Genetic Susceptibility to Ketosis in Cattle: Current State of Research

Мұқаба

Дәйексөз келтіру

Толық мәтін

Ашық рұқсат Ашық рұқсат
Рұқсат жабық Рұқсат берілді
Рұқсат жабық Тек жазылушылар үшін

Аннотация

High-yield productivity in dairy cows is due to intense functioning of all organs and organism systems, that predisposes animals to various forms of disorders of metabolic processes. Progress of energy disbalance in high-yield dairy cows during lactation contributes to the development of systemic metabolic disorders, negatively affecting milk production and reproductive potential of animals. Interest in breeding ketosis resistant cattle is global and finding of mutations, gene variants and molecular and genetic processes contributing to one or another phenotype are considered as key steps in understanding a degree of susceptibility to ketosis. These steps will also give an insight in etiology of ketosis and provide basis for designing novel effective breeding programs. In this paper we present an overview of studies based on genetic and molecular research methods in finding genetic markers of cattle ketosis development. We discuss comprehensive SNPs localization of GWAS meta-analysis data, protein-protein interactions of associated with SNPs candidate genes via STRING, as well as SNPs annotation of associated biological processes. We provide candidate gene expression profiles for associated with ketosis tissues based on human data with GTEx tool.

Авторлар туралы

O. Sokolova

Ural Federal Agrarian Scientific Research Centre, Ural Branch
of Russian Academy of Sciences

Хат алмасуға жауапты Автор.
Email: nauka_sokolova@mail.ru
Russia, 620142, Ekaterinburg

M. Bytov

Ural Federal Agrarian Scientific Research Centre, Ural Branch
of Russian Academy of Sciences

Email: nauka_sokolova@mail.ru
Russia, 620142, Ekaterinburg

A. Belousov

Ural Federal Agrarian Scientific Research Centre, Ural Branch
of Russian Academy of Sciences

Email: nauka_sokolova@mail.ru
Russia, 620142, Ekaterinburg

N. Bezborodova

Ural Federal Agrarian Scientific Research Centre, Ural Branch
of Russian Academy of Sciences

Email: nauka_sokolova@mail.ru
Russia, 620142, Ekaterinburg

V. Zubareva

Ural Federal Agrarian Scientific Research Centre, Ural Branch
of Russian Academy of Sciences

Email: nauka_sokolova@mail.ru
Russia, 620142, Ekaterinburg

N. Martynov

Ural Federal Agrarian Scientific Research Centre, Ural Branch
of Russian Academy of Sciences

Email: nauka_sokolova@mail.ru
Russia, 620142, Ekaterinburg

O. Zaitseva

Ural Federal Agrarian Scientific Research Centre, Ural Branch
of Russian Academy of Sciences

Email: nauka_sokolova@mail.ru
Russia, 620142, Ekaterinburg

I. Shkuratova

Ural Federal Agrarian Scientific Research Centre, Ural Branch
of Russian Academy of Sciences

