DOI QR코드

DOI QR Code

Identification of genomic regions and genes associated with subclinical ketosis in periparturient dairy cows

  • Jihwan Lee (Dairy Science Division, National Institute of Animal Science, RDA) ;
  • KwangHyeon Cho (Department of Beef and Dairy Science, Korea National College of Agriculture and Fisheries) ;
  • Kent A. Weigel (Department of Animal and Dairy Sciences, University of Wisconsin) ;
  • Heather M. White (Department of Animal and Dairy Sciences, University of Wisconsin) ;
  • ChangHee Do (Department of Animal and Dairy Sciences, Chungnam National University) ;
  • Inchul Choi (Department of Animal and Dairy Sciences, Chungnam National University)
  • Received : 2023.06.09
  • Accepted : 2023.09.12
  • Published : 2024.05.31

Abstract

Subclinical ketosis (SCK) is a prevalent metabolic disorder that occurs during the transition to lactation period. It is defined as a high blood concentration of ketone bodies (beta-hydroxybutyric acid f ≥ 1.2 mmol/L) within the first few weeks of lactation, and often presents without clinical signs. SCK is mainly caused by negative energy balance (NEB). The objective of this study is to identify single nucleotide polymorphisms (SNPs) associated with SCK using genome-wide association studies (GWAS), and to predict the biological functions of proximal genes using gene-set enrichment analysis (GSEA). Blood samples were collected from 112 Holstein cows between 5 and 18 days postpartum to determine the incidence of SCK. Genomic DNA extracted from both SCK and healthy cows was examined using the Illumina Bovine SNP50K BeadChip for genotyping. GWAS revealed 194 putative SNPs and 163 genes associated with those SNPs. Additionally, GSEA showed that the genes retrieved by Database for Annotation, Visualization, and Integrated Discovery (DAVID) belonged to calcium signaling, starch and sucrose, immune network, and metabolic pathways. Furthermore, the proximal genes were found to be related to germ cell and early embryo development. In summary, this study proposes several feasible SNPs and genes associated with SCK through GWAS and GSEA. These candidates can be utilized in selective breeding programs to reduce the genetic risk for SCK and subfertility in high-performance dairy cows.

Keywords

Acknowledgement

We also thank to Ryan S. Pralle who helped to collect, combine, and provide the dataset.

