Skip to main content
Log in

Comparative CpG methylation kinetic patterns of cis-regulatory regions of heat stress–related genes in Sahiwal and Frieswal cattle upon persistent heat stress

  • Original Paper
  • Published:
International Journal of Biometeorology Aims and scope Submit manuscript

Abstract

The kinetic patterns of CpG methylation of the cis-regulatory region of heat stress–related genes on exposed to heat stress (at 42 °C) between the Sahiwal and Frieswal cattle was compared in the present study. Using an in vitro whole blood culture model, cells were continuously exposed to heat stress (at 42 °C) for 6 h. Methylation levels of five genes, viz., GPX1, HSP70, HSP90, c-FOS, and JUN were estimated by SyberGreen-based quantitative methylation-specific PCR (qMSP) assay. CpG methylation kinetics at different time points of heat stress (0.5, 1, 2, 4, 6 h) were analyzed using mixed ANOVA. The initial methylation level, estimated at 37 °C, of HSP70 was significantly high in the Sahiwal breed. A significant (p<0.001) time-dependent hypomethylation of an antioxidant gene (GPX1) CpG islands was detected at the acute phase of the stress. Heat shock protein gene (HSP70) showed a similar CpG methylation kinetics where the hypomethylation was prominent from 1 h and persisted up to 4 h. The heat stress responses of both Sahiwal and Frieswal cattle were identical as there was no distinctiveness in the methylation kinetics of CpG islands of studied genes. The acclimatization of Frieswal cattle—a breed developed in India over the years to the tropical climatic conditions, maybe one of the reasons for this similarity. Thus, the present study results could pave a path to understand the molecular mechanism of heat stress and adaptation of indigenous and crossbred cattle populations to the changing scenario in tropical climate conditions.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2

Similar content being viewed by others

Data availability

Data has been presented within the manuscript. The datasets used and/or analyzed during the current study also available from the corresponding author on reasonable request.

References

  • Bahrami S, Drabløs F (2016) Gene regulation in the immediate-early response process Advances in Biological Regulation Gene regulation in the immediate-early response process. Adv Biol Regul. 62:37–49. https://doi.org/10.1016/j.jbior.2016.05.001

    Article  CAS  Google Scholar 

  • Bartels A, Han Q, Nair P, Stacey L, Gaynier H, Mosley M, Huang Q, Pearson J, Hsieh TF, An YQ, Xiao W (2018) Dynamic DNA methylation in plant growth and development. Int J Mol Sci 19:2144

    Article  Google Scholar 

  • Brenet F, Moh M, Funk P, Feierstein E, Viale AJ, Socci ND, Scandura JM (2011) DNA methylation of the first exon is tightly linked to transcriptional silencing. PLoS One 6:e14524

    Article  CAS  Google Scholar 

  • Broekman MMTJ, Roelofs HMJ, Hoentjen F, Wiegertjes R, Stoel N, Joosten LA, de Jong DJ, Wanten GJA (2015) LPS-stimulated whole blood cytokine production is not related to disease behavior in patients with quiescent Crohn’s disease. PLoS One 10:1–12. https://doi.org/10.1371/journal.pone.0133932

    Article  CAS  Google Scholar 

  • Cho YG, Chang X, Park I-S et al (2011) Promoter methylation of leukemia inhibitory factor receptor gene in colorectal carcinoma. Int J Oncol 39:337–344

    CAS  Google Scholar 

  • Dalcin VC, Fischer V, Daltro D d S et al (2016) Physiological parameters for thermal stress in dairy cattle. Rev Bras Zootec 45:458–465. https://doi.org/10.1590/S1806-92902016000800006

    Article  Google Scholar 

  • Damsgaard CT, Lauritzen L, Calder PC, Kjær TMR, Frøkiær H (2009) Whole-blood culture is a valid low-cost method to measure monocytic cytokines - A comparison of cytokine production in cultures of human whole-blood, mononuclear cells and monocytes. J Immunol Methods 340:95–101. https://doi.org/10.1016/j.jim.2008.10.005

    Article  CAS  Google Scholar 

  • Darst RP, Pardo CE, Ai L, et al (2010) Bisulfite sequencing of DNA. Curr Protoc Mol Biol Chapter 7:Unit 7.9.1-17. doi: https://doi.org/10.1002/0471142727.mb0709s91

  • Davidović RS, Božović AM, Mandušić VL, Krajnović MM (2014) Methylation-specific PCR : four steps in primer design. 9:1127–1139. doi: https://doi.org/10.2478/s11535-014-0324-z

  • de Barros FRO, Paula-Lopes FF (2018) Cellular and epigenetic changes induced by heat stress in bovine preimplantation embryos. Mol Reprod Dev 85:810–820