Email: nauka_sokolova@mail.ru
Russia, 620142, Ekaterinburg

Әдебиет тізімі

  1. Berry D.P., Bermingham M.L., Good M.,More S.J. Genetics of animal health and disease in cattle // Irish Veterinary J. 2011. V. 64. № 5. P. 1–10. https://doi.org/10.1186/2046-0481-64-5
  2. Zinovieva N.A. Haplotypes affecting fertility in holstein cattle // Sel’skokhozyaistvennaya Biologiya. 2016. V. 51. P. 423–435. https://doi.org/10.15389/agrobiology.2016.4.423eng
  3. Brito L.F., Bedere N., Douhard F. et al. Review: Genetic selection of high-yielding dairy cattle toward sustainable farming systems in a rapidly changing world // Animal. 2021. V. 15. P. 1–14. https://doi.org/10.1016/j.animal.2021.100292
  4. Белоусов А.И., Красноперов А.С., Опарина О.Ю., Суздальцева М.А. Метаболические признаки алиментарного кетоза у высокопродуктивных коров // Труды ВИЭВ. 2018. Т. 80. № 1. С. 88–100. https://doi.org/10.30917/ATT-PRINT-2018-1
  5. Белоусов А.И., Соколова О.В., Беспамятных Е.Н. Применение биохимического скрининга при оценке продуктивного здоровья высокопродуктивных коров в Свердловской области // Вопр. нормативно-правового регулирования в ветеринарии. 2018. Т. 4. С. 278–280. https://doi.org/10.17238/issn2072-6023.2018.4.278
  6. Михайлова И.И., Евглевская Е.П., Михайлова О.И. и др. Патобиохимические изменения в метаболическом статусе высокопродуктивных коров // Ветеринарная патология. 2016. Т. 1. № 55. С. 75–80.
  7. Kessel S., Stroehl M., Meyer H.H.D. et al. Individual variability in physiological adaptation to metabolic stress during early lactation in dairy cows kept under equal conditions // J. Animal Sci. 2008. V. 86. № 11. P. 2903–2912. https://doi.org/10.2527/jas.2008-1016
  8. van Dorland H.A., Richter S., Morel I. et al. Variation in hepatic regulation of metabolism during the dry period and in early lactation in dairy cows // J. Dairy Sci. 2009. V. 92. № 5. P. 1924–1940. https://doi.org/10.3168/jds.2008-1454
  9. Ковалюк Н.В., Якушева Л.И., Кузьминова Е.В. и др. Связь полиморфизмов гена лептина с предрасположенностью крупного рогатого скота к кетозу // Генетика и разведение животных. 2020. Т. 3. С. 20–26. https://doi.org/10.31043/2410-2733-2020-3-20-26
  10. Kroezen V., Schenkel F.S., Miglior F. et al. Candidate gene association analyses for ketosis resistance in Holsteins // J. Dairy Sci. 2018. V. 101. № 6. P. 5240–5249. https://doi.org/10.3168/jds.2017-13374
  11. Huang H., Cao J., Hanif Q. et al. Genome-wide association study identifies energy metabolism genes for resistance to ketosis in Chinese Holstein cattle // Anim. Genet. 2019. V. 50. № 4. P. 376–380. https://doi.org/10.1111/age.12802
  12. Nayeri S., Schenkel F., Fleming A. et al. Genome-wide association analysis for β-hydroxybutyrate concentration in Milk in Holstein dairy cattle // BMC Genetics. 2019. V. 20. № 58. P. 1–17. https://doi.org/10.1186/s12863-019-0761-9
  13. Yan Z., Huang H., Freebern E. et al. Integrating RNA-Seq with GWAS reveals novel insights into the molecular mechanism underpinning ketosis in cattle // BMC Genomics. 2020. V. 21. № 489. P. 1–12. https://doi.org/10.1186/s12864-020-06909-z
  14. Wu Z.L., Chen S.Y., Qin C. et al. Clinical ketosis-associated alteration of gene expression in Holstein cows // Genes (Basel). 2020. V. 11. № 219. P. 1–11. https://doi.org/10.3390/genes11020219
  15. Mohsin M.A., Yu H., He R. et al. Differentiation of subclinical ketosis and liver function test indices in adipose tissues associated with hyperketonemia in postpartum dairy cattle // Front. Vet. Sci. 2021. V. 8. P. 1–14. https://doi.org/10.3389/fvets.2021.796494
