79 research outputs found

    New insight into the SSC8 genetic determination of fatty acid composition in pigs

    Get PDF
    [EN] Background:Fat content and fatty acid composition in swine are becoming increasingly studied because of their effect on sensory and nutritional quality of meat. A QTL (quantitative trait locus) for fatty acid composition in backfat was previously detected on porcine chromosome 8 (SSC8) in an Iberian x Landrace F-2 intercross. More recently, a genome-wide association study detected the same genomic region for muscle fatty acid composition in an Iberian x Landrace backcross population. ELOVL6, a strong positional candidate gene for this QTL, contains a polymorphism in its promoter region (ELOVL6:c.-533C < T), which is associated with percentage of palmitic and palmitoleic acids in muscle and adipose tissues. Here, a combination of single-marker association and the haplotype-based approach was used to analyze backfat fatty acid composition in 470 animals of an Iberian x Landrace F2 intercross genotyped with 144 SNPs (single nucleotide polymorphisms) distributed along SSC8. Results:Two trait-associated SNP regions were identified at 93 Mb and 119 Mb on SSC8. The strongest statistical signals of both regions were observed for palmitoleic acid (C16:1(n-7)) content and C18:0/C16:0 and C18:1(n-7)/C16:1 (n-7) elongation ratios. MAML3 and SETD7 are positional candidate genes in the 93 Mb region and two novel microsatellites in MAML3 and nine SNPs in SETD7 were identified. No significant association for the MAML3 microsatellite genotypes was detected. The SETD7:c. 700G > T SNP, although statistically significant, was not the strongest signal in this region. In addition, the expression of MAML3 and SETD7 in liver and adipose tissue varied among animals, but no association was detected with the polymorphisms in these genes. In the 119 Mb region, the ELOVL6:c.-533C > T polymorphism showed a strong association with percentage of palmitic and palmitoleic fatty acids and elongation ratios in backfat. Conclusions:Our results suggest that the polymorphisms studied in MAML3 and SETD7 are not the causal mutations for the QTL in the 93 Mb region. However, the results for ELOVL6 support the hypothesis that the ELOVL6:c.-533C > T polymorphism has a pleiotropic effect on backfat and intramuscular fatty acid composition and that it has a role in the determination of the QTL in the 119 Mb region.This work was funded by MICINN AGL2008-04818-C03/GAN and MINECO AGL2011-29821-C02 and the Innovation Programme Consolider-Ingenio 2010 (CSD2007-00036). M. Revilla is a Master's student of Animal Breeding and Biotechnology of Reproduction (Polytechnical University of Valencia and Autonomous University of Barcelona). Y. Ramayo-Caldas was funded by a FPU grant (AP2008-01450), J. Corominas by a FPI scholarship from the Ministry of Education (BES-2009-018223) and A. Puig-Oliveras by a PIF scholarship (458-01-1/2011). This manuscript has been proofread by Chuck Simons, a native English speaking university instructor in English.Revilla, M.; Ramayo-Caldas, Y.; Castelló, A.; Corominas, J.; Puig-Oliveras, A.; Ibañez Escriche, N.; Muñoz, M.... (2014). New insight into the SSC8 genetic determination of fatty acid composition in pigs. Genetics Selection Evolution. 46. https://doi.org/10.1186/1297-9686-46-28S46Clarke, R., Frost, C., Collins, R., Appleby, P., & Peto, R. (1997). Dietary lipids and blood cholesterol: quantitative meta-analysis of metabolic ward studies. BMJ, 314(7074), 112-112. doi:10.1136/bmj.314.7074.112Mensink, R. P., & Katan, M. B. (1992). Effect of dietary fatty acids on serum lipids and lipoproteins. A meta-analysis of 27 trials. Arteriosclerosis and Thrombosis: A Journal of Vascular Biology, 12(8), 911-919. doi:10.1161/01.atv.12.8.911Hunter, J. E., Zhang, J., & Kris-Etherton, P. M. (2009). Cardiovascular disease risk of dietary stearic acid compared with trans, other saturated, and unsaturated fatty acids: a systematic review. The American Journal of Clinical Nutrition, 91(1), 46-63. doi:10.3945/ajcn.2009.27661Astrup, A., Dyerberg, J., Elwood, P., Hermansen, K., Hu, F. B., Jakobsen, M. U., … Willett, W. C. (2011). The role