مطالعه‌ی پویش کل ژنوم جهت شناسایی جایگاه‌های ژنتیکی مرتبط با بیماری یون در گاوهای هلشتاین ایران

نوع مقاله : مقاله پژوهشی

نویسندگان

1 گروه علوم دامی، دانشکدگان کشاورزی و منابع طبیعی دانشگاه تهران، کرج، ایران

2 گروه علوم دامی، دانشکدگان کشاورزی و منابع طبیعی دانشگاه تهران، کرج، ایران.

3 گروه ژنتیک و اصلاح دام، دانشکده کشاورزی و منابع طبیعی دانشگاه اراک، اراک، ایران

4 گروه ژنتیک واصلاح دام، پردیس بین المللی ارس دانشگاه تهران، جلفا، ایران.

5 گروه ژنتیک واصلاح دام، گروه علوم دامی، دانشکدگان کشاورزی و منابع طبیعی دانشگاه تهران، کرج، ایران

چکیده

پاراتوبرکلوزیس یک بیماری مسری، مزمن و روده­ای در نشخوارکنندگان است که در اثر عفونت زیرگونه مایکوباکتریوم آویوم پاراتوبرکلوزیس (MAP) ایجاد می­شود و خسارات اقتصادی فراوانی به صنعت پرورش دام  تحمیل  می­کند. در حال حاضر واکسن یا درمان موثری برای عفونت MAP  وجود ندارد، بنابراین بررسی مناطق ژنتیکی مرتبط با حساسیت به عفونت MAP می­تواند درک بهتری از مکانیسم‌های پاراتوبرکلوزیس ارائه دهد و به بهبود ژنتیکی حیوانات کمک کند. هدف این پژوهش شناسایی مناطق ژنومی و ژن‌های کاندیدا مرتبط با حساسیت به عفونت MAP در گاوهای شیری هلشتاین ایران با استفاده از روش پویش کل ژنوم (GWAS) بود. در این پژوهش،  از ۱۵۰ راس گاو نمونه گرفته شد. پس از استخراج DNA و سرم، نمونه­های DNA با استفاده از تراشه­های k30 (SNPchip30k) (شرکت ایلومینا) تعیین ژنوتیپ شدند. کنترل کیفیت نشانگرها براساس شاخص­های فراوانی آلل نادر (05/0PMAF <)، ژنوتیپ از دست رفته (05/0PMIND >)، نرخ تعیین ژنوتیپ (05/0PGENO >) و تعادل هاردی واینبرگ (PH-W < 1×10-6) توسط نرم­افزار PLINK انجام شد. بعد از  کنترل کیفیت، ۲۸۷۴۹ نشانگر روی ۱۴۲راس گاو (۹۹ راس بیمار و ۴۳ راس شاهد) برای ادامه آنالیز باقی ماند. پس از انجام آنالیز پویش ژنومی (GWAS) در نرم افزار PLINK ۱۶ نشانگر به عنوان نشانگر های معنی دار شناسایی شدند که بیشتر روی  کروموزوم­های ۳۰ و ۶ قرار داشتند. با بررسی بیوانفورماتیکی مناطق ژنومی معنی­­دار در پایگاه­های برخط  ensemble و genecards، ژن­های کاندیدای برای بیماری یون  شناسایی شد. مهم­ترین ژن­های شناسایی شده  LMX1A، THSD7A، ELMOD2، ATP6AP2، RNF150، SLIT3، SDE2، PARP1، PBX1، TFB2M، SMYD3 و PYCR2 بودند. آنالیز هستی­شناسی نشان داد که بیشتر این ژن­ها در سیستم عصبی، سیستم اسکلت سلولی، تنظیم سیتوکینز، دستگاه گلژی، فعالیت کاتالیزوری، انتقال یون کلسیم و مقاومت در برابر بیماری­ها نقش دارند. تجزیه و تحلیل پویش کل ژنوم  و آنالیز هستی شناسی می­تواند به شناسایی ژن­های کاندیدا و مناطق مرتبط با حساسیت بهMAP   در گاوهای شیری کمک ­کند که نقش مهمی در درمان و پیشگیری بیماری یون دارد .