  16. Loor J.J., Everts R.E., Bionaz M. et al. Nutrition-induced ketosis alters metabolic and signaling gene networks in liver of periparturient dairy cows // Physiol. Genomics. 2007. V. 32. № 1. P. 105–116. https://doi.org/10.1152/physiolgenomics.00188.2007
  17. Visscher P.M., Wray N.R., Zhang Q. et al. 10 Years of GWAS discovery: biology, function, and translation // Am. J. Hum. Genet. 2017. V. 101. № 1. P. 5–22. https://doi.org/10.1016/j.ajhg.2017.06.005
  18. Giacomini K.M., Yee S.W., Mushiroda T. et al. Genome-wide association studies of drug response and toxicity: An opportunity for genome medicine // Nat. Rev. Drug Discov. 2017. V. 16. № 1. P. 1–3. https://doi.org/10.1038/nrd.2016.234
  19. Shu L., Blencowe M., Yang X. Translating GWAS findings to novel therapeutic targets for coronary artery disease // Front. Cardiovasc. Med. 2018. V. 5. P. 1–9. https://doi.org/10.3389/fcvm.2018.00056
  20. Hillreiner M., Flinspach C., Pfaffl M.W., Kliem H. Effect of the ketone body beta-hydroxybutyrate on the innate defense capability of primary bovine mammary epithelial cells // PLoS One. 2016. V. 11. № 6. P. 1–18. https://doi.org/10.1371/journal.pone.0157774
  21. Esposito G., Irons P.C., Webb E.C., Chapwanya A. Interactions between negative energy balance, metabolic diseases, uterine health and immune response in transition dairy cows // Animal Reproduction Sci. 2014. V. 144. № 3. P. 60–71. https://doi.org/10.1016/j.anireprosci.2013.11.007
  22. Zhang S., Liu G., Xu C. et al. Perilipin 1 mediates lipid metabolism homeostasis and inhibits inflammatory cytokine synthesis in bovine adipocytes // Front. Immunol. 2018. V. 9. P. 1–14. https://doi.org/10.3389/fimmu.2018.00467
  23. Ha N.T., Gross J.J., van Dorland A. et al. Gene-based mapping and pathway analysis of metabolic traits in dairy cows // PLoS One. 2015. V. 10. № 3. P. 1–15. https://doi.org/10.1371/journal.pone.0122325
  24. Soares R.A.N., Vargas G., Muniz M.M.M. et al. Differential gene expression in dairy cows under negative energy balance and ketosis: A systematic review and meta-analysis // J. Dairy Sci. 2021. V. 104. № 1. P. 602–615. https://doi.org/10.3168/jds.2020-18883
  25. McLaren W., Gil L., Hunt S.E. et al. The Ensembl Variant Effect Predictor // Genome Biology. 2016. V. 17. № 1. P. 1–14. https://doi.org/10.1186/s13059-016-0974-4
  26. Huang D., Ovcharenko I. Identifying causal regulatory SNPs in ChIP-seq enhancers // Nucl. Acids Res. 2015. V. 43. № 1. P. 225–236. https://doi.org/10.1093/nar/gku1318
  27. Calvo S.E., Pagliarini D.J., Mootha V.K. Upstream open reading frames cause widespread reduction of protein expression and are polymorphic among humans // Proc. Natl Acad. Sci. USA. 2009. V. 106. № 18. P. 7507–7512. https://doi.org/10.1073/pnas.0810916106
  28. Chen J., Tian W. Explaining the disease phenotype of intergenic SNP through predicted long range regulation // Nucl. Acids Res. 2016. V. 44. № 18. P. 8641–8654. https://doi.org/10.1093/nar/gkw519
  29. Schmidt S.F., Larsen B.D., Loft A. et al. Acute TNF-induced repression of cell identity genes is mediated by NFκB-directed redistribution of cofactors from super-enhancers // Genome Research. 2015. V. 25. № 9. P. 1281–1294. https://doi.org/10.1101/gr.188300.114
  30. Vlahopoulos S.A. Aberrant control of NF-κB in cancer permits transcriptional and phenotypic plasticity, to curtail dependence on host tissue: molecular mode // Cancer Biology & Medicine. 2017. V. 14. № 3. P. 254–270. https://doi.org/10.20892/j.issn.2095-3941.2017.0029