of reducing intakes of saturated fat in the prevention of cardiovascular disease: where does the evidence stand in 2010? The American Journal of Clinical Nutrition, 93(4), 684-688. doi:10.3945/ajcn.110.004622Harris, W. S., Poston, W. C., & Haddock, C. K. (2007). Tissue n−3 and n−6 fatty acids and risk for coronary heart disease events. Atherosclerosis, 193(1), 1-10. doi:10.1016/j.atherosclerosis.2007.03.018Lopez-Huertas, E. (2010). Health effects of oleic acid and long chain omega-3 fatty acids (EPA and DHA) enriched milks. A review of intervention studies. Pharmacological Research, 61(3), 200-207. doi:10.1016/j.phrs.2009.10.007Guo, T., Ren, J., Yang, K., Ma, J., Zhang, Z., & Huang, L. (2009). Quantitative trait loci for fatty acid composition in longissimus dorsi and abdominal fat: results from a White Duroc × Erhualian intercross F2population. Animal Genetics, 40(2), 185-191. doi:10.1111/j.1365-2052.2008.01819.xUemoto, Y., Soma, Y., Sato, S., Ishida, M., Shibata, T., Kadowaki, H., … Suzuki, K. (2011). Genome-wide mapping for fatty acid composition and melting point of fat in a purebred Duroc pig population. Animal Genetics, 43(1), 27-34. doi:10.1111/j.1365-2052.2011.02218.xClop, A., Ovilo, C., Perez-Enciso, M., Cercos, A., Tomas, A., Fernandez, A., … Noguera, J. L. (2003). Detection of QTL affecting fatty acid composition in the pig. Mammalian Genome, 14(9), 650-656. doi:10.1007/s00335-002-2210-7Ramayo-Caldas, Y., Mercadé, A., Castelló, A., Yang, B., Rodríguez, C., Alves, E., … Folch, J. M. (2012). Genome-wide association study for intramuscular fatty acid composition in an Iberian × Landrace cross1. Journal of Animal Science, 90(9), 2883-2893. doi:10.2527/jas.2011-4900Muñoz, M., Rodríguez, M. C., Alves, E., Folch, J. M., Ibañez-Escriche, N., Silió, L., & Fernández, A. I. (2013). Genome-wide analysis of porcine backfat and intramuscular fat fatty acid composition using high-density genotyping and expression data. BMC Genomics, 14(1), 845. doi:10.1186/1471-2164-14-845Ramos, A. M., Crooijmans, R. P. M. A., Affara, N. A., Amaral, A. J., Archibald, A. L., Beever, J. E., … Groenen, M. A. M. (2009). Design of a High Density SNP Genotyping Assay in the Pig Using SNPs Identified and Characterized by Next Generation Sequencing Technology. PLoS ONE, 4(8), e6524. doi:10.1371/journal.pone.0006524Estellé, J., Mercadé, A., Pérez-Enciso, M., Pena, R. N., Silió, L., Sánchez, A., & Folch, J. M. (2009). Evaluation ofFABP2as candidate gene for a fatty acid composition QTL in porcine chromosome 8. Journal of Animal Breeding and Genetics, 126(1), 52-58. doi:10.1111/j.1439-0388.2008.00754.xEstellé, J., Fernández, A. I., Pérez-Enciso, M., Fernández, A., Rodríguez, C., Sánchez, A., … Folch, J. M. (2009). A non-synonymous mutation in a conserved site of theMTTPgene is strongly associated with protein activity and fatty acid profile in pigs. Animal Genetics, 40(6), 813-820. doi:10.1111/j.1365-2052.2009.01922.xPurcell, S., Neale, B., Todd-Brown, K., Thomas, L., Ferreira, M. A. R., 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. doi:10.1086/519795Pérez-Enciso, M., & Misztal, I. (2011). Qxpak.5: Old mixed model solutions for new genomics problems. BMC Bioinformatics, 12(1). doi:10.1186/1471-2105-12-202Storey, J. D., & Tibshirani, R. (2003). Statistical significance for genomewide studies. Proceedings of the National Academy of Sciences, 100(16), 9440-9445. doi:10.1073/pnas.1530509100Druet, T., & Georges, M. (2009). A Hidden Markov Model Combining Linkage and Linkage Disequilibrium Information for Haplotype Reconstruction and Quantitative Trait Locus Fine Mapping. Genetics, 184(3), 789-798. doi:10.1534/genetics.109.108431Werle, E., Schneider, C., Renner, M., Völker, M., & Fiehn, W. (1994). Convenient single-step, one tube purification of PCR products for direct sequencing. Nucleic Acids Research, 22(20), 4354-4355. doi:10.1093/nar/22.20.4354Ballester, M., Cordón, R., & Folch, J. M. (2013). DAG Expression: High-Throughput Gene Expression Analysis of Real-Time PCR Data Using Standard Curves for Relative Quantification. PLoS ONE, 8(11), e80385. doi:10.1371/journal.pone.0080385Karim, L., Takeda, H., Lin, L., Druet, T., Arias, J. A. C., Baurain, D., … Coppieters, W. (2011). Variants