کلیدواژه‌ها

موضوعات


عنوان مقاله [English]

Genome-wide association study to identify genomic regions associated with Johne's disease in Iranian Holstein cattle

نویسندگان [English]

  • younes doosti 1
  • Mohammad Moradi Shahrbabak 2
  • Hossein Moradi Shahrbabak 1
  • Hossein Mohammadi 3
  • Mahdi JavanNikkhah 4
  • Farnaz Arjmand Kermani 5
1 Department of Animal Sciences, College of Agriculture & Natural Resources, University of Tehran, Karaj, Iran
2 Department of Animal Sciences, College of Agriculture & Natural Resources, University of Tehran, Karaj, Iran
3 Department of Animal Sciences, Faculty of Agriculture and Natural Resources, Arak University, Arak, Iran
4 College of Agriculture &Natural Resources, University of Tehran, Ares International Campus, jolfa, Iran
5 College of Agriculture &Natural Resources, University of Tehran, Karaj, Iran
چکیده [English]

Paratuberculosis  known as Johne's disease, is a contagious, chronic, intestinal disease in ruminants, which is caused by Mycobacterium avium paratuberculosis subspecies (MAP) and causes a huge economic damage to the livestock industry. There is no effective treatment nor vaccine for MAP infection. Thus, investigating the genetic regions associated with susceptibility to MAP infection can provide a better understanding of paratuberculosis mechanisms and contribute to the genetic improvement of animals. The aim of this study was to identify genomic regions and candidate genes associated with susceptibility to MAP infection in Holstein dairy cattle using a genome-wide association study (GWAS). For this purpose, blood samples of 150 cows were collected, and DNA and serum of them were extracted.The prepared DNA samples were genotyped using bovine SNPchip30k (Illumina). Quality control of genotypes was performed based on the minor allele frequency (PMAF < 0.05), missing genotype (PMIND > 0.05), genotyping rate (PGENO > 0.05), and Hardy-Weinberg equilibrium (PH-W < 1) 10-6) using PLINK software. The association was performed using a mixed linear model in PLINK software. After quality control, 28749 markers on 142 cows (99 cases and 43 controls) were remained for the further analysis. Associations analysis showed that 16 markers located in chromosomes 30 and 6, were associated with  Johne's disease. Positional candidate genes for Johne's disease were identified. The most important identified genes were LMX1A, THSD7A, ELMOD2, ATP6AP2, RNF150, SLIT3, SDE2, PARP1, PBX1, TFB2M, SMYD3 and PYCR2. Gene ontology analysis showed that most of the identified genes are involved in the nervous system, cytoskeleton system, regulation of cytokines, Golgi apparatus, catalytic activity, calcium ion transport, and resistance to diseases. Whole genome-wide association study (GWAS) and ontology analysis (GO) can help to identify candidate genes and regions related to sensitivity to MAP in dairy cows, which can play an important role in the treatment and prevention of Johne's disease.

کلیدواژه‌ها [English]

  • Genotype
  • GWAS
  • Paratuberculosis
  • SNP
  • Somatic

Extended Abstract

Introduction and Objective

   Paratuberculosis is a contagious, chronic, intestinal disease in ruminants, which is caused by Mycobacterium avium paratuberculosis subspecies (MAP) infection and causes great economic damage to the livestock industry worldwide. This disease is one of the most important from the economic point of view and one of the most widespread diseases in terms of prevalence, which causes a decrease in fat and protein and an increase in milk somatic cells in dairy cows. There is no effective treatment for MAP infection or vaccine for this disease so far. Thus investigating the genetic regions related to susceptibility to MAP infection can provide a better understanding of paratuberculosis mechanisms and contribute to the genetic improvement of animals. The aim of this study was to identify genomic regions and candidate genes associated with susceptibility to MAP infection in Iranian Holstein dairy cattle using the whole genome-wide association study (GWAS).

 

Material and Methods

   For this purpose, blood samples of 150 cows in an industrial farm were collected, and DNA and serum of them were extracted. The prepared DNA samples were genotyped using k30 microarrays (SNPchip30k) (by Illumina). Quality control of genotypes based on rare allele frequency components (PMAF < 0.05), missing genotype (PMIND > 0.05), genotyping rate (PGENO > 0.05), and Hardy-Weinberg equilibrium (PH-W < 1) 10-6) and significance test were performed by PLINK software. Analysis of the ontology of genes was done by the online database https://www.Uniprot.org and finally, the ontology diagram of genes was drawn and analyzed by the online database PANTHER.