  31. Birney E., Stamatoyannopoulos J.A., Dutta A. et al. Identification and analysis of functional elements in 1% of the human genome by the ENCODE pilot project // Nature. 2007. V. 447. P. 799–816. https://doi.org/10.1038/nature05874
  32. Mucaki E.J., Shirley B.C., Rogan P.K. Expression changes confirm genomic variants predicted to result in allele-specific, alternative mRNA splicing // Front. Genet. 2020. V. 11. P. 1–16. https://doi.org/10.3389/fgene.2020.00109
  33. Kalsotra A., Cooper T.A. Functional consequences of developmentally regulated alternative splicing // Nat. Rev. Genet. 2011. V. 12. № 10. P. 715–729. https://doi.org/10.1038/nrg3052
  34. Chasman D., Adams R.M. Predicting the functional consequences of non-synonymous single nucleotide polymorphisms: Structure-based assessment of amino acid variation // J. Mol. Biol. 2001. V. 307. № 2. P. 683–706. https://doi.org/10.1006/jmbi.2001.4510
  35. Dakal T.C., Kala D., Dhiman G. et al. Predicting the functional consequences of non-synonymous single nucleotide polymorphisms in IL8 gene // Scientific Reports. 2017. V. 7. № 1. P. 1–18. https://doi.org/10.1038/s41598-017-06575-4
  36. Sharma J., Keeling K.M., Rowe S.M. Pharmacological approaches for targeting cystic fibrosis nonsense mutations // Eur. J. Med. Chem. 2020. V. 200. P. 1–11. https://doi.org/10.1016/j.ejmech.2020.112436
  37. Petersen G.M., Parmigiani G., Thomas D. Missense mutations in Disease Genes: A Bayesian Approach to Evaluate Causality // Am. J. Human Genet. 1998. V. 62. № 6. P. 1516–1524. https://doi.org/10.1086/301871
  38. van der Velden A.W., Thomas A.A. The role of the 5' untranslated region of an mRNA in translation regulation during development // Int. J. Biochem. Cell Biol. 1999. V. 31. № 1. P. 87–106. https://doi.org/10.1016/s1357-2725(98)00134-4
  39. Jansen Ralf-P. mRNA localization: message on the move // Nature Reviews Mol. Cell Biol. 2001. V. 2. № 4. P. 247–256. https://doi.org/10.1038/35067016
  40. Bashirullah A., Cooperstock R.L., Lipshitz H.D. Spatial and temporal control of RNA stability // Proc. Natl Acad. Sci. USA. 2001. V. 98. № 13. P. 7025–7028. https://doi.org/10.1073/pnas.111145698
  41. Jiang H., Lucy M.C. Variants of the 5'-untranslated region of the bovine growth hormone receptor mRNA: Isolation, expression and effects on translational efficiency // Gene. 2001. V. 265. № 1. P. 45–53. https://doi.org/10.1016/s0378-1119(01)00356-0
  42. Hu L., Ma Y., Liu L. et al. Detection of functional polymorphisms in the hsp70 gene and association with cold stress response in Inner-Mongolia Sanhe cattle // Cell Stress and Chaperones. 2019. V. 24. № 2. P. 409–418. https://doi.org/10.1007/s12192-019-00973-5
  43. Dhamija S., Menon M.B. Non-coding transcript variants of protein-coding genes – what are they good for? // RNA Biology. 2018. V. 15. № 8. P. 1025–1031. https://doi.org/10.1080/15476286.2018.1511675
  44. Nelson C.D., Reinhardt T.A., Thacker T.C. et al. Modulation of the bovine innate immune response by production of 1α,25-dihydroxyvitamin D3 in bovine monocytes // J. Dairy Sci. 2010. V. 93. № 3. P. 1041–1049. https://doi.org/10.3168/jds.2009-2663
  45. Yu-fei S., Liu J., Wang X. et al. Essential role of the first intron in the transcription of hsp90β gene // FEBS Letters. 1997. V. 413. № 1. P. 92–98. https://doi.org/10.1016/S0014-5793(97)00883-1
  46. Jo B., Choi S.S. Introns: The functional benefits of introns in genomes // Genomics Inform. 2015. V. 13. № 4. P. 112–118. https://doi.org/10.5808/GI.2015.13.4.112
  47. Huang D., Chowdhury S., Wang H. et al. Multiomic analysis identifies CPT1A as a potential therapeutic target in platinum-refractory, high-grade serous ovarian cancer // Cell Reports Med. 2021. V. 2. № 12. P. 1–32. https://doi.org/10.1016/j.xcrm.2021.100471