modulating the expression of a chromosome domain encompassing PLAG1 influence bovine stature. Nature Genetics, 43(5), 405-413. doi:10.1038/ng.814Oyama, T., Harigaya, K., Sasaki, N., Okamura, Y., Kokubo, H., Saga, Y., … Kitagawa, M. (2011). Mastermind-like 1 (MamL1) and mastermind-like 3 (MamL3) are essential for Notch signaling in vivo. Development, 138(23), 5235-5246. doi:10.1242/dev.062802Pajvani, U. B., Qiang, L., Kangsamaksin, T., Kitajewski, J., Ginsberg, H. N., & Accili, D. (2013). Inhibition of Notch uncouples Akt activation from hepatic lipid accumulation by decreasing mTorc1 stability. Nature Medicine, 19(8), 1054-1060. doi:10.1038/nm.3259Syreeni, A., El-Osta, A., Forsblom, C., Sandholm, N., Parkkonen, M., … Tarnow, L. (2011). Genetic Examination of SETD7 and SUV39H1/H2 Methyltransferases and the Risk of Diabetes Complications in Patients With Type 1 Diabetes. Diabetes, 60(11), 3073-3080. doi:10.2337/db11-0073Chakrabarti, S. K., Francis, J., Ziesmann, S. M., Garmey, J. C., & Mirmira, R. G. (2003). Covalent Histone Modifications Underlie the Developmental Regulation of Insulin Gene Transcription in Pancreatic β Cells. Journal of Biological Chemistry, 278(26), 23617-23623. doi:10.1074/jbc.m303423200Ramayo-Caldas, Y., Mach, N., Esteve-Codina, A., Corominas, J., Castelló, A., Ballester, M., … Folch, J. M. (2012). Liver transcriptome profile in pigs with extreme phenotypes of intramuscular fatty acid composition. BMC Genomics, 13(1), 547. doi:10.1186/1471-2164-13-547Corominas, J., Ramayo-Caldas, Y., Puig-Oliveras, A., Pérez-Montarelo, D., Noguera, J. L., Folch, J. M., & Ballester, M. (2013). Polymorphism in the ELOVL6 Gene Is Associated with a Major QTL Effect on Fatty Acid Composition in Pigs. PLoS ONE, 8(1), e53687. doi:10.1371/journal.pone.005368

    Genome-wide linkage analysis of QTL for growth and body composition employing the PorcineSNP60 BeadChip

    Get PDF
    [EN] Background: The traditional strategy to map QTL is to use linkage analysis employing a limited number of markers. These analyses report wide QTL confidence intervals, making very difficult to identify the gene and polymorphisms underlying the QTL effects. The arrival of genome-wide panels of SNPs makes available thousands of markers increasing the information content and therefore the likelihood of detecting and fine mapping QTL regions. The aims of the current study are to confirm previous QTL regions for growth and body composition traits in different generations of an Iberian x Landrace intercross (IBMAP) and especially identify new ones with narrow confidence intervals by employing the PorcineSNP60 BeadChip in linkage analyses. Results: Three generations (F3, Backcross 1 and Backcross 2) of the IBMAP and their related animals were genotyped with PorcineSNP60 BeadChip. A total of 8,417 SNPs equidistantly distributed across autosomes were selected after filtering by quality, position and frequency to perform the QTL scan. The joint and separate analyses of the different IBMAP generations allowed confirming QTL regions previously identified in chromosomes 4 and 6 as well as new ones mainly for backfat thickness in chromosomes 4, 5, 11, 14 and 17 and shoulder weight in chromosomes 1, 2, 9 and 13; and many other to the chromosome-wide signification level. In addition, most of the detected QTLs displayed narrow confidence intervals, making easier the selection of positional candidate genes. Conclusions: The use of higher density of markers has allowed to confirm results obtained in previous QTL scans carried out with microsatellites. Moreover several new QTL regions have been now identified in regions probably not covered by markers in previous scans, most of these QTLs displayed narrow confidence intervals. Finally, prominent putative biological and positional candidate genes underlying those QTL effects are listed based on recent porcine genome annotation.This work was funded by MICINN projects AGL2008-04818-C03/GAN and CSD2007-00036. DPM was funded by a FPI Ph.D grant from the Spanish Ministerio de Educacion (BES-2009-025417). YR was funded by a FPU Ph.D grant from the Spanish Ministerio de Educacion (AP2008-01450). We want to thanks to Dr. Martien Groenen (Wageningen, NL) for the SNP annotation on porcine genome assembly, to Anna Mercade for her technical assistance with the SNPs genotyping and to Rita Benitez and Fabian Garcia for technical support.Fernández, A.; Pérez-Montarelo, D.; Barragan, C.; Ramayo-Caldas, Y.; Ibáñez-Escriche, N.; Castelló, A.; Noguera, J.... (2012). Genome-wide linkage analysis of QTL for growth and body composition employing the PorcineSNP60 BeadChip. BMC Genetics. 