Bioinformatics analysis of differentially gene expression profiles related to heat stress in brain, liver, and leg muscle of broiler chickens based on microarray technique

Document Type : Research Paper

Authors

1 Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran

2 Department of Animal Sciences, Faculty of Agriculture, University of Tabriz, Tabriz, Iran

3 Department of Animal Science, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran

4 Iranian Fisheries Science Research Institute, Agricultural Research Education and Extension Organization (AREEO), Ahvaz, Iran

5 Agri-Food and Biosciences Institute, Hillsborough, UK and Assistant professor, School of Biological Sciences, Queen’s University Belfast, Belfast, UK

Abstract

In the poultry industry, the heat stress caused by high environmental temperature has a negative influence on broiler chicken performance and has become a major challenge. Transcriptome profile analysis of the data and identification of patterns of differential gene expression in related tissues can be involved in the discovery of molecular mechanisms resistant to heat stress. The main purpose of this study was to use transcriptome profiles of three tissues brain, liver, and leg muscle of two groups of the control and heat stress broiler chickens to identify candidate genes associated with heat stress. By the analysis of microarray data to express the gene differences, 657 significant genes (P<0.05) were extracted, which a total of 94 genes showed significant expression differences (FDR < 0.05, Fold change > ± 2). Then, by studying the ontology of the relevant genes resulting from data analysis and literature mining as well as the reconstructed protein-protein interaction network, hub genes including NSDHL, DHCR24, LSS, FDPS, PCK1, ACTA1, HSP90AA1, HSPA2, HSPB1, HSF1, CRYAB, APOB, and IL6 were identified. Annotation results of these genes indicated that they have a role in the main process of metabolic and signaling pathways related to the ion transport system, steroid, antibodies, cholesterol biosynthesis, lipid metabolism, immune system function, and various signaling pathways such as MAP kinase, RET, and ERK. Overall, the present study can provide new insights into evidence of the pathways activated by these genes to identify effective genes and a better understanding of biological processes related to heat stress.

Keywords

Main Subjects


Extended Abstract

Introduction

Transcriptome analysis is an important and valuable tool for identifying genes and their function in the mechanism of action of heat stress and for identifying the inherent genetic mechanisms to deal with it. Transcriptome analysis is actually a method of determining and identifying gene activity and expression. Using the resulting gene expression pattern, it is possible to uncover how biological systems are regulated at the transcriptional level. Therefore, the main objective of the current research is to identify candidate genes, using the analysis of samples based on microarray technology, in relation to the three tissues brain, liver and thigh muscles in two groups of broilers under control and under heat stress, to verify their expression levels as significant expression in the control group compared to those under heat stress.

 

Materials and Methods

This dataset contains the expression information of three tissues of brain, liver and thigh muscles belonging to the research of China in 2012. For each tissue, a total of 6 specimens (three specimens as control (at a temperature of 281 °C in the growth hall) and three specimens under heat stress (at a temperature of 401 °C in the growth hall), totaling 18 specimens, were used. Analysis of microarray data on expression of gene differences extracted 657 significant genes (P<0.05). with a total of 94 genes that show significant differences in expression (FDR < 0.05, fold change > 2). Subsequently, the ontology of the relevant genes emerging from the data analysis and literature review as well as the reconstructed protein-protein interaction network is examined.

 

Results and discussion

Interestingly, the results of this work highlighted different hub genes NSDHL, DHCR24, LSS, FDPS, PCK1, ACTA1, HSP90AA1, HSPA2, HSPB1, HSF1, CRYAB, APOB and IL6. Annotation results of these genes indicate that they play a role in the main process of metabolic and signaling pathways related to ion transport system, steroids, antibodies, cholesterol biosynthesis, lipid metabolism, immune system function and various signaling pathways such as MAP kinase. RET and ERK.