  48. Ren Q., Guo M., Yang F. et al. Association of CPT1A gene polymorphism with the risk of gestational diabetes mellitus: A case-control study // J. Assist. Reprod. Genet. 2021. V. 38. № 7. P. 1861–1869. https://doi.org/10.1007/s10815-021-02143-y
  49. Mayr C. What are 3' UTRs doing? // Cold Spring Harb. Perspect. Biol. 2019. V. 11. № 10. P. 1–17. https://doi.org/10.1101/cshperspect.a034728
  50. Chekulaeva M., Landthaler M. Eyes on translation // Mol. Cell. 2016. V. 63. № 6. P. 918–925. https://doi.org/10.1016/j.molcel.2016.08.031
  51. Mayr C. Regulation by 3'-untranslated regions // Annu. Rev. Genet. 2017. V. 51. P. 171–194. https://doi.org/10.1146/annurev-genet-120116-024704
  52. Szklarczyk D., Gable A.L., Nastou K.C. et al. The STRING database in 2021: Customizable protein–protein networks, and functional characterization of user-uploaded gene/measurement sets // Nucl. Acids Res. 2021. V. 49. № D1. P. 605–612. https://doi.org/10.1093/nar/gkaa1074
  53. Freed A.S., Schwarz A.C., Brei B.K. et al. CHRNB1-associated congenital myasthenia syndrome: Expanding the clinical spectrum // Am. J. Med. Genetics. Part A. 2021. V. 185. № 3. P. 827–835. https://doi.org/10.1002/ajmg.a.62011
  54. Andre E., Beckerandre M. Expression of an N-terminally truncated form of human focal adhesion kinase in brain // Biochem. Biophys. Res. Communications. 1993. V. 190. № 1. P. 140–147. https://doi.org/10.1006/bbrc.1993.1022
  55. Li M., Zhong Di, Li G. Regulatory role of local tissue signal Del-1 in cancer and inflammation: a review // Cellular & Mol. Biol. Letters. 2021. V. 26. № 1. P. 1–12. https://doi.org/10.1186/s11658-021-00274-9
  56. Hsiao C.T., Cheng H.W., Huang C.M. et al. Fibronectin in cell adhesion and migration via N-glycosylation // Oncotarget. 2017. V. 8. № 41. P. 70653–70668. https://doi.org/10.18632/oncotarget.19969
  57. Dutta S., Mana-Capelli S., Paramasivam M. et al. TRIP6 inhibits Hippo signaling in response to tension at adherens junctions // EMBO Reports. 2018. V. 19. № 2. P. 337–350. https://doi.org/10.15252/embr.201744777
  58. Warfel J.D., Vandanmagsar B., Dubuisson O.S. et al. Examination of carnitine palmitoyltransferase 1 abundance in white adipose tissue: Implications in obesity research // Am. J. Physiol. Regul. Integr. Comp. Physiol. 2017. V. 312. № 5. P. 816–820. https://doi.org/10.1152/ajpregu.00520.2016
  59. Price N.T., Jackson V.N., Müller J. et al. Alternative exon usage in the single CPT1 gene of Drosophila generates functional diversity in the kinetic properties of the enzyme: Differential expression of alternatively spliced variants in Drosophila tissues // J. Biol. Chemistry. 2010. V. 285. № 11. P. 7857–7865. https://doi.org/10.1074/jbc.M109.072892
  60. Riancho J.A., Vázquez L., García-Pérez M.A. et al. Association of ACACB polymorphisms with obesity and diabetes // Mol. Genet. Metabolism. 2011. V. 104. № 4. P. 670–676. https://doi.org/10.1016/j.ymgme.2011.08.013
  61. Hellwege J.N., Stallings S., Torstenson E.S. et al. Heritability and genome-wide association study of benign prostatic hyperplasia (BPH) in the eMERGE network // Sci. Rep. 2019. V. 9. № 1. P. 1–10. https://doi.org/10.1038/s41598-019-42427-z
  62. Lao-On U., Cliff T.S., Dalton S., Jitrapakdee S. Pyruvate carboxylase supports basal ATP-linked respiration in human pluripotent stem cell-derived brown adipocytes // Biochem. Biophys. Res. Communications. 2021. V. 569. P. 139–146. https://doi.org/10.1016/j.bbrc.2021.06.096