13:1-11. https://doi.org/10.1186/1471-2156-13-41S11113Van Laere, A.-S., Nguyen, M., Braunschweig, M., Nezer, C., Collette, C., Moreau, L., … Andersson, L. (2003). A regulatory mutation in IGF2 causes a major QTL effect on muscle growth in the pig. Nature, 425(6960), 832-836. doi:10.1038/nature02064John, S., Shephard, N., Liu, G., Zeggini, E., Cao, M., Chen, W., … Kennedy, G. C. (2004). Whole-Genome Scan, in a Complex Disease, Using 11,245 Single-Nucleotide Polymorphisms: Comparison with Microsatellites. The American Journal of Human Genetics, 75(1), 54-64. doi:10.1086/422195Mercadé, A., Estellé, J., Noguera, J. L., Folch, J. M., Varona, L., Silió, L., … Pérez-Enciso, M. (2005). On growth, fatness, and form: A further look at porcine Chromosome 4 in an Iberian × Landrace cross. Mammalian Genome, 16(5), 374-382. doi:10.1007/s00335-004-2447-4Óvilo, C., Pérez-Enciso, M., Barragán, C., Clop, A., Rodríguez, C., Oliver, M. A., … Noguera, J. L. (2000). A QTL for intramuscular fat and backfat thickness is located on porcine Chromosome 6. Mammalian Genome, 11(4), 344-346. doi:10.1007/s003350010065Cristina, Ó., Oliver, A., Noguera, J. L., Clop, A., Barragán, C., Varona, L., … Silió, L. (2002). Test for positional candidate genes for body composition on pig chromosome 6. Genetics Selection Evolution, 34(4). doi:10.1186/1297-9686-34-4-465ÓVILO, C., FERNÁNDEZ, A., NOGUERA, J. L., BARRAGÁN, C., LETÓN, R., RODRÍGUEZ, C., … TORO, M. (2005). Fine mapping of porcine chromosome 6 QTL and LEPR effects on body composition in multiple generations of an Iberian by Landrace intercross. Genetical Research, 85(1), 57-67. doi:10.1017/s0016672305007330Óvilo, C., Fernández, A., Fernández, A. I., Folch, J. M., Varona, L., Benítez, R., … Silió, L. (2010). Hypothalamic expression of porcine leptin receptor (LEPR), neuropeptide Y (NPY), and cocaine- and amphetamine-regulated transcript (CART) genes is influenced by LEPR genotype. Mammalian Genome, 21(11-12), 583-591. doi:10.1007/s00335-010-9307-1Estellé, J., Fernández, A. I., Pérez-Enciso, M., Fernández, A., Rodríguez, C., Sánchez, A., … Folch, J. M. (2009). A non-synonymous mutation in a conserved site of theMTTPgene is strongly associated with protein activity and fatty acid profile in pigs. Animal Genetics, 40(6), 813-820. doi:10.1111/j.1365-2052.2009.01922.xEstellé, J., Pérez-Enciso, M., Mercadé, A., Varona, L., Alves, E., Sánchez, A., & Folch, J. M. (2006). Characterization of the porcine FABP5 gene and its association with the FAT1 QTL in an Iberian by Landrace cross. Animal Genetics, 37(6), 589-591. doi:10.1111/j.1365-2052.2006.01535.xMercadé, A., Pérez-Enciso, M., Varona, L., Alves, E., Noguera, J. L., Sánchez, A., & Folch, J. M. (2006). Adipocyte fatty-acid binding protein is closely associated to the porcine FAT1 locus on chromosome 41. Journal of Animal Science, 84(11), 2907-2913. doi:10.2527/jas.2005-663Evans, D. M., & Cardon, L. R. (2004). Guidelines for Genotyping in Genomewide Linkage Studies: Single-Nucleotide–Polymorphism Maps Versus Microsatellite Maps. The American Journal of Human Genetics, 75(4), 687-692. doi:10.1086/424696Gonzalez-Neira, A., Rosa-Rosa, J., Osorio, A., Gonzalez, E., Southey, M., Sinilnikova, O., … Benitez, J. (2007). Genomewide high-density SNP linkage analysis of non-BRCA1/2 breast cancer families identifies various candidate regions and has greater power than microsatellite studies. BMC Genomics, 8(1), 299. doi:10.1186/1471-2164-8-299Chioza, B. A., Aicardi, J., Aschauer, H., Brouwer, O., Callenbach, P., Covanis, A., … Everett, K. V. (2009). Genome wide high density SNP-based linkage analysis of childhood absence epilepsy identifies a susceptibility locus on chromosome 3p23-p14. Epilepsy Research, 