References

  1. Lucy MC. Reproductive loss in high-producing dairy cattle: where will it end? J Dairy Sci. 2001;84:1277-93. https://doi.org/10.3168/jds.S0022-0302(01)70158-0 
  2. Walsh RB, Walton JS, Kelton DF, LeBlanc SJ, Leslie KE, Duffield TF. The effect of subclinical ketosis in early lactation on reproductive performance of postpartum dairy cows. J Dairy Sci. 2007;90:2788-96. https://doi.org/10.3168/jds.2006-560 
  3. Drackley JK, Cardoso FC. Prepartum and postpartum nutritional management to optimize fertility in high-yielding dairy cows in confined TMR systems. Animal. 2014;8:5-14. https://doi.org/10.1017/S1751731114000731 
  4. Andersson L. Subclinical ketosis in dairy cows. Vet Clin North Am Food Anim Pract. 1988;4:233-51. https://doi.org/10.1016/S0749-0720(15)31046-X 
  5. Duffield TF, Sandals D, Leslie KE, Lissemore K, McBride BW, Lumsden JH, et al. Efficacy of monensin for the prevention of subclinical ketosis in lactating dairy cows. J Dairy Sci. 1998;81:2866-73. https://doi.org/10.3168/jds.S0022-0302(98)75846-1 
  6. Oetzel GR. Monitoring and testing dairy herds for metabolic disease. Vet Clin North Am Food Anim Pract. 2004;20:651-74. https://doi.org/10.1016/j.cvfa.2004.06.006 
  7. Raboisson D, Mounie M, Maigne E. Diseases, reproductive performance, and changes in milk production associated with subclinical ketosis in dairy cows: a meta-analysis and review. J Dairy Sci. 2014;97:7547-63. https://doi.org/10.3168/jds.2014-8237 
  8. Suthar VS, Canelas-Raposo J, Deniz A, Heuwieser W. Prevalence of subclinical ketosis and relationships with postpartum diseases in European dairy cows. J Dairy Sci. 2013;96:2925-38. https://doi.org/10.3168/jds.2012-6035 
  9. Rathbun FM, Pralle RS, Bertics SJ, Armentano LE, Cho K, Do C, et al. Relationships between body condition score change, prior mid-lactation phenotypic residual feed intake, and hyperketonemia onset in transition dairy cows. J Dairy Sci. 2017;100:3685-96. https://doi.org/10.3168/jds.2016-12085 
  10. Rutherford AJ, Oikonomou G, Smith RF. The effect of subclinical ketosis on activity at estrus and reproductive performance in dairy cattle. J Dairy Sci. 2016;99:4808-15. https://doi.org/10.3168/jds.2015-10154 
  11. Asl AN, Nazifi S, Ghasrodashti AR, Olyaee A. Prevalence of subclinical ketosis in dairy cattle in the Southwestern Iran and detection of cutoff point for NEFA and glucose concentrations for diagnosis of subclinical ketosis. Prev Vet Med. 2011;100:38-43. https://doi.org/10.1016/j.prevetmed.2011.02.013 
  12. Duffield TF, Lissemore KD, McBride BW, Leslie KE. Impact of hyperketonemia in early lactation dairy cows on health and production. J Dairy Sci. 2009;92:571-80. https://doi.org/10.3168/jds.2008-1507 
  13. McArt JAA, Nydam DV, Oetzel GR. Epidemiology of subclinical ketosis in early lactation dairy cattle. J Dairy Sci. 2012;95:5056-66. https://doi.org/10.3168/jds.2012-5443 
  14. Loor JJ, Everts RE, Bionaz M, Dann HM, Morin DE, Oliveira R, et al. Nutrition-induced ketosis alters metabolic and signaling gene networks in liver of periparturient dairy cows. Physiol Genomics. 2007;32:105-16. https://doi.org/10.1152/physiolgenomics.00188.2007 
  15. Kuhla B, Albrecht D, Kuhla S, Metges CC. Proteome analysis of fatty liver in feed-deprived dairy cows reveals interaction of fuel sensing, calcium, fatty acid, and glycogen metabolism. Physiol Genomics. 2009;37:88-98. https://doi.org/10.1152/physiolgenomics.90381.2008 
  16. Pryce JE, Parker Gaddis KL, Koeck A, Bastin C, Abdelsayed M, Gengler N, et al. Invited review: opportunities for genetic improvement of metabolic diseases. J Dairy Sci. 2016;99:6855-73. https://doi.org/10.3168/jds.2016-10854 