  63. Rossi S.M., Konstantinidou G. Targeting long chain acyl-СоА synthetases for cancer therapy // Int. J. Mol. Sci. 2019. V. 20. № 15. https://doi.org/10.3390/ijms20153624
  64. Lee K., Kerner J., Hoppel C.L. Mitochondrial carnitine palmitoyltransferase 1a (CPT1a) is part of an outer membrane fatty acid transfer complex // J. Biol. Chem. 2011. V. 286. № 29. P. 25655–25662. https://doi.org/10.1074/jbc.M111.228692
  65. Nickkho-Amiry M., McVey R., Holland C. Peroxisome proliferator–activated receptors modulate proliferation and angiogenesis in human endometrial carcinoma // Mol. Cancer Res. 2012. V. 10. № 3. P. 441–453. https://doi.org/10.1158/1541-7786.MCR-11-0233
  66. Mostaghel E.A., Cho E., Zhang A. et al. Association of tissue abiraterone levels and SLCO genotype with intraprostatic steroids and pathologic response in men with high-risk localized prostate cancer // Clin. Cancer Res. 2017. V. 23. № 16. P. 4592–4601. https://doi.org/10.1158/1078-0432.CCR-16-2245
  67. Lu X., Chan T., Cheng Z. et al. The 5'-AMP-activated protein kinase regulates the function and expression of human organic anion transporting polypeptide 1A2 // Mol. Pharmacology. 2018. V. 94. № 6. P. 1–9. https://doi.org/10.1124/mol.118.113423
  68. Nayeri S., Stothard P. Tissues, metabolic pathways and genes of key importance in lactating dairy cattle // Springer Sci. Rev. 2016. V. 4. № 2. P. 49–77. https://doi.org/10.1007/s40362-016-0040-3
  69. Boyle E.I., Weng S., Gollub J. et al. GO::TermFinder – open source software for accessing Gene Ontology information and finding significantly enriched Gene Ontology terms associated with a list of genes // Bioinformatics. 2004. V. 20. № 18. P. 3710–3715. https://doi.org/10.1093/bioinformatics/bth456
  70. Supek F., Bošnjak M., Škunca N., Šmuc T. REVIGO summarizes and visualizes long lists of Gene Ontology terms // PLoS One. 2011. V. 6. № 7. P. 1–9. https://doi.org/10.1371/journal.pone.0021800
  71. Zhang G., Hailemariam D., Dervishi E. et al. Dairy cows affected by ketosis show alterations in innate immunity and lipid and carbohydrate metabolism during the dry off period and postpartum // Res. Veterinary Sci. 2016. V. 107. P. 246–256. https://doi.org/10.1016/j.rvsc.2016.06.012
  72. Gulinski P. Ketone bodies – causes and effects of their increased presence in cows’ body fluids: A review // Vet. World. 2021. V. 14. № 6. P. 1492–1503. https://doi.org/10.14202/vetworld.2021.1492-1503
  73. Wang Y., Gao Y., Xia C. et al. Pathway analysis of plasma different metabolites for dairy cow ketosis // Italian J. Animal Sci. 2016. V. 15. № 3. P. 545–551. https://doi.org/10.1080/1828051X.2016.1180643
  74. Zhang G., Ametaj B.N. Ketosis an old story under a new approach // Dairy. 2020. V. 1. № 1. P. 42–60. https://doi.org/10.3390/dairy1010005
  75. Dufour D.R., Lott J.A., Nolte F.S. et al. Diagnosis and monitoring of hepatic injury. II. Recommendations for use of laboratory tests in screening, diagnosis, and monitoring // Clin. Chem. 2000. V. 46. № 12. P. 2050–2068.https://doi.org/10.1093/clinchem/46.12.2050
  76. Tsukamoto K., Teramoto T. Carbohydrate and lipid metabolism in liver cirrhosis // Nihon Rinsho. 1994. V. 52. № 1. P. 150–158.
  77. Natesan V., Kim S.J. Lipid metabolism, disorders and therapeutic drugs – review // Biomol. Ther. (Seoul). 2021. V. 29. № 6. P. 596–604. https://doi.org/10.4062/biomolther.2021.122
  78. McCabe M., Waters S., Morris D. et al. RNA-seq analysis of differential gene expression in liver from lactating dairy cows divergent in negative energy balance // BMC Genomics. 2012. V. 13. P. 1–11. https://doi.org/10.1186/1471-2164-13-193