87(2-3), 247-255. doi:10.1016/j.eplepsyres.2009.09.010Ramos, A. M., Crooijmans, R. P. M. A., Affara, N. A., Amaral, A. J., Archibald, A. L., Beever, J. E., … Groenen, M. A. M. (2009). Design of a High Density SNP Genotyping Assay in the Pig Using SNPs Identified and Characterized by Next Generation Sequencing Technology. PLoS ONE, 4(8), e6524. doi:10.1371/journal.pone.0006524Purcell, S., Neale, B., Todd-Brown, K., Thomas, L., Ferreira, M. A. R., 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. doi:10.1086/519795Barrett, J. C., Fry, B., Maller, J., & Daly, M. J. (2004). Haploview: analysis and visualization of LD and haplotype maps. Bioinformatics, 21(2), 263-265. doi:10.1093/bioinformatics/bth457Andersson, L., Haley, C., Ellegren, H., Knott, S., Johansson, M., Andersson, K., … et, al. (1994). Genetic mapping of quantitative trait loci for growth and fatness in pigs. Science, 263(5154), 1771-1774. doi:10.1126/science.8134840Marklund, L., Nyström, P.-E., Stern, S., Andersson-Eklund, L., & Andersson, L. (1999). Confirmed quantitative trait loci for fatness and growth on pig chromosome 4. Heredity, 82(2), 134-141. doi:10.1038/sj.hdy.6884630Fan, B., Onteru, S. K., Du, Z.-Q., Garrick, D. J., Stalder, K. J., & Rothschild, M. F. (2011). Genome-Wide Association Study Identifies Loci for Body Composition and Structural Soundness Traits in Pigs. PLoS ONE, 6(2), e14726. doi:10.1371/journal.pone.0014726Bidanel, J.-P., Milan, D., Iannuccelli, N., Amigues, Y., Boscher, M.-Y., Bourgeois, F., … Chevalet, C. (2001). Detection of quantitative trait loci for growth and fatness in pigs. Genetics Selection Evolution, 33(3). doi:10.1186/1297-9686-33-3-289Geldermann, H., Čepica, S., Stratil, A., Bartenschlager, H., & Preuss, S. (2010). Genome-wide mapping of Quantitative Trait Loci for fatness, fat cell characteristics and fat metabolism in three porcine F2 crosses. Genetics Selection Evolution, 42(1). doi:10.1186/1297-9686-42-31Quintanilla, R., Milan, D., & Bidanel, J.-P. (2002). A further look at quantitative trait loci affecting growth and fatness in a cross between Meishan and Large White pig populations. Genetics Selection Evolution, 34(2), 193. doi:10.1186/1297-9686-34-2-193Sławińska, A., Siwek, M., Knol, E. F., Roelofs-Prins, D. T., van Wijk, H. J., Dibbits, B., & Bednarczyk, M. (2009). Validation of the QTL on SSC4 for meat and carcass quality traits in a commercial crossbred pig population. Journal of Animal Breeding and Genetics, 126(1), 43-51. doi:10.1111/j.1439-0388.2008.00753.xMilan, D., Bidanel, J.-P., Iannuccelli, N., Riquet, J., Amigues, Y., Gruand, J., … Chevalet, C. (2002). Detection of quantitative trait loci for carcass composition traits in pigs. Genetics Selection Evolution, 34(6), 705. doi:10.1186/1297-9686-34-6-705Guo, T., Ren, J., Yang, K., Ma, J., Zhang, Z., & Huang, L. (2009). Quantitative trait loci for fatty acid composition in longissimus dorsi and abdominal fat: results from a White Duroc × Erhualian intercross F2population. Animal Genetics, 40(2), 185-191. doi:10.1111/j.1365-2052.2008.01819.xLiu, G., Kim, J. J., Jonas, E., Wimmers, K., Ponsuksili, S., Murani, E., … Schellander, K. (2008). Combined line-cross and half-sib QTL analysis in Duroc–Pietrain population. Mammalian Genome, 19(6), 429-438. doi:10.1007/s00335-008-9132-yKIM, C. W., HONG, Y. H., YUN, S.-I., LEE, S.-R., KIM, Y. H., KIM, M.-S., … CHANG, K.-T. (2006). Use of Microsatellite Markers to Detect Quantitative Trait Loci in Yorkshire Pigs. Journal of Reproduction and Development, 52(2), 229-237. doi:10.1262/jrd.17046Liu, G., Jennen, D. G. J., Tholen, E., Juengst, H., Kleinwächter, T., Hölker, M., … Wimmers, K. (2007). A genome scan reveals QTL for growth, fatness, leanness and meat quality in a Duroc-Pietrain resource population. Animal Genetics, 38(3), 241-252. doi:10.1111/j.1365-2052.2007.01592.xXu, X. L., Xu, X. W., Pan, P. W., Li, K., Jiang, Z. H., Yu, M., … Liu, B. (2009). Porcine skeletal muscle differentially expressed geneCMYA1: isolation, characterization, mapping, expression and association analysis with carcass traits. Animal Genetics, 40(3), 255-261. doi:10.1111/j.1365-2052.2008.01825.xRamos, A. M., Bastiaansen, J. W. M., Plastow, G. S., & Rothschild, M. F. (2009). Genes located on a SSC17 meat quality QTL region are associated with growth in outbred pig populations. Animal Genetics, 40(5), 774-778. doi:10.1111/j.1365-2052.2009.01907.xRusso, V., Fontanesi, L., Scotti, E., Beretti, F., Davoli, R., Nanni Costa, L., … Buttazzoni, L. (2008). Single nucleotide polymorphisms in several porcine cathepsin genes are associated with growth, carcass, and production traits in Italian Large White pigs1. Journal of Animal Science, 86(12), 3300-3314. doi:10.2527/jas.2008-0920Tsai, F.