMNL), representing the front-line defense involved in pathogen clearance upon invasion. As such, they play a pivotal role in immune and inflammatory responses. Isolated PMNL from 5 mid-lactating Holstein dairy cows were used to evaluate the in vitro effect of methionine (Met) and choline (Chol) supplementation on mRNA expression of genes related to the Met cycle and innate immunity. The target genes are associated with the Met cycle, cell signaling, inflammation, antimicrobial and killing mechanisms, and pathogen recognition. Treatments were allocated in a 3 × 3 factorial arrangement, including 3 Lys-to-Met ratios (L:M, 3.6:1, 2.9:1, or 2.4:1) and 3 levels of supplemental Chol (0, 400, or 800 μg/mL). Three replicates per treatment group were incubated for 2 h at 37°C and 5% atmospheric CO2. Both betaine-homocysteine S-methyltransferase and choline dehydrogenase were undetectable, indicating that PMNL (at least in vitro) cannot generate Met from Chol through the betaine pathway. The PMNL incubated without Chol experienced a specific state of inflammatory mediation [greater interleukin-1β (IL1B), myeloperoxidase (MPO), IL10, and IL6] and oxidative stress [greater cysteine sulfinic acid decarboxylase (CSAD), cystathionine gamma-lyase (CTH), glutathione reductase (GSR), and glutathione synthase (GSS)]. However, data from the interaction L:M × Chol indicated that this negative state could be overcome by supplementing additional Met. This was reflected in the upregulation of methionine synthase (MTR) and toll-like receptor 2 (TLR2); that is, pathogen detection ability. At the lowest level of supplemental Chol, Met downregulated GSS, GSR, IL1B, and IL6, suggesting it could reduce cellular inflammation and enhance antioxidant status. At 400 µg/mL Chol, supplemental Met upregulated PMNL recognition capacity [higher TLR4 and L-selectin (SELL)]. Overall, enhancing the supply of methyl donors to isolated unstimulated PMNL from mid-lactating dairy cows leads to a low level of PMNL activation and upregulates a cytoprotective mechanism against oxidative stress. Enhancing the supply of Met coupled with adequate Chol levels enhances the gene expression of PMNL pathogen-recognition mechanism. These data suggest that Chol supply to PMNL exposed to low levels of Met effectively downregulated the entire repertoire of innate inflammatory-responsive genes. Thus, Met availability in PMNL during an inflammatory challenge may be sufficient for mounting an appropriate biologic response

    Rumen-protected methionine compared with rumen-protected choline improves immunometabolic status in dairy cows during the peripartal period.

    Get PDF
    The immunometabolic status of peripartal cows is altered due to changes in liver function, inflammation, and oxidative stress. Nutritional management during this physiological state can affect the biological components of immunometabolism. The objectives of this study were to measure concentrations of biomarkers in plasma, liver tissue, and milk, and also polymorphonuclear leukocyte function to assess the immunometabolic status of cows supplemented with rumen-protected methionine (Met) or choline (CHOL). Forty-eight multiparous Holstein cows were used in a randomized complete block design with 2×2 factorial arrangement of Met (Smartamine M, Adisseo NA, Alpharetta, GA) and CHOL (ReaShure, Balchem Inc., New Hampton, NY) level (with or without). Treatments (12 cows each) were control (CON), no Met or CHOL; CON and Met (SMA); CON and CHOL (REA); and CON and Met and CHOL (MIX). From -50 to -21d before expected calving, all cows received the same diet [1.40Mcal of net energy for lactation (NEL)/kg of DM] with no Met or CHOL. From -21d to calving, cows received the same close-up diet (1.52Mcal of NEL/kg of DM) and were assigned randomly to each treatment. From calving to 30d, cows were on the same postpartal diet (1.71Mcal of NEL/kg of DM) and continued to receive the same treatments until 30d. The Met supplementation was adjusted daily at 0.08% DM of diet, and CHOL was supplemented at 60g/cow per day. Liver (-10, 7, 21, and 30d) and blood (-10, 4, 8, 20, and 30d) samples were harvested for biomarker analyses. Neutrophil and monocyte phagocytosis and oxidative burst were assessed at d 1, 4, 14, and 28d. The Met-supplemented cows tended to have greater plasma paraoxonase. Greater plasma albumin and IL-6 as well as a tendency for lower haptoglobin were detected in Met- but not CHOL-supplemented cows. Similarly, cows fed Met compared with CHOL had greater concentrations of total and reduced glutathione (a potent intracellular antioxidant) in liver tissue. Upon a pathogen challenge in vitro, blood polymorphonuclear leukocyte phagocytosis capacity and oxidative burst activity were greater in Met-supplemented cows. Overall, liver and blood biomarker analyses revealed favorable changes in liver function, inflammation status, and immune response in Met-supplemented cows