 

Results

   After quality control, 142 cows (99 cases and 43 controls) and 28749 markers were left for further analysis. Finally, 16 markers were considered higher than the significant threshold, and the most significant markers were located in chromosomes 30 and 6 respectively. Genes associated with selected markers were identified by ensemble and genecards sites. The most important identified genes related to these disease  were LMX1A, THSD7A, ELMOD2, ATP6AP2, RNF150, SLIT3, SDE2, PARP1, PBX1, TFB2M, SMYD3 and PYCR2. Ontology analysis showed that most of these genes are involved in  coding  proteins , cytoplasmic and membrane activities, the nervous system, cytoskeleton system, disease resistance, cytokine regulator, organ Golgi, catalytic activity, calcium ion transport. Also, some of these genes were related to other activities such as reproduction and fertility, mastitis, antiviral response, clinical ketosis, and immune response in dairy cows, as well as muscle growth and meat traits in beef cows.

 

Conclusion

     Whole genome-wide association study (GWAS) and ontology analysis (GO) can help to identify candidate genes and regions related to sensitivity to MAP in dairy cows, which can play an important role in the treatment and prevention of Johne's disease. The most important identified genes related to these disease  were LMX1A, THSD7A, ELMOD2, ATP6AP2, RNF150, SLIT3, SDE2, PARP1, PBX1, TFB2M, SMYD3 and PYCR2.

.