-J., Yang, C.-F., Chen, C.-C., Chuang, L.-M., Lu, C.-H., Chang, C.-T., … Wu, J.-Y. (2010). A Genome-Wide Association Study Identifies Susceptibility Variants for Type 2 Diabetes in Han Chinese. PLoS Genetics, 6(2), e1000847. doi:10.1371/journal.pgen.1000847Silva, K. M., Bastiaansen, J. W. M., Knol, E. F., Merks, J. W. M., Lopes, P. S., Guimarães, S. E. F., & van Arendonk, J. A. M. (2010). Meta-analysis of results from quantitative trait loci mapping studies on pig chromosome 4. Animal Genetics, 42(3), 280-292. doi:10.1111/j.1365-2052.2010.02145.xFontanesi, L., Scotti, E., Buttazzoni, L., Dall’Olio, S., Davoli, R., & Russo, V. (2009). A single nucleotide polymorphism in the porcine cathepsin K (CTSK) gene is associated with back fat thickness and production traits in Italian Duroc pigs. Molecular Biology Reports, 37(1), 491-495. doi:10.1007/s11033-009-9678-0Ojeda, A., Estellé, J., Folch, J. M., & Pérez-Enciso, M. (2008). Nucleotide variability and linkage disequilibrium patterns at the porcineFABP5gene. Animal Genetics, 39(5), 468-473. doi:10.1111/j.1365-2052.2008.01752.xHan, S.-H., Shin, K.-Y., Lee, S.-S., Ko, M.-S., Jeong, D. K., Oh, H.-S., … Cho, I.-C. (2009). SINE indel polymorphism of AGL gene and association with growth and carcass traits in Landrace × Jeju black pig F2 population. Molecular Biology Reports, 37(1), 467-471. doi:10.1007/s11033-009-9644-xYamauchi, T., Kamon, J., Ito, Y., Tsuchida, A., Yokomizo, T., Kita, S., … Kadowaki, T. (2003). Cloning of adiponectin receptors that mediate antidiabetic metabolic effects. Nature, 423(6941), 762-769. doi:10.1038/nature01705Grundberg, E., Brandstrom, H., Ribom, E., Ljunggren, O., Mallmin, H., & Kindmark, A. (2004). Genetic variation in the human vitamin D receptor is associated with muscle strength, fat mass and body weight in Swedish women. European Journal of Endocrinology, 323-328. doi:10.1530/eje.0.1500323Muñoz, G., Alcázar, E., Fernández, A., Barragán, C., Carrasco, A., de Pedro, E., … Rodríguez, M. C. (2011). Effects of porcine MC4R and LEPR polymorphisms, gender and Duroc sire line on economic traits in Duroc×Iberian crossbred pigs. Meat Science, 88(1), 169-173. doi:10.1016/j.meatsci.2010.12.018Krzęcio, E., Koćwin-Podsiadła, M., Kurył, J., Zybert, A., Sieczkowska, H., & Antosik, K. (2008). The effect of interaction between genotype CAST/RsaI (calpastatin) and MYOG/MspI (myogenin) on carcass and meat quality in pigs free of RYR1T allele. Meat Science, 80(4), 1106-1115. doi:10.1016/j.meatsci.2008.05.002Wyszyńska-Koko, J., Pierzchała, M., Flisikowski, K., Kamyczek, M., Różycki, M., & Kurył, J. (2006). Polymorphisms in coding and regulatory regions of the porcineMYF6 andMYOG genes and expression of theMYF6 gene inm. longissimus dorsi versus productive traits in pigs. Journal of Applied Genetics, 47(2), 131-138. doi:10.1007/bf03194612IKEDA, T., KANAZAWA, T., OTSUKA, S., ICHII, O., HASHIMOTO, Y., & KON, Y. (2009). Expression of Caspase Family and Muscle- and Apoptosis-Specific Genes during Skeletal Myogenesis in Mouse Embryo. Journal of Veterinary Medical Science, 71(9), 1161-1168. doi:10.1292/jvms.71.1161Lin, Z., Lou, Y., & Squires, E. J. (2006). Functional polymorphism in porcine CYP2E1 gene: Its association with skatole levels. The Journal of Steroid Biochemistry and Molecular Biology, 99(4-5), 231-237. doi:10.1016/j.jsbmb.2005.07.001Aubert, J., Begriche, K., Knockaert, L., Robin, M. A., & Fromenty, B. (2011). Increased expression of cytochrome P450 2E1 in nonalcoholic fatty liver disease: Mechanisms and pathophysiological role. Clinics and Research in Hepatology and Gastroenterology, 35(10), 630-637. doi:10.1016/j.clinre.2011.04.015Latreille, M., Laberge, M.-K., Bourret, G., Yamani, L., & Larose, L. (2011). Deletion of Nck1 attenuates hepatic ER stress signaling and improves glucose tolerance and insulin signaling in liver of obese mice. American Journal of Physiology-Endocrinology and Metabolism, 300(3), E423-E434. doi:10.1152/ajpendo.00088.2010Akerfeldt, M. C., & Laybutt, D. R. (2011). Inhibition of Id1 Augments Insulin Secretion and Protects Against High-Fat Diet-Induced Glucose Intolerance. Diabetes, 60(10), 2506-2514. doi:10.2337/db11-008