    Photoprotection in intact cells of photosynthetic bacteria: quenching of bacteriochlorophyll fluorescence by carotenoid triplets.

    Get PDF
    Upon high light excitation in photosynthetic bacteria, various triplet states of pigments can accumulate leading to harmful effects. Here, the generation and lifetime of flash-induced carotenoid triplets (3Car) have been studied by observation of the quenching of bacteriochlorophyll (BChl) fluorescence in different strains of photosynthetic bacteria including Rvx. gelatinosus (anaerobic and semianaerobic), Rsp. rubrum, Thio. roseopersicina, Rba. sphaeroides 2.4.1 and carotenoid- and cytochrome-deficient mutants Rba. sphaeroides Ga, R-26, and cycA, respectively. The following results were obtained: (1) 3Car quenching is observed during and not exclusively after the photochemical rise of the fluorescence yield of BChl indicating that the charge separation in the reaction center (RC) and the carotenoid triplet formation are not consecutive but parallel processes. (2) The photoprotective function of 3Car is not limited to the RC only and can be described by a model in which the carotenoids are distributed in the lake of the BChl pigments. (3) The observed lifetime of 3Car in intact cells is the weighted average of the lifetimes of the carotenoids with various numbers of conjugated double bonds in the bacterial strain. (4) The lifetime of 3Car measured in the light is significantly shorter (1-2 mus) than that measured in the dark (2-10 mus). The difference reveals the importance of the dynamics of 3Car before relaxation. The results will be discussed not only in terms of energy levels of the 3Car but also in terms of the kinetics of transitions among different sublevels in the excited triplet state of the carotenoid

    The effect of calving in the summer on the hepatic transcriptome of Holstein cows during the peripartal period

    Get PDF
    The liver is the main metabolic organ coordinating the adaptations that take place during the peripartal period of dairy cows. A successful transition into lactation, rather than management practices alone, depends on environmental factors such as temperature, season of parturition, and photoperiod. Therefore, we analyzed the effect of calving season on the hepatic transcriptome of dairy cows during the transition period. A total of 12 Holstein dairy cows were assigned into 2 groups based on calving season (6 cows March-April, spring; 6 cows June-July, summer, SU). The RNA was extracted from liver samples obtained at -30, 3, and 35 DIM via percutaneous biopsy and hybridized to the Agilent 44K Bovine (V2) Gene Expression Microarray (Agilent Technologies Inc., Santa Clara, CA). A quantitative PCR on 22 target genes was performed to verify and expand the analyses. A total of 4,307 differentially expressed genes were detected (false discovery rate ≤0.05) in SU compared with spring. Furthermore, 73 unique differentially expressed genes were detected in SU compared with spring cows after applying a fold-change threshold ≥3 and ≤-3. For Kyoto Encyclopedia of Genes and Genomes pathways analysis of differentially expressed genes, we used the dynamic impact approach. Ingenuity Pathway Analysis software was used to analyze upstream transcription regulators and perform gene network analysis. Among metabolic pathways, energy metabolism from lipids, carbohydrates, and amino acids was strongly affected by calving in SU, with a reduced level of fatty acid synthesis, oxidation, re-esterification, and synthesis of lipoproteins, leading to hepatic lipidosis. Glycan-synthesis was downregulated in SU cows probably as a mechanism to counteract the progression of this lipidosis. In contrast, calving in the SU resulted in upregulation of gluconeogenesis but also greater use of glucose as an energy source. Among nonmetabolic pathways, the heat-shock response was obviously activated in SU cows but was also associated with inflammatory and intracellular stress response. Furthermore, data support a recent finding that cows experience endoplasmic reticulum stress around parturition. Transcription regulator analysis revealed how metabolic changes are related to important regulatory mechanisms, including epigenetic modification. The holistic analyses of the liver transcriptome response to calving in the summer at high environmental temperatures underscore how transition cows should be carefully managed during this period, as they experience alterations in liver energy metabolism and inflammatory state increasing susceptibility to health disorders in early postpartum