Baker, D.C., Barker, I.K., Brown, C.C.,Caswell, J.L., and Wittiams, K.J. (2007). Kennedy & Palmer's Pathology of Domestic Animals, Volume 2, 5th ed.W.B. Saunders, Philadelphia, p: 222-230.
Beaudeau, F., Belliard, M., Joly, A., & Seegers, H. (2007). Reduction in milk yield associated with Mycobacterium avium subspecies paratuberculosis (Map) infection in dairy cows. Veterinary research, 38(4), 625-634.
Ben-Jemaa, S., Senczuk, G., Ciani, E., Ciampolini, R., Catillo, G., Boussaha, M., & Mastrangelo, S. (2021). Genome-Wide Analysis Reveals Selection Signatures Involved in Meat Traits and Local Adaptation in Semi-Feral Maremmana Cattle. Frontiers in genetics, 12, 675569.‏
Bunel, A., Nivet, A. L., Blondin, P., Vigneault, C., Richard, F. J., & Sirard, M. A. (2020). The effects of LH inhibition with cetrorelix on cumulus cell gene expression during the luteal phase under ovarian coasting stimulation in cattle. Domestic Animal Endocrinology, 72, 106429.‏
Carignano, H. A., Roldan, D. L., Beribe, M. J., Raschia, M. A., Amadio, A., Nani, J. P., & Miretti, M. M. (2018). Genome-wide scan for commons SNPs affecting bovine leukemia virus infection level in dairy cattle. BMC genomics, 19(1), 1-15.‏
Chang, T., Xia, J., Xu, L., Wang, X., Zhu, B., Zhang, L., & Gao, H. (2018). A genome‐wide association study suggests several novel candidate genes for carcass traits in Chinese Simmental beef cattle. Animal genetics, 49(4), 312-316.‏
Collins, M. T., Gardner, I. A., Garry, F. B., Roussel, A. J., & Wells, S. J. (2006). Consensus recommendations on diagnostic testing for the detection of paratuberculosis in cattle in the United States. Journal of the American Veterinary Medical Association, 229(12), 1912-1919.
Dash, S., Singh, A., Dixit, S. P., & Kumar, A. (2022). Identification of Selection Signatures for Milk Performance Traits among Indigenous Dairy Cattle Breeds using High Density Genomic Information. Indian Journal of Animal Research, 1, 7.‏
De Matteis, G., Reale, A., Grandoni, F., Meyer-Ficca, M. L., Scatà, M. C., Meyer, R. G., & Moioli, B. (2018). Assessment of Poly (ADP-ribose) Polymerase1 (PARP1) expression and activity in cells purified from blood and milk of dairy cattle. Veterinary immunology and immunopathology, 202, 102-108.‏
De Vos, J. (2018). A Genome wide association study of carcass traits based on Real Time Ultrasound in South African Nguni cattle (Doctoral dissertation, University of Pretoria).‏
Douarre, P. E., Cashman, W., Buckley, J., Coffey, A., & O'Mahony, J. M. (2010). Isolation and detection of Mycobacterium avium subsp. paratuberculosis (MAP) from cattle in Ireland using both traditional culture and molecular based methods. Gut Pathogens, 2(1), 1-7.
Gao, Y., Jiang, J., Yang, S., Cao, J., Han, B., Wang, Y., & Sun, D. (2018). Genome-wide association study of Mycobacterium avium subspecies paratuberculosis infection in Chinese Holstein. BMC genomics, 19(1), 1-10.
Hermon-Taylor, J., & El-Zaatari, F. A. K. (2004). The Mycobacterium avium subspecies paratuberculosis problem and its relation to the causation of Crohn’s disease. Pathogenic Mycobacteria in Water.
Huo, J., Lu, S., Kwong, J. Q., Bround, M. J., Grimes, K. M., Sargent, M. A., & Molkentin, J. D. (2020). MCUb induction protects the heart from postischemic remodeling. Circulation research, 127(3), 379-390.‏
Jiang, Z., Kunej, T., Wibowo, T. A., Michal, J. J., Zhang, Z., Gaskins, C. T., & Wright Jr, R. W. (2006, August). The basal nucleus-encoded mitochondrial transcription genes and meat quality in beef cattle. In Proceedings of the 8th World Congress on Genetics Applied to Livestock Production (pp. 13-18). Belo Horizonte, Minas Gerais, Brazil, Instituto Prociencia.‏
Kaminski, S. (2019). Missense mutation in SDE2 gene–new lethal defect transmitted into Polish Holstein-Friesian cattle. Polish Journal of Veterinary Sciences, (3).‏
Kanamori, Y., Yamada, T., Asano, H., Kida, R., Qiao, Y., Abd Eldaim, M. A., & Funaba, M. (2014). Effects of vitamin a status on expression of ucp1 and brown/beige adipocyte-related genes in white adipose tissues of beef cattle. Journal of Veterinary Medical Science, 14-0137.‏
Kirkpatrick, B. W., & Shook, G. E. (2011). Genetic susceptibility to paratuberculosis. Veterinary Clinics: Food Animal Practice, 27(3), 559-571.‏