 

Conclusion

In general, identified genes (particularly hub genes) from data analysis and resource review in various metabolic and signaling pathways related to the ion transport system, steroid, antibody and cholesterol biosynthesis, lipid metabolism, immune system function, and various signaling pathways. Like MAP kinase, RET, and ERK play a role that may help improve our understanding of the important role of the three tissues brain, liver, and thigh muscles in performance and resilience to thermal stress in poultry and provide important molecular evidence for this association The level of gene expression in these tissues, along with other layers of omics, may lead to genetic enhancement of this trait in broilers and breeding strategies in the poultry industry.

Adu-Asiamah, P., Zhang, Y., Amoah, K., Leng, Q.Y., Zheng, J.H., Yang, H., Zhang, W.L. & Zhang, L. (2021). Evaluation of physiological and molecular responses to acute heat stress in two chicken breeds. Animal, 15(2), 100106.
Åkerfelt, M., Morimoto, R. I., & Sistonen, L. (2010). Heat shock factors: integrators of cell stress, development and lifespan. Nature Reviews Molecular Cell Biology, 11(8), 545-555.
Altan, Ö.Z.G.E., Pabuçcuoğlu, A., Altan, A., Konyalioğlu, S. & Bayraktar, H. (2003). Effect of heat stress on oxidative stress, lipid peroxidation and some stress parameters in broilers. British Poultry Science, 44(4), 545-550.
Al-Zghoul, M.B., El-Bahr, S.M., Al-Rukibat, R.K., Abd Elhafeed, S.D., Althnaian, T.A. & Al-Ramadan, S.Y. (2015). Biochemical and molecular investigation of thermal manipulation protocols during broiler embryogenesis and subsequent thermal challenge. BMC Veterinary Research, 11(1), 1-9.
Barrett, N.W., Rowland, K., Schmidt, C.J., Lamont, S.J., Rothschild, M.F., Ashwell, C.M. & Persia, M.E. (2019). Effects of acute and chronic heat stress on the performance, egg quality, body temperature, and blood gas parameters of laying hens. Poultry Science, 98(12), 6684-6692.
Bigland, C.H. & Triantaphyllopoulos, D.C. (1961). Chicken prothrombin, thrombin, and fibrinogen. American Journal of Physiology-Legacy Content, 200(5), 1013-1017.
Brede, M., Nagy, G. b., Philipp, M., Sørensen, J. B., Lohse, M. J., & Hein, L. (2003). Differential control of adrenal and sympathetic catecholamine release by α2-adrenoceptor subtypes. Molecular Endocrinology, 17(8), 1640-1646.
Chang, Y., Östling, P., Åkerfelt, M., Trouillet, D., Rallu, M., Gitton, Y., El Fatimy, R., Fardeau, V., Le Crom, S., Morange, M. & Sistonen, L. (2006). Role of heat-shock factor 2 in cerebral cortex formation and as a regulatorof p35 expression. Genes & Development, 20(7), 836-847.
Chen, X., Li, R., & Geng, Z. (2015). Cold stress initiates the Nrf2/UGT1A1/L-FABP signaling pathway in chickens. Poultry Science, 94(11), 2597-2603.
Cheung, A.S., de Rooy, C., Levinger, I., Rana, K., Clarke, M.V., How, J.M., Garnham, A., McLean, C., Zajac, J.D., Davey, R.A. & Grossmann, M. (2017). Actin alpha cardiac muscle 1 gene expression is upregulated in the skeletal muscle of men undergoing androgen deprivation therapy for prostate cancer. The Journal of Steroid Biochemistry and Molecular Biology, 174, 56-64.