  79. Carithers L.J., Moore H.M. The Genotype-Tissue Expression (GTEx) project // Biopreserv. Biobank. 2015. V. 13. № 5. P. 307–308. https://doi.org/10.1038/ng.2653
  80. Goldinger A., Henders A.K., McRae A.F. et al. Genetic and nongenetic variation revealed for the principal components of human gene expression // Genetics. 2013. V. 195. № 3. P. 1117–1128. https://doi.org/10.1534/genetics.113.153221
  81. Genin E., Feingold J., Clerget-Darpoux F. Identifying modifier genes of monogenic disease: strategies and difficulties // Hum. Genet. 2008. V. 124. № 4. P. 357–368. https://doi.org/10.1007/s00439-008-0560-2
  82. Wright G.E.B., Caron N.S., Ng B. et al. Gene expression profiles complement the analysis of genomic modifiers of the clinical onset of Huntington disease // Hum. Mol. Genet. 2020. V. 29. № 16. P. 2788–2802. https://doi.org/10.1093/hmg/ddaa184
  83. Shahzad K., Lopreiato V., Liang Y. et al. Hepatic metabolomics and transcriptomics to study susceptibility to ketosis in response to prepartal nutritional management // J. Anim. Sci. Biotechnol. 2019. V. 10. P. 96. https://doi.org/10.1186/s40104-019-0404-z
  84. Pralle R.S., Li W., Murphy B.N. et al. Novel facets of the liver transcriptome are associated with the susceptibility and resistance to lipid-related metabolic disorders in periparturient Holstein cows // Animals (Basel). 2021. V. 11. № 9. P. 1–22. https://doi.org/10.3390/ani11092558
  85. Mezzetti M., Cattaneo L., Passamonti M.M. et al. The transition period updated: A review of the new insights into the adaptation of dairy cows to the new lactation // Dairy. 2021. V. 2. № 4. P. 617–636. https://doi.org/10.3390/dairy2040048
  86. Cuiyu Z., Chang Z., Jiang Z. et al. The relationship between insulin resistance and type II ketosis in dairy cows // Acta Scientiae Veterinariae. 2019. V. 47. № 1. P. 1–8. https://doi.org/10.22456/1679-9216.93425
  87. Klein S.L., Scheper C., Brügemann K. et al. Phenotypic relationships, genetic parameters, genome-wide associations, and identification of potential candidate genes for ketosis and fat-to-protein ratio in German Holstein cows // J. Dairy Sci. 2019. V. 102. № 7. P. 6276–6287. https://doi.org/10.3168/jds.2019-16237
  88. Heringstad B., Chang Y.M., Gianola D., Klemetsdal G. Genetic analysis of clinical mastitis, milk fever, ketosis, and retained placenta in three lactations of Norwegian red cows // J. Dairy Sci. 2005. V. 88. № 9. P. 3273–3281. https://doi.org/10.3168/jds.S0022-0302(05)73010-1
  89. Koeck A., Jamrozik J., Schenkel F.S. et al. Genetic analysis of milk β-hydroxybutyrate and its association with fat-to-protein ratio, body condition score, clinical ketosis, and displaced abomasum in early first lactation of Canadian Holsteins // J. Dairy Sci. 2014. V. 97. № 11. P. 7286–7292. https://doi.org/10.3168/jds.2014-8405
  90. Blanco-Gómez A., Castillo-Lluva S., Del Mar S.F.M. et al. Missing heritability of complex diseases: Enlightenment by genetic variants from intermediate phenotypes // BioEssays. 2016. V. 38. № 7. P. 664–673. https://doi.org/10.1002/bies.201600084
  91. Belay T.K., Svendsen M., Kowalski Z.M., Ådnøy T. Genetic parameters of blood β-hydroxybutyrate predicted from milk infrared spectra and clinical ketosis, and their associations with milk production traits in Norwegian Red cows // J. Dairy Sci. 2017. V. 100. № 8. P. 6298–6311. https://doi.org/10.3168/jds.2016-12458
  92. Oreland L., Lagravinese G., Toffoletto S. et al. Personality as an intermediate phenotype for genetic dissection of alcohol use disorder // J. Neural. Transm. (Vienna). 2018. V. 125. № 1. P. 107–130. https://doi.org/10.1007/s00702-016-1672-9