Kiser, J. N., White, S. N., Johnson, K. A., Hoff, J. L., Taylor, J. F., & Neibergs, H. L. (2017). Identification of loci associated with susceptibility to Mycobacterium avium subspecies paratuberculosis (Map) tissue infection in cattle. Journal of Animal Science, 95(3), 1080-1091.‏
Koets, A., Santema, W., Mertens, H., Oostenrijk, D., Keestra, M., Overdijk, M., & Rutten, V. P. M. G. (2010). Susceptibility to paratuberculosis infection in cattle is associated with single nucleotide polymorphisms in Toll-like receptor 2 which modulate immune responses against Mycobacterium avium subspecies paratuberculosis. Preventive veterinary medicine, 93(4), 305-315.‏
Kupper, J. D., Brandt, H. R., & Erhardt, G. (2014). Genetic association between NOD 2 polymorphism and infection status by M ycobacterium avium ssp. paratuberculosis in G erman H olstein cattle. Animal Genetics, 45(1), 114-116.‏
Lin, S., Wan, Z., Zhang, J., Xu, L., Han, B., & Sun, D. (2020). Genome-wide association studies for the concentration of albumin in colostrum and serum in Chinese Holstein. Animals, 10(12), 2211.‏
Mallikarjunappa, S., Brito, L. F., Pant, S. D., Schenkel, F. S., Meade, K. G., & Karrow, N. A. (2021). Johne's Disease in Dairy Cattle: An Immunogenetic Perspective. Frontiers in Veterinary Science, 8.‏
Mallikarjunappa, S., Sargolzaei, M., Brito, L. F., Meade, K. G., Karrow, N. A., & Pant, S. D. (2018). Uncovering quantitative trait loci associated with resistance to Mycobacterium avium ssp. paratuberculosis infection in Holstein cattle using a high-density single nucleotide polymorphism panel. Journal of dairy science, 101(8), 7280-7286.‏
Matteis, G., Grandoni, F., Zampieri, M., Reale, A., & Scatà, M. C. (2021). New Insights into the Significance of PARP-1 Activation: Flow Cytometric Detection of Poly (ADP-Ribose) as a Marker of Bovine Intramammary Infection. Cells, 10(3), 599.‏
Meszaros, G., Taferner, R., & Sölkner, J. (2016). Pleiotropic and epistatic interactions between stillbirth and calving ease in cattle. Acta Agriculture Slovenica, 5, 56.‏
Nielsen, S. S., & Toft, N. (2009). A review of prevalences of paratuberculosis in farmed animals in Europe. Preventive veterinary medicine, 88(1), 1-14.
Pant, S. D., Schenkel, F. S., Verschoor, C. P., You, Q., Kelton, D. F., Moore, S. S., & Karrow, N. A. (2010). A principal component regression based genome wide analysis approach reveals the presence of a novel QTL on BTA7 for MAP resistance in holstein cattle. Genomics, 95(3), 176-182.‏
Pant, S. D., Verschoor, C. P., Schenkel, F. S., You, Q., Kelton, D. F., & Karrow, N. A. (2014). Bovine CLEC7A genetic variants and their association with seropositivity in Johne's disease ELISA. Gene, 537(2), 302-307.‏
Pant, S. D., Verschoor, C. P., Schenkel, F. S., You, Q., Kelton, D. F., & Karrow, N. A. (2011). Bovine PGLYRP1 polymorphisms and their association with resistance to Mycobacterium avium ssp. paratuberculosis. Animal genetics, 42(4), 354-360.‏
Purcell, S., Neale, B., Todd-Brown, K., Thomas, L., Ferreira, M. A., Bender, D., & Sham, P. C. (2007). PLINK: a tool set for whole-genome association and population-based linkage analyses. The American journal of human genetics, 81(3), 559-575.
Radostits, O. M., Gay, C. C., Hinchcliff, K. W., & Constable, P. D. (2007). A textbook of the diseases of cattle, horses, sheep, pigs and goats. Veterinary medicine, 10, 2045-2050.
Ruiz-Larranaga, O., Garrido, J. M., Iriondo, M., Manzano, C., Molina, E., Montes, I., & Estonba, A. (2010). SP110 as a novel susceptibility gene for Mycobacterium avium subspecies paratuberculosis infection in cattle. Journal of Dairy Science, 93(12), 5950-5958.‏
Ruiz-Larranaga, O., Manzano, C., Iriondo, M., Garrido, J. M., Molina, E., Vazquez, P., & Estonba, A. (2011). Genetic variation of toll-like receptor genes and infection by Mycobacterium avium ssp. paratuberculosis in Holstein-Friesian cattle. Journal of dairy science, 94(7), 3635-3641.‏
Settles, M., Zanella, R., McKay, S. D., Schnabel, R. D., Taylor, J. F., Whitlock, R., & Neibergs, H. (2009). A whole genome association analysis identifies loci associated with Mycobacterium avium subsp. paratuberculosis infection status in US holstein cattle. Animal genetics, 40(5), 655-662.‏