  17. Weigel KA, Pralle RS, Adams H, Cho K, Do C, White HM. Prediction of whole-genome risk for selection and management of hyperketonemia in Holstein dairy cattle. J Anim Breed Genet. 2017;134:275-85. https://doi.org/10.1111/jbg.12259 
  18. Askland K, Read C, Moore J. Pathways-based analyses of whole-genome association study data in bipolar disorder reveal genes mediating ion channel activity and synaptic neurotransmission. Hum Genet. 2009;125:63-79. https://doi.org/10.1007/s00439-008-0600-y 
  19. Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci. 2005;102:15545-50. https://doi.org/10.1073/pnas.0506580102 
  20. Torkamani A, Topol EJ, Schork NJ. Pathway analysis of seven common diseases assessed by genome-wide association. Genomics. 2008;92:265-72. https://doi.org/10.1016/j.ygeno.2008.07.011 
  21. Iwersen M, Falkenberg U, Voigtsberger R, Forderung D, Heuwieser W. Evaluation of an electronic cowside test to detect subclinical ketosis in dairy cows. J Dairy Sci. 2009;92:2618-24. https://doi.org/10.3168/jds.2008-1795 
  22. Lee SH, Lee D, Lee M, Ryoo SH, Seo S, Choi I. Analysis of single nucleotide polymorphisms related to heifer fertility in Hanwoo (Korean cattle). Anim Biotechnol. 2022;33:964-9. https://doi.org/10.1080/10495398.2020.1856124 
  23. Tetens J, Heuer C, Heyer I, Klein MS, Gronwald W, Junge W, et al. Polymorphisms within the APOBR gene are highly associated with milk levels of prognostic ketosis biomarkers in dairy cows. Physiol Genomics. 2015;47:129-37. https://doi.org/10.1152/physiolge nomics.00126.2014 
  24. Mahmoudi A, Zargaran A, Amini HR, Assadi A, Vajdi Hokmabad R, Eghbalsaied S. A SNP in the 3'-untranslated region of AMPKγ1 may associate with serum ketone body and milk production of Holstein dairy cows. Gene. 2015;574:48-52. https://doi.org/10.1016/j.gene.2015.07.077 
  25. Pralle RS, Schultz NE, White HM, Weigel KA. Hyperketonemia GWAS and parity-dependent SNP associations in Holstein dairy cows intensively sampled for blood β-hydroxybutyrate concentration. Physiol Genomics. 2020;52:347-57. https://doi.org/10.1152/physiolgenomics.00016.2020 
  26. Bough KJ, Wetherington J, Hassel B, Pare JF, Gawryluk JW, Greene JG, et al. Mitochondrial biogenesis in the anticonvulsant mechanism of the ketogenic diet. Ann Neurol. 2006;60:223-35. https://doi.org/10.1002/ana.20899 
  27. Peiris H, Duffield MD, Fadista J, Jessup CF, Kashmir V, Genders AJ, et al. A syntenic cross species Aneuploidy genetic screen links RCAN1 expression to β-cell mitochondrial dysfunction in Type 2 diabetes. PLOS Genet. 2016;12:e1006033. https://doi.org/10.1371/journal.pgen.1006033 
  28. Bonner-Weir S, Aguayo-Mazzucato C. Pancreatic β-cell heterogeneity revisited. Nature. 2016;535:365-6. https://doi.org/10.1038/nature18907 
  29. Rask-Andersen M, Almen MS, Lind L, Schioth HB. Association of the LINGO2-related SNP rs10968576 with body mass in a cohort of elderly Swedes. Mol Genet Genomics. 2015;290:1485-91. https://doi.org/10.1007/s00438-015-1009-7 
  30. van der Louw EJTM, Desadien R, Vehmeijer FOL, van der Sijs H, Catsman-Berrevoets CE, Neuteboom RF. Concomitant lamotrigine use is associated with decreased efficacy of the ketogenic diet in childhood refractory epilepsy. Seizure. 2015;32:75-7. https://doi.org/10.1016/j.seizure.2015.09.007 
  31. Zhang L, Chu X, Wang H, Xie H, Guo C, Cao L, et al. Dysregulations of UDP-glucuronosyltransferases in rats with valproic acid and high fat diet induced fatty liver. Eur J Pharmacol. 2013;721:277-85. https://doi.org/10.1016/j.ejphar.2013.09.024 