  93. Zuk O., Hechter E., Sunyaev S.R., Lander E.S. The mystery of missing heritability: Genetic interactions create phantom heritability // Proc. Natl Acad. Sci. USA. 2012. V. 109. № 4. P. 1193–1198. https://doi.org/10.1073/pnas.1119675109
  94. Marian A.J. Elements of missing heritability // Curr. Opin. Cardiol. 2012. V. 27. № 3. P. 197–201. https://doi.org/10.1097/HCO.0b013e328352707d
  95. van Calker D., Serchov T. The “missing heritability”–problem in psychiatry: Is the interaction of genetics, epigenetics and transposable elements a potential solution? // Neurosci. Biobehav. Rev. 2021. V. 126. P. 23–42. https://doi.org/10.1016/j.neubiorev.2021.03.019
  96. Manolio T.A., Collins F.S., Cox N.J. et al. Finding the missing heritability of complex diseases // Nature. 2009. V. 461. № 7265. P. 747–753. https://doi.org/10.1038/nature08494
  97. Genin E. Missing heritability of complex diseases: case solved? // Hum. Genet. 2020. V. 139. № 1. P. 103–113. https://doi.org/10.1007/s00439-019-02034-4
  98. Wagner G.P., Zhang J. The pleiotropic structure of the genotype-phenotype map: the evolvability of complex organisms // Nat. Rev. Genet. 2011. V. 12. № 3. P. 204–213. https://doi.org/10.1038/nrg2949
  99. Li Y., Huang J., Amos C.I. Genetic association analysis of complex diseases incorporating intermediate phenotype information // PLoS One. 2012. V. 7. № 10. P. 1–9. https://doi.org/10.1371/journal.pone.0046612
  100. Hackinger S., Zeggini E. Statistical methods to detect pleiotropy in human complex traits // Open Biol. 2017. V. 7. № 11. P. 1–13. https://doi.org/10.1098/rsob.170125
  101. Bone W.P., Siewert K.M., Jha A. et al. Multi-trait association studies discover pleiotropic loci between Alzheimer’s disease and cardiometabolic traits // Alzheimers Res. Ther. 2021. V. 13. P. 1–14. https://doi.org/10.1186/s13195-021-00773-z
  102. Якушева Л.И., Абрамов А.А., Ковалюк Н.В., Сацук В.Ф. Связь полиморфизмов R25C и A80V гена лептина быков-производителей с оценкой их дочерей на предрасположенность к возникновению кетоза // Сб. науч. трудов Краснодарского науч. центра по зоотехнии и ветеринарии. 2019. Т. 8. № 3. С. 24–27. https://doi.org/10.34617/y47d-6h82
  103. Komisarek J. Impact of LEP and LEPR gene polymorphisms on functional traits in Polish Holstein-Friesian cattle // Anim. Sci. Pap. Rep. 2010. V. 28.
  104. Mahmoudi A., Zargaran A., Amini H.R. et al. A SNP in the 3'-untranslated region of AMPKgamma1 may associate with serum ketone body and milk production of Holstein dairy cows // Gene. 2015. V. 574. № 1. P. 48–52. https://doi.org/10.1016/j.gene.2015.07.077
  105. Yang L., Bai J., Ju Z. et al. Effect of functional single nucleotide polymorphism g.-572 A>G of apolipoprotein A1 gene on resistance to ketosis in Chinese Holstein cows // Res. Vet. Sci. 2021. V. 135. P. 310–316. https://doi.org/10.1016/j.rvsc.2020.10.006
  106. Tetens J., Heuer C., Heyer I. et al. Polymorphisms within the APOBR gene are highly associated with milk levels of prognostic ketosis biomarkers in dairy cows // Physiol. Genomics. 2015. V. 47. № 4. P. 129–137. https://doi.org/10.1152/physiolgenomics.00126.2014

Қосымша файлдар

Қосымша файлдар
Әрекет
1. JATS XML
2.

Жүктеу (161KB)
3.

Жүктеу (692KB)
4.

Жүктеу (132KB)
5.

Жүктеу (189KB)

© О.В. Соколова, М.В. Бытов, А.И. Белоусов, Н.А. Безбородова, В.Д. Зубарева, Н.А. Мартынов, О.С. Зайцева, И.А. Шкуратова, 2023

Осы сайт cookie-файлдарды пайдаланады

Біздің сайтты пайдалануды жалғастыра отырып, сіз сайттың дұрыс жұмыс істеуін қамтамасыз ететін cookie файлдарын өңдеуге келісім бересіз.< / br>< / br>cookie файлдары туралы< / a>