Sevilla, I., Li, L., Amonsin, A., Garrido, J. M., Geijo, M. V., Kapur, V., & Juste, R. A. (2008). Comparative analysis of Mycobacterium avium subsp. paratuberculosis isolates from cattle, sheep and goats by short sequence repeat and pulsed-field gel electrophoresis typing. BMC Microbiology, 8(1), 1-8.
Smith, B. P. (2009). Large Animal Internal Medicine. Mosby Elsevier, St. Louis, MO, USA.
Soares, R. A. N., Vargas, G., Duffield, T., Schenkel, F., & Squires, E. J. (2021). Genome-wide association study and functional analyses for clinical and subclinical ketosis in Holstein cattle. Journal of Dairy Science, 104(9), 10076-10089.‏
Sohal, J.S., Singh S.V., Subhodh, S., Singh A.V., Singh, P.K., Sheoran, N. (2007): Mycobacteri  um avium subspecies paratuberculosis diagnosis and strain typing present status and future developments. Indian J Exp Biol. 45(10):843-52.
Tabatabayi, AH. and R. Firouzi. 2001. Disease of Animals due to Bacteria (3rd ed ed.). University of Tehran press.
Teo, Y. Y., Fry, A. E., Clark, T. G., Tai, E. S., & Seielstad, M. (2007). On the usage of HWE for identifying genotyping errors. Annals of Human genetics71(5), 701-703.
Thompson-Crispi, K. A., Sargolzaei, M., Ventura, R., Abo-Ismail, M., Miglior, F., Schenkel, F., & Mallard, B. A. (2014). A genome-wide association study of immune response traits in Canadian Holstein cattle. BMC genomics, 15(1), 1-10.‏
Van Hulzen, K. J. E., Koets, A. P., Nielen, M., Hoeboer, J., Van Arendonk, J. A. M., & Heuven, H. C. M. (2012). Genetic variation for infection status as determined by a specific antibody response against Mycobacterium avium subspecies paratuberculosis in milk of Dutch dairy goats. Journal of dairy science, 95(10), 6145-6151.‏
Van Hulzen, K. J. E., Schopen, G. C. B., van Arendonk, J. A. M., Nielen, M., Koets, A. P., Schrooten, C., & Heuven, H. C. M. (2012). Genome-wide association study to identify chromosomal regions associated with antibody response to Mycobacterium avium subspecies paratuberculosis in milk of Dutch Holstein-Friesians. Journal of Dairy Science, 95(5), 2740-2748.‏
Vansnick, E., Vercammen, F., Bauwens, L., D’Haese, E., Nelis, H., & Geysen, D. (2005). A survey for Mycobacterium avium subspecies paratuberculosis in the Royal Zoological Society of Antwerp. The Veterinary Journal170(2), 249-256.
Verschoor, C. P., Pant, S. D., You, Q., Schenkel, F. S., Kelton, D. F., & Karrow, N. A. (2010). Polymorphisms in the gene encoding bovine interleukin-10 receptor alpha are associated with Mycobacterium avium ssp. paratuberculosis infection status. BMC genetics, 11(1), 1-9.‏
Zare, Y., Shook, G. E., Collins, M. T., & Kirkpatrick, B. W. (2014). Heritability estimates for susceptibility to Mycobacterium avium subspecies paratuberculosis infection defined by ELISA and fecal culture test results in Jersey cattle. Journal of Dairy Science, 97(7), 4562-4567.‏
Dickinson, R. E., Dallol, A., Bieche, I., Krex, D., Morton, D., Maher, E. R., & Latif, F. (2004). Epigenetic inactivation of SLIT3 and SLIT1 genes in human cancers. British journal of cancer, 91(12), 2071-2078.‏
Dubos, A., Castells-Nobau, A., Meziane, H., Oortveld, M. A., Houbaert, X., Iacono, G., & Herault, Y. (2015). Conditional depletion of intellectual disability and Parkinsonism candidate gene ATP6AP2 in fly and mouse induces cognitive impairment and neurodegeneration. Human molecular genetics, 24(23), 6736-6755.‏
Litonin, D., Sologub, M., Shi, Y., Savkina, M., Anikin, M., Falkenberg, M., & Temiakov, D. (2010). Human mitochondrial transcription revisited: only TFAM and TFB2M are required for transcription of the mitochondrial genes in vitro. Journal of Biological Chemistry, 285(24), 18129-18133.‏
Scholz, B. A., Sumida, N., de Lima, C. D. M., Chachoua, I., Martino, M., Tzelepis, I., & Ohlsson, R. (2019). WNT signaling and AHCTF1 promote oncogenic MYC expression through super-enhancer-mediated gene gating. Nature genetics, 51(12), 1723-1731.‏
Zou, H. L., Su, C. J., Shi, M., Zhao, G. Y., Li, Z. Y., Guo, C., & Ding, Y. Q. (2009). Expression of the LIM-homeodomain gene Lmx1a in the postnatal mouse central nervous system. Brain research bulletin, 78(6), 306-312.‏v
Deng, W., Wu, J., Zheng, W., Wang, Q., Li, D., & Kuang, H. (2023). RNF150 suppresses papillary thyroid carcinoma with ASK1 ubiquitination presenting a direct target via inactivating p38 signaling axis. Cell Biology International.‏