Ciocca, D. R., Cappello, F., Cuello-Carrion, E., & Arrigo, A. P. (2015). Molecular approaches to target heat shock proteins for cancer treatment. Frontiers in Clinical Drug Research-Anti-Cancer Agents, 2, 3-47.
Coble, D.J., Fleming, D., Persia, M.E., Ashwell, C.M., Rothschild, M.F., Schmidt, C.J. & Lamont, S.J. (2014). RNA-seq analysis of broiler liver transcriptome reveals novel responses to high ambient temperature. BMC Genomics, 15(1), 1-12.
Creagh, E., Sheehan, D., & Cotter, T. (2000). Heat shock proteins–modulators of apoptosis in tumour cells. Leukemia, 14(7), 1161-1173.
Cunningham, D., Swartzlander, D., Liyanarachchi, S., Davuluri, R.V. & Herman, G.E. (2005). Changes in gene expression associated with loss of function of the NSDHL sterol dehydrogenase in mouse embryonic fibroblasts. Journal of Lipid Research, 46(6), 1150-1162.
Davis, S. & Meltzer, P.S. (2007). GEOquery: a bridge between the Gene Expression Omnibus (GEO) and BioConductor. Bioinformatics, 23(14), 1846-1847.
de Graauw, M., Tijdens, I., Cramer, R., Corless, S., Timms, J.F. & Van De Water, B. (2005). Heat shock protein 27 is the major differentially phosphorylated protein involved in renal epithelial cellular stress response and controls focal adhesion organization and apoptosis. Journal of Biological Chemistry, 280(33), 29885-29898.
Dietschy, J. M., & Wilson, J. D. (1970). Regulation of cholesterol metabolism. New England Journal of Medicine, 282(22), 1241-1249.
Du, P., Kibbe, W.A. & Lin, S.M. (2008). Lumi: a pipeline for processing Illumina microarray. Bioinformatics, 24(13), 1547-1548.
Fujimoto, M., Hayashida, N., Katoh, T., Oshima, K., Shinkawa, T., Prakasam, R., Tan, K., Inouye, S., Takii, R. & Nakai, A. (2010). A novel mouse HSF3 has the potential to activate nonclassical heat-shock genes during heat shock. Molecular Biology of the Cell, 21(1), 106-116.
Gautier, L., Cope, L., Bolstad, B.M. & Irizarry, R.A. (2004). Affy-analysis of Affymetrix GeneChip data at the probe level. Bioinformatics, 20(3), 307-315.
Ghafouri, F., Bahrami, A., Sadeghi, M., Miraei-Ashtiani, S.R., Bakherad, M., Barkema, H.W. & Larose, S. (2021). Omics multi-layers networks provide novel mechanistic and functional insights into fat storage and lipid metabolism in poultry. Frontiers in Genetics, 12.
Groenendijk, B. C., Van der Heiden, K., Hierck, B. P., & Poelmann, R. E. (2007). The role of shear stress on ET-1, KLF2, and NOS-3 expression in the developing cardiovascular system of chicken embryos in a venous ligation model. Physiology, 22(6), 380-389.
Hietbrink, F., Koenderman, L., Rijkers, G. T., & Leenen, L. P. (2006). Trauma: the role of the innate immune system. World Journal of Emergency Surgery, 1(1), 15.
Huber, W., Carey, V.J., Gentleman, R., Anders, S., Carlson, M., Carvalho, B.S., Bravo, H.C., Davis, S., Gatto, L., Girke, T. & Gottardo, R. (2015). Orchestrating high-throughput genomic analysis with Bioconductor. Nature Methods, 12(2), 115-121.
Huising, M., Van Schooten, C., Taverne-Thiele, A., Hermsen, T., Verburg-van Kemenade, B., & Flik, G. (2004). Structural characterisation of a cyprinid (Cyprinus carpio L.) CRH, CRH-BP and CRH-R1, and the role of these proteins in the acute stress response. Journal of Molecular Endocrinology, 32(3), 627-648.