    Early socialization and environmental enrichment of lactating piglets affects the caecal microbiota and metabolomic response after weaning

    Get PDF
    The aim of this study was to determine the possible impact of early socialization and an enriched neonatal environment to improve adaptation of piglets to weaning. We hypothesized that changes in the microbiota colonization process and in their metabolic response and intestinal functionality could help the animals face weaning stress. A total of 48 sows and their litters were allotted into a control (CTR) or an enriched treatment (ENR), in which piglets from two adjacent pens were combined and enriched with toys. The pattern of caecal microbial colonization, the jejunal gene expression, the serum metabolome and the intestinal physiology of the piglets were assessed before (-2 d) and after weaning (+ 3d). A differential ordination of caecal microbiota was observed after weaning. Serum metabolome suggested a reduced energetic metabolism in ENR animals, as evidenced by shifts in triglycerides and fatty acids, VLDL/LDL and creatine regions. The TLR2 gene showed to be downregulated in the jejunum of ENR pigs after weaning. The integration of gene expression, metabolome and microbiota datasets confirmed that differences between barren and enriched neonatal environments were evident only after weaning. Our results suggest that improvements in adaptation to weaning could be mediated by a better response to the post-weaning stress.info:eu-repo/semantics/publishedVersio

    Gut barrier-microbiota imbalances in early life lead to higher sensitivity to inflammation in a murine model of C-section delivery

    Get PDF
    Most interactions between the host and its microbiota occur at the gut barrier, and primary colonizers are essential in the gut barrier maturation in the early life. The mother-offspring transmission of microorganisms is the most important factor influencing microbial colonization in mammals, and C-section delivery (CSD) is an important disruptive factor of this transfer. Recently, the deregulation of symbiotic host-microbe interactions in early life has been shown to alter the maturation of the immune system, predisposing the host to gut barrier dysfunction and inflammation. The main goal of this study is to decipher the role of the early-life gut microbiota-barrier alterations and its links with later-life risks of intestinal inflammation in a murine model of CSD. The higher sensitivity to chemically induced inflammation in CSD mice is related to excessive exposure to a too diverse microbiota too early in life. This early microbial stimulus has short-term consequences on the host homeostasis. It switches the pup's immune response to an inflammatory context and alters the epithelium structure and the mucus-producing cells, disrupting gut homeostasis. This presence of a too diverse microbiota in the very early life involves a disproportionate short-chain fatty acids ratio and an excessive antigen exposure across the vulnerable gut barrier in the first days of life, before the gut closure. Besides, as shown by microbiota transfer experiments, the microbiota is causal in the high sensitivity of CSD mice to chemical-induced colitis and in most of the phenotypical parameters found altered in early life. Finally, supplementation with lactobacilli, the main bacterial group impacted by CSD in mice, reverts the higher sensitivity to inflammation in ex-germ-free mice colonized by CSD pups' microbiota. Early-life gut microbiota-host crosstalk alterations related to CSD could be the linchpin behind the phenotypic effects that lead to increased susceptibility to an induced inflammation later in life in mice