  32. Bell AW. Regulation of organic nutrient metabolism during transition from late pregnancy to early lactation. J Anim Sci. 1995;73:2804-19. https://doi.org/10.2527/1995.7392804x 
  33. Horst RL, Goff JP, Reinhardt TA. Calcium and vitamin D metabolism in the dairy cow. J Dairy Sci. 1994;77:1936-51. https://doi.org/10.3168/jds.S0022-0302(94)77140-X 
  34. Horst RL, Goff JP, Reinhardt TA, Buxton DR. Strategies for preventing milk fever in dairy cattle. J Dairy Sci. 1997;80:1269-80. https://doi.org/10.3168/jds.S0022-0302(97)76056-9 
  35. Goff JP, Horst RL. Physiological changes at parturition and their relationship to metabolic disorders. J Dairy Sci. 1997;80:1260-8. https://doi.org/10.3168/jds.S0022-0302(97)76055-7 
  36. Mallard BA, Dekkers JC, Ireland MJ, Leslie KE, Sharif S, Vankampen CL, et al. Alteration in immune responsiveness during the peripartum period and its ramification on dairy cow and calf health. J Dairy Sci. 1998;81:585-95. https://doi.org/10.3168/jds.S0022-0302(98)75612-7 
  37. Suriyasathaporn W, Heuer C, Noordhuizen-Stassen EN, Schukken YH. Hyperketonemia and the impairment of udder defense: a review. Vet Res. 2000;31:397-412. https://doi.org/10.1051/vetres:2000128 
  38. Sato S, Suzuki T, Okada K. Suppression of lymphocyte blastogenesis in cows with puerperal metritis and mastitis. J Vet Med Sci. 1995;57:373-5. https://doi.org/10.1292/jvms.57.373 
  39. Suriyasathaporn W, Daemen AJJM, Noordhuizen-Stassen EN, Dieleman SJ, Nielen M, Schukken YH. β-hydroxybutyrate levels in peripheral blood and ketone bodies supplemented in culture media affect the in vitro chemotaxis of bovine leukocytes. Vet Immunol Immunopathol. 1999;68:177-86. https://doi.org/10.1016/S0165-2427(99)00017-3 
  40. Zdzisinska B, Filar J, Paduch R, Kaczor J, Lokaj I, Kandefer-Szerszen M. The influence of ketone bodies and glucose on interferon, tumor necrosis factor production and NO release in bovine aorta endothelial cells. Vet Immunol Immunopathol. 2000;74:237-47. https://doi.org/10.1016/S0165-2427(00)00175-6 
  41. Haines BP, Rigby PWJ. Expression of the Lingo/LERN gene family during mouse embryogenesis. Gene Expr Patterns. 2008;8:79-86. https://doi.org/10.1016/j.modgep.2007.10.003 
  42. Li Y, Liu H, Yu Q, Liu H, Huang T, Zhao S, et al. Growth hormone promotes in vitro maturation of human oocytes. Front Endocrinol. 2019;10:485. https://doi.org/10.3389/fendo.2019.00485 
  43. Rogers NT, Halet G, Piao Y, Carroll J, Ko MSH, Swann K. The absence of a Ca2+ signal during mouse egg activation can affect parthenogenetic preimplantation development, gene expression patterns, and blastocyst quality. Reproduction. 2006;132:45-57. https://doi.org/10.1530/rep.1.01059 
  44. Li T, Shi Y, Wang P, Guachalla LM, Sun B, Joerss T, et al. Smg6/Est1 licenses embryonic stem cell differentiation via nonsense-mediated mRNA decay. EMBO J. 2015;34:1630-47. https://doi.org/10.15252/embj.201489947 
  45. MacLean JA 2nd, Rao MK, Doyle KMH, Richards JS, Wilkinson MF. Regulation of the Rhox5 homeobox gene in primary granulosa cells: preovulatory expression and dependence on SP1/SP3 and GABP. Biol Reprod. 2005;73:1126-34. https://doi.org/10.1095/biolreprod.105.042747 
  46. Okunade GW, Miller ML, Pyne GJ, Sutliff RL, O'Connor KT, Neumann JC, et al. Targeted ablation of plasma membrane Ca2+-ATPase (PMCA) 1 and 4 indicates a major housekeeping function for PMCA1 and a critical role in hyperactivated sperm motility and male fertility for PMCA4. J Biol Chem. 2004;279:33742-50. https://doi.org/10.1074/jbc.M404628200