IPCC (Intergovernmental Panel on Climate change) (2007) Climate Change 2007: The Physical Science Basis, Contribution of Working Group I to the Fourth Assessment Report of the Intergovernmental Panel on Climate Change. Available: http://www.ipcc.ch/publications_and_data/publications_ipcc_fourth_assessment_report_wg1_report_the_physical_science_basis.htm. Accessed 2009 Sept 30.
Javadi Esfehani, Y. (2014). CEP78, a novel centrosomal protein.
John Peter, A. T., Herrmann, B., Antunes, D., Rapaport, D., Dimmer, K. S., & Kornmann, B. (2017). Vps13-Mcp1 interact at vacuole–mitochondria interfaces and bypass ER–mitochondria contact sites. Journal of Cell Biology, 216(10), 3219-3229.
Judge, L.M., Perez-Bermejo, J.A., Truong, A., Ribeiro, A.J., Yoo, J.C., Jensen, C.L., Mandegar, M.A., Huebsch, N., Kaake, R.M., So, P.L. & Srivastava, D. (2017). A BAG3 chaperone complex maintains cardiomyocyte function during proteotoxic stress. JCI Insight, 2(14).
Krishnamurthy, K., Kanagasabai, R., Druhan, L. J., & Ilangovan, G. (2011). Small Heat Shock Proteins and Doxorubicin-Induced Oxidative Stress in the Heart. In Studies on Experimental Models, Springer, 105-130.
Kumar, V., Sinha, A. K., Romano, N., Allen, K. M., Bowman, B. A., Thompson, K. R., & Tidwell, J. H. (2018). Metabolism and nutritive role of cholesterol in the growth, gonadal development, and reproduction of crustaceans. Reviews in Fisheries Science & Aquaculture, 26(2), 254-273.
Laing, N.G., Dye, D.E., Wallgren‐Pettersson, C., Richard, G., Monnier, N., Lillis, S., Winder, T.L., Lochmüller, H., Graziano, C., Mitrani‐Rosenbaum, S. & Twomey, D. (2009). Mutations and polymorphisms of the skeletal muscle α‐actin gene (ACTA1). Human Mutation, 30(9), 1267-1277.
Lara, L.J. & Rostagno, M.H. (2013). Impact of heat stress on poultry production. Animals, 3(2), 356-369.
Lee, M., Park, H., Heo, J.M., Choi, H.J. & Seo, S. (2021). Multi-tissue transcriptomic analysis reveals that L-methionine supplementation maintains the physiological homeostasis of broiler chickens than D-methionine under acute heat stress. Plos One, 16(1), e0246063.
Li, J., Xing, S., Zhao, G., Zheng, M., Yang, X., Sun, J., Wen, J. & Liu, R. (2020). Identification of diverse cell populations in skeletal muscles and biomarkers for intramuscular fat of chicken by single-cell RNA sequencing. BMC Genomics, 21(1), 1-11.
Li, Y., Zeng, Y., Mooney, S. M., Yin, B., Mizokami, A., Namiki, M., & Getzenberg, R. H. (2011). Resistance to paclitaxel increases the sensitivity to other microenvironmental stresses in prostate cancer cells. Journal of Cellular Biochemistry, 112(8), 2125-2137.
Liu, J., Zhao, H., Wang, Y., Shao, Y., Zong, H., Zeng, X., & Xing, M. (2019). Arsenic trioxide and/or copper sulfate induced apoptosis and autophagy associated with oxidative stress and perturbation of mitochondrial dynamics in the thymus of Gallus gallus. Chemosphere, 219, 227-235.
Lu, Z., He, X.F., Ma, B.B., Zhang, L., Li, J.L., Jiang, Y., Zhou, G.H. & Gao, F. (2019). Increased fat synthesis and limited apolipoprotein B cause lipid accumulation in the liver of broiler chickens exposed to chronic heat stress. Poultry Science, 98(9), 3695-3704.
Luo, Q.B., Song, X.Y., Ji, C.L., Zhang, X.Q. & Zhang, D.X. (2014). Exploring the molecular mechanism of acute heat stress exposure in broiler chickens using gene expression profiling. Gene, 546(2), 200-205.