    Genome-wide association study confirm major QTL for backfat fatty acid composition on SSC14 in Duroc pigs

    Get PDF
    Background: Fatty acid composition contributes importantly to meat quality and is essential to the nutritional value of the meat. Identification of genetic factors underlying levels of fatty acids can be used to breed for pigs with healthier meat. The aim of this study was to conduct genome-wide association studies (GWAS) to identify QTL regions affecting fatty acid composition in backfat from the pig breeds Duroc and Landrace. Results: Using data from the Axiom porcine 660 K array, we performed GWAS on 454 Duroc and 659 Landrace boars for fatty acid phenotypes measured by near-infrared spectroscopy (NIRS) technology (C16:0, C16:1n-7, C18:0, C18:1n-9, C18:2n-6, C18:3n-3, total saturated fatty acids, monounsaturated fatty acids and polyunsaturated fatty acids). Two QTL regions on SSC4 and SSC14 were identified in Duroc for the de novo synthesized fatty acids traits, whereas one QTL on SSC8 was detected in Landrace for C16:1n-7. The QTL region on SSC14 has been reported in previous studies and a putative causative mutation has been suggested in the promoter region of the SCD gene. Whole genome re-sequencing data was used for genotype imputation and to fine map the SSC14 QTL region in Norwegian Duroc. This effort confirms the location of the QTL on this chromosome as well as suggesting other putative candidate genes in the region. The most significant single nucleotide polymorphisms (SNPs) located on SSC14 explain between 55 and 76% of the genetic variance and between 27 and 54% of the phenotypic variance for the de novo synthesized fatty acid traits in Norwegian Duroc. For the QTL region on SSC8 in Landrace, the most significant SNP explained 19% of the genetic variance and 5% of the phenotypic variance for C16:1n-7. Conclusions: This study confirms a major QTL affecting fatty acid composition on SSC14 in Duroc, which can be used in genetic selection to increase the level of fatty acid desaturation. The SSC14 QTL was not segregating in the Landrace population, but another QTL on SSC8 affecting C16:1n-7 was identified and might be used to increase the level of desaturation in meat products from this breed

    Genomic characteristics of cattle copy number variations

    Get PDF
    <p>Abstract</p> <p>Background</p> <p>Copy number variation (CNV) represents another important source of genetic variation complementary to single nucleotide polymorphism (SNP). High-density SNP array data have been routinely used to detect human CNVs, many of which have significant functional effects on gene expression and human diseases. In the dairy industry, a large quantity of SNP genotyping results are becoming available and can be used for CNV discovery to understand and accelerate genetic improvement for complex traits.</p> <p>Results</p> <p>We performed a systematic analysis of CNV using the Bovine HapMap SNP genotyping data, including 539 animals of 21 modern cattle breeds and 6 outgroups. After correcting genomic waves and considering the pedigree information, we identified 682 candidate CNV regions, which represent 139.8 megabases (~4.60%) of the genome. Selected CNVs were further experimentally validated and we found that copy number "gain" CNVs were predominantly clustered in tandem rather than existing as interspersed duplications. Many CNV regions (~56%) overlap with cattle genes (1,263), which are significantly enriched for immunity, lactation, reproduction and rumination. The overlap of this new dataset and other published CNV studies was less than 40%; however, our discovery of large, high frequency (> 5% of animals surveyed) CNV regions showed 90% agreement with other studies. These results highlight the differences and commonalities between technical platforms.</p> <p>Conclusions</p> <p>We present a comprehensive genomic analysis of cattle CNVs derived from SNP data which will be a valuable genomic variation resource. Combined with SNP detection assays, gene-containing CNV regions may help identify genes undergoing artificial selection in domesticated animals.</p
    corecore