    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

    Get PDF
    The common practice of increasing dietary energy density during the close-up dry period (last ∼3 wk prepartum) has been recently associated with a higher incidence of metabolic disorders after calving. Despite these reports, over-feeding of metabolizable energy (ME) during the far-off, nonlactating period is a common management policy aimed at achieving optimum calving body condition score (BCS) in pasture-based systems, as cows are generally thinner than total mixed ration cows at the end of lactation. Our hypothesis was that both far-off and close-up overfeeding influence the peripartum adipose tissue changes associated with energy balance and inflammatory state. Sixty mid-lactation, grazing dairy cows of mixed age and breed were randomly allocated to 1 of 2 groups that were managed through late lactation to achieve a low and high BCS (approximately 4.25 and 5.0 on a 10-point scale) at dry-off. The low BCS cows were then overfed ME to ensure that they achieved the same BCS as the higher BCS group by calving. Within each rate of BCS gain treatment, cows were offered 65, 90, or 120% of their pre-calving ME requirements for 3 wk pre-calving in a 2 × 3 factorial arrangement of treatments (i.e., 10 cows/treatment). Subcutaneous adipose tissue was collected via biopsy at −1, 1, and 4 wk relative to parturition. Quantitative PCR was used to measure mRNA and microRNA expression of targets related to adipogenesis and inflammation. Cows overfed in the far-off period had increased expression of miR-143 and miR-378 prepartum (−1 wk) indicating greater adipogenesis, consistent with their rapid gain in BCS following dry-off. Furthermore, the lower postpartum expression of IL6, TNF, TLR4, TLR9, and miR-145, and a higher abundance of miR-99a indicated lower body fat mobilization in early lactation in the same group. In the close-up period, feeding either 65 or 120% of ME requirements caused changes in FASN, IL1B, IL6R, TLR9, and the microRNA miR-143, miR-155, and miR-378. Their respective expression patterns indicate a tentative negative-feedback mechanism in metabolically compromised, feed-restricted cows, and a possible immune-related stimulation of lipolysis in apparently static adipocytes in overfed cows. Data from cows fed 90% of ME requirements indicate the existence of a balance between lipolytic (inflammatory-related) and anti-lipolytic signals, to prime the mobilization machinery in light of imminent lactation. Overall, results indicate that far-off dry cow nutrition influences peripartum adipose tissue metabolism, with neither strategy negatively affecting the physiological adaptation to lactation. Furthermore, to ensure a favorable transition, cows should be subjected to a small feed restriction in the close-up period, irrespective of far-off nutritional management

    Benefits of Amino Acid Balancing on Metabolism, Inflammation, and Oxidative Stress

    Full text link
    This information was presented at the 2016 Cornell Nutrition Conference for Feed Manufacturers, organized by the Department of Animal Science In the College of Agriculture and Life Sciences at Cornell University. Softcover copies of the entire conference proceedings may be purchased at http://ansci.cals.cornell.edu/extension-outreach/adult-extension/dairy-management/order-proceedings-resources
    corecore