McInnes, L., Healy, J. & Melville, J. (2018). Umap: Uniform manifold approximation and projection for dimension reduction. arXiv preprint arXiv:1802.03426.
Mutryn, M.F., Brannick, E.M., Fu, W., Lee, W.R. & Abasht, B. (2015). Characterization of a novel chicken muscle disorder through differential gene expression and pathway analysis using RNA-sequencing. BMC Genomics, 16(1), 1-19.
Nawaz, A.H., Amoah, K., Leng, Q.Y., Zheng, J.H., Zhang, W.L. & Zhang, L. (2021). Poultry Response to Heat Stress: Its Physiological, Metabolic, and Genetic Implications on Meat Production and Quality Including Strategies to Improve Broiler Production in a Warming World. Frontiers in Veterinary Science, 814.
Ohba, K., Sasaki, S., Matsushita, A., Iwaki, H., Matsunaga, H., Suzuki, S., Ishizuka, K., Misawa, H., Oki, Y. & Nakamura, H. (2011). GATA2 mediates thyrotropin-releasing hormone-induced transcriptional activation of the thyrotropin β gene. PLoS ONE, 6(4), e18667.
Osei-Amponsah, R., Chauhan, S.S., Leury, B.J., Cheng, L., Cullen, B., Clarke, I.J. & Dunshea, F.R. (2019). Genetic selection for thermotolerance in ruminants. Animals, 9(11), 948.
Patwari, P., Emilsson, V., Schadt, E.E., Chutkow, W.A., Lee, S., Marsili, A., Zhang, Y., Dobrin, R., Cohen, D.E., Larsen, P.R. & Zavacki, A.M. (2011). The arrestin domain-containing 3 protein regulates body mass and energy expenditure. Cell Metabolism, 14(5), 671-683.
Pawar, S.S., Sajjanar, B., Lonkar, V.D., Kurade, N.P., Kadam, A.S., Nirmal, A.V., Brahmane, M.P. & Bal, S.K. (2016). Assessing and mitigating the impact of heat stress in poultry. Adv. Anim. Vet. Sci, 4(6), 332-341.
Pearson, G., Robinson, F., Beers Gibson, T., Xu, B.e., Karandikar, M., Berman, K., & Cobb, M. H. (2001). Mitogen-activated protein (MAP) kinase pathways: regulation and physiological functions. Endocrine Reviews, 22(2), 153-183.
Qin, S., Minami, Y., Kurosaki, T., & Yamamura, H. (1997). Distinctive functions of Syk and Lyn in mediating osmotic stress-and ultraviolet C irradiation-induced apoptosis in chicken B cells. Journal of Biological Chemistry, 272(29), 17994-17999.
Rabindran, S.K., Giorgi, G., Clos, J. & Wu, C. (1991). Molecular cloning and expression of a human heat shock factor, HSF1. Proceedings of the National Academy of Sciences, 88(16), 6906-6910.
Rezaei Sinaki, M., Sadeghi, M., Bahrami, A., & Moradi Shahrbabak, M. (2020). Identification of genes, biological pathways and signaling affecting heat stress with ‎microarray data sets in poultry. Iranian Journal of Animal Science, 51(3), 243-251. (In Farsi)
Ritchie, M.E., Phipson, B., Wu, D.I., Hu, Y., Law, C.W., Shi, W. & Smyth, G.K. (2015). limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Research, 43(7), e47-e47.
Romanelli, M.G., Lorenzi, P., Sangalli, A., Diani, E. & Mottes, M. (2009). Characterization and functional analysis of cis-acting elements of the human farnesyl diphosphate synthetase (FDPS) gene 5′ flanking region. Genomics, 93(3), 227-234.
Shannon, P., Markiel, A., Ozier, O., Baliga, N.S., Wang, J.T., Ramage, D., Amin, N., Schwikowski, B. & Ideker, T. (2003). Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Research, 13(11), 2498-2504.