    Article  Google Scholar 

  • Diederich M, Hansmann T, Heinzmann J, Barg-Kues B, Herrmann D, Aldag P, Baulain U, Reinhard R, Kues W, Weißgerber C, Haaf T, Niemann H (2012) DNA methylation and mRNA expression profiles in bovine oocytes derived from prepubertal and adult donors. Reproduction 144:319–330. https://doi.org/10.1530/REP-12-0134

    Article  CAS  Google Scholar 

  • Duan J, Jiang Z, Alqahtani F et al (2019) Methylome dynamics of bovine gametes and in vivo early embryos. Front Genet 10:512

    Article  CAS  Google Scholar 

  • Ducrocq V, Laloe D, Swaminathan M, Rognon X, Tixier-Boichard M, Zerjal T (2018) Genomics for ruminants in developing countries: From principles to practice. Front Genet 9:1–7. https://doi.org/10.3389/fgene.2018.00251

    Article  Google Scholar 

  • Fang X, Zhao Z, Yu H, Li G, Jiang P, Yang Y, Yang R, Yu X (2017) Comparative genome-wide methylation analysis of longissimus dorsi muscles between Japanese black (Wagyu) and Chinese Red Steppes cattle. PLoS One 12:e0182492

    Article  Google Scholar 

  • Feil R, Fraga MF (2012) Epigenetics and the environment: emerging patterns and implications. Nat Rev Genet 13:97–109

    Article  CAS  Google Scholar 

  • Gao F, Luo Y, Li S, Li J, Lin L, Nielsen AL, Sørensen CB, Vajta G, Wang J, Zhang X, du Y, Yang H, Bolund L (2011) Comparison of gene expression and genome-wide DNA methylation profiling between phenotypically normal cloned pigs and conventionally bred controls. PLoS One 6:e25901

    Article  CAS  Google Scholar 

  • Hao Y, Cui Y, Gu X (2016) Genome-wide DNA methylation profiles changes associated with constant heat stress in pigs as measured by bisulfite sequencing. Sci Rep 6:22–25. https://doi.org/10.1038/srep27507

    Article  CAS  Google Scholar 

  • Hernández HG, Tse MY, Pang SC, Arboleda H, Forero DA (2013) Optimizing methodologies for PCR-based DNA methylation analysis. Biotechniques 55:181–197

    Article  Google Scholar 

  • Horowitz M (2016) Epigenetics and cytoprotection with heat acclimation. J Appl Physiol 120:702–710

    Article  CAS  Google Scholar 

  • Hsieh HJ, Li NQ, Frangos JA (1993) Pulsatile and steady flow induces c-fos expression in human endothelial cells. J Cell Physiol 154:143–151

    Article  CAS  Google Scholar 

  • Jablonka E, Raz G (2009) Transgenerational epigenetic inheritance: prevalence, mechanisms, and implications for the study of heredity and evolution. Q Rev Biol 84:131–176

    Article  Google Scholar 

  • Jost JP, Saluz HP (1993) Steroid hormone dependent changes in DNA methylation and its significance for the activation or silencing of specific genes. DNA Methylation. Springer, In, pp 425–451

    Google Scholar 

  • Kisliouk T, Cramer T, Meiri N (2017) Methyl CpG level at distal part of heat-shock protein promoter HSP70 exhibits epigenetic memory for heat stress by modulating recruitment of POU2F1-associated nucleosome-remodeling deacetylase (NuRD) complex. J Neurochem 141:358–372. https://doi.org/10.1111/jnc.14014

    Article  CAS  Google Scholar 

  • Kuroda A, Rauch TA, Todorov I, Ku HT, al-Abdullah IH, Kandeel F, Mullen Y, Pfeifer GP, Ferreri K (2009) Insulin gene expression is regulated by DNA methylation. PLoS One 4:e6953

    Article  Google Scholar 

  • Livak KJ, Schmittgen TD (2001) Analysis of relative gene expression data using real-time quantitative PCR and the 2-ΔΔCT method. Methods 25:402–408. https://doi.org/10.1006/meth.2001.1262

    Article  CAS  Google Scholar 

  • Lorincz MC, Dickerson DR, Schmitt M, Groudine M (2004) Intragenic DNA methylation alters chromatin structure and elongation efficiency in mammalian cells. Nat Struct Mol Biol 11:1068–1075

    Article  CAS  Google Scholar 

  • Lowdon RF, Jang HS, Wang T (2016) Evolution of epigenetic regulation in vertebrate genomes. Trends Genet 32:269–283

    Article  CAS  Google Scholar 

  • Miozzo F, Sabéran-Djoneidi D, Mezger V (2015) HSFs, stress sensors and sculptors of transcription compartments and epigenetic landscapes. J Mol Biol 427:3793–3816

    Article  CAS  Google Scholar 

  • Nguyen TTT, Bowman PJ, Haile-Mariam M, Pryce JE, Hayes BJ (2016) Genomic selection for tolerance to heat stress in Australian dairy cattle. J Dairy Sci 99:2849–2862. https://doi.org/10.3168/jds.2015-9685