Soleimani, A.F., Zulkifli, I., Omar, A.R. & Raha, A.R. (2011). Physiological responses of 3 chicken breeds to acute heat stress. Poultry Science, 90(7), 1435-1440.
Song, X.Y., Luo, Q.B. & Zhang, X.Q. (2012). Gene Expression Profiling of Three Tissues in Chicken with Heat Stress by Affymetrix Microarray. Available online at: https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE23592
St-Pierre, N.R., Cobanov, B. & Schnitkey, G. (2003). Economic losses from heat stress by US livestock industries. Journal of Dairy Science, 86, 52-77.
Szklarczyk, D., Gable, A.L., Lyon, D., Junge, A., Wyder, S., Huerta-Cepas, J., Simonovic, M., Doncheva, N.T., Morris, J.H., Bork, P. & Jensen, L.J. (2019). STRING v11: protein–protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Research, 47(D1), 607-613.
Tang, S., Yin, B., Song, E., Chen, H., Cheng, Y., Zhang, X., Bao, E. & Hartung, J. (2016). Aspirin upregulates αB-Crystallin to protect the myocardium against heat stress in broiler chickens. Scientific Reports, 6(1), 1-8.
Wang, Y., Jia, X., Hsieh, J.C., Monson, M.S., Zhang, J., Shu, D., Nie, Q., Persia, M.E., Rothschild, M.F. & Lamont, S.J. (2021). Transcriptome Response of Liver and Muscle in Heat-Stressed Laying Hens. Genes, 12(2), 255.
Weber, A., Wasiliew, P. & Kracht, M. (2010). Interleukin-1 (IL-1) pathway. Science Signaling, 3(105).
Wolc, A., Arango, J., Settar, P., Fulton, J.E., O’Sullivan, N.P. & Dekkers, J.C.M. (2019). Genome wide association study for heat stress induced mortality in a white egg layer line. Poultry Science, 98(1), 92-96.
Wu, S., Guo, W., Liang, S., Lu, H., Sun, W., Ren, X., Sun, Q. & Yang, X. (2018). Systematic analysis of the regulatory roles of microRNAs in postnatal maturation and metergasis of liver of breeder cocks. Scientific reports, 8(1), 1-14.
Xie, J., Tang, L., Lu, L., Zhang, L., Xi, L., Liu, H.C., Odle, J. & Luo, X. (2014). Differential expression of heat shock transcription factors and heat shock proteins after acute and chronic heat stress in laying chickens (Gallus gallus). PloS One, 9(7), e102204.
Xu, J., Yin, B., Huang, B., Tang, S., Zhang, X., Sun, J., & Bao, E. (2019). Co-enzyme Q10 protects chicken hearts from in vivo heat stress via inducing HSF1 binding activity and Hsp70 expression. Poultry Science, 98(2), 1002-1011.
Zhang, J., Lv, C., Mo, C., Liu, M., Wan, Y., Li, J. & Wang, Y. (2021). Single-Cell RNA Sequencing Analysis of Chicken Anterior Pituitary: A Bird’s-Eye View on Vertebrate Pituitary. Frontiers in Physiology, 12.
Zhang, Q., Shi, H., Liu, W., Wang, Y., Wang, Q. & Li, H. (2013). Differential expression of L-FABP and L-BABP between fat and lean chickens. Genet Mol Res, 12(4), 4192-4206.
Zhang, W., Kong, L., Zhang, X., & Luo, Q. (2014). Alteration of HSF3 and HSP70 mRNA expression in the tissues of two chicken breeds during acute heat stress. Genet Mol Res, 13(4).
Zhao, P., Guo, Y., Zhang, W., Chai, H., Xing, H. & Xing, M. (2017). Neurotoxicity induced by arsenic in Gallus Gallus: regulation of oxidative stress and heat shock protein response. Chemosphere, 166, 238-245.
Zhao, Y., Wong, L. & Goh, W.W.B. (2020). How to do quantile normalization correctly for gene expression data analyses. Scientific Reports, 10(1), 1-11.