    Article  CAS  Google Scholar 

  • Renaudeau D, Collin A, Yahav S, de Basilio V, Gourdine JL, Collier RJ (2012) Adaptation to hot climate and strategies to alleviate heat stress in livestock production. Animal 6:707–728

    Article  CAS  Google Scholar 

  • Runcie DE, Garfield DA, Babbitt CC et al (2012) Genetics of gene expression responses to temperature stress in a sea urchin gene network. Mol Ecol 21:4547–4562

    Article  CAS  Google Scholar 

  • Sengar GS, Deb R, Singh U, Junghare V, Hazra S, Raja TV, Alex R, Kumar A, Alyethodi RR, Kant R, Jakshara S, Joshi CG (2018a) Identification of differentially expressed microRNAs in Sahiwal (Bos indicus) breed of cattle during thermal stress. Cell Stress Chaperones. 23:1019–1032. https://doi.org/10.1007/s12192-018-0911-4

    Article  CAS  Google Scholar 

  • Sengar GS, Deb R, Singh U, Raja TV, Kant R, Sajjanar B, Alex R, Alyethodi RR, Kumar A, Kumar S, Singh R, Jakhesara SJ, Joshi CG (2018b) Differential expression of microRNAs associated with thermal stress in Frieswal (Bos taurus x Bos indicus) crossbred dairy cattle. Cell Stress Chaperones 23:155–170. https://doi.org/10.1007/s12192-017-0833-6

    Article  CAS  Google Scholar 

  • Sevane N, Martínez R, Bruford MW (2019) Genome-wide differential DNA methylation in tropically adapted Creole cattle and their Iberian ancestors. Anim Genet 50:15–26. https://doi.org/10.1111/age.12731

    Article  CAS  Google Scholar 

  • Silberman DM, Acosta GB, Zubilete MAZ (2016) Long-term effects of early life stress exposure: Role of epigenetic mechanisms. Pharmacol Res 109:64–73

    Article  CAS  Google Scholar 

  • Skibiel AL, Peñagaricano F, Amorin R et al (2018) In utero heat stress alters the offspring epigenome. Sci Rep 8:14609

    Article  CAS  Google Scholar 

  • Verma N, Alyethodi RR, Kathuria A, Alex R, Hussain S, Singh U, Tyagi S, Sirohi AS, Kumar S, Deb R, Sengar GS, Raja TV, Prakash B (2020) Effect of heat stress on superoxide anion production in native and crossbred cattle under in vitro whole blood culture model. J Therm Biol 87:102457

    Article  CAS  Google Scholar 

  • Vinoth A, Thirunalasundari T, Shanmugam M, Uthrakumar A, Suji S, Rajkumar U (2018) Evaluation of DNA methylation and mRNA expression of heat shock proteins in thermal manipulated chicken. Cell Stress Chaperones 23:235–252

    Article  CAS  Google Scholar 

  • Wu J, Zhang W, Li C (2020) Recent Advances in Genetic and Epigenetic Modulation of Animal Exposure to High Temperature. Front Genet 11. https://doi.org/10.3389/fgene.2020.00653

  • Xu Q, Zhang Y, Sun D, Wang Y, Yu Y (2007) Analysis on DNA methylation of various tissues in chicken. Anim Biotechnol 18:231–241

    Article  CAS  Google Scholar 

  • Zemach A, McDaniel IE, Silva P, Zilberman D (2010) Genome-wide evolutionary analysis of eukaryotic DNA methylation. Science (80) 328:916–919

  • Zhu Y, Lu L, Liao X, Li W, Zhang L, Ji C, Lin X, Liu HC, Odle J, Luo X (2017) Maternal dietary manganese protects chick embryos against maternal heat stress via epigenetic-activated antioxidant and anti-apoptotic abilities. Oncotarget 8:89665–89680

    Article  Google Scholar 

Download references

Acknowledgements

The authors are thankful to Director, Central Institute for Research on Cattle, Meerut, India, for providing the necessary facilities for conducting the study. We also acknowledge the Director, Frieswal for providing the biological samples.

Funding

The ICAR supported the research under Institutional funding and work is done as part of MSc dissertation work.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Rafeeque R. Alyethodi.

Ethics declarations

Competing of Interest

None of the authors' have any competing interests, including financial and non-financial.

Ethical approval and consent to participate

Research has been approved by the Institute Animal Ethics Committee (IAEC) of the ICAR-Central Institute for Research on Cattle, Meerut, UP, India, under CPCSEA guidelines.

Code availability

Not applicable

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Verma, N., Alyethodi, R.R., Kathuria, A. et al. Comparative CpG methylation kinetic patterns of cis-regulatory regions of heat stress–related genes in Sahiwal and Frieswal cattle upon persistent heat stress. Int J Biometeorol 65, 1025–1032 (2021). https://doi.org/10.1007/s00484-021-02084-2

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00484-021-02084-2

Keywords

Navigation