53 research outputs found

    The effects of low levels of aflatoxin B1 on health, growth performance and reproductivity in male rabbits

    Get PDF
    [EN] This study investigated the chronic effects of relatively low exposure to aflatoxin B1 (AFB1) on the growth performance, immune situation and reproduction in male rabbits. Bucks (n=32, 4.82±0.22 kg) were individually assigned to 4 treatments (8 replicates each) using a randomised complete block design. Four diets containing 0, 0.02, 0.05, and 0.1 mg AFB1/kg, were provided to bucks for 8 wk. Growth performance and semen quality were measured. Blood, organ and tissue samples were collected to measure haematological indices, liver function, organ weights and immune parameters. Compared to control, AFB1-contaminated diets reduced body weight and average daily gain (P<0.05), altered certain haematological indices and liver function with decreased monocytes percentage and mean corpuscular haemoglobin concentration, and increased plateletcrit and albumin (P<0.05), slightly impaired reproductive parameters with enhanced ratio of morphologically abnormal sperm cells at early stage and reduced post-stage acrosome integrity, testis weight and serum testosterone concentration (P<0.05), decreased immune function with reduced relative liver weight (%) and tumour necrosis factor-α levels in serum and liver tissue, and increased serum 8-hydroxy-2’-deoxyguanosine levels (P<0.05). Furthermore, bucks fed diets with relatively high AFB1 (0.05 and 0.1 mg AFB1/kg) had reduced red blood cell and haematocrit (P<0.05) in contrast with the low AFB1 group (0.02 mg AFB1/kg). In conclusion, diets containing 0.05 and 0.1 mg AFB1/kg had negative effects on bucks’ growth performance, haematology, reproductivity and immune function, whereas diet containing 0.02 mg AFB1/kg had only minor effects on the parameters measured.The study was funded by the Fundamental Research Funds for the Central Universities (XDJK2015C081).Sun, Y.; Dong, G.; E, G.; Liao, M.; Tao, L.; Lv, J. (2018). The effects of low levels of aflatoxin B1 on health, growth performance and reproductivity in male rabbits. World Rabbit Science. 26(2):123-133. https://doi.org/10.4995/wrs.2018.7433SWORD123133262Abdelaziz S.A., Hamada M.M. 2007. Phytic acid exposure alters AflatoxinB1-induced reproductive and oxidative toxicity in Albino Rats (Rattus norvegicus). eCAM, 6: 331-3471. https://doi.org/10.1093/ecam/nem137Abdel-Wahhab M.A., Nada S.A., Khalil F.A.2002. Physiological and toxicological responses in rats fed aflatoxin-contaminated diet with or without sorbent materials. Animal Feed Sci. Tech., 97: 209-219. https://doi.org/10.1016/S0377-8401(01)00342-XAbnet C.C. 2007.Carcinogenic food contaminants. Cancer Invest., 25: 189-196. https://doi.org/10.1080/07357900701208733Adedara I.A., Nanjappa M.K., Farombi E.O., Akingbemi B.T. 2014. Aflatoxin B1 disrupts the androgen biosynthetic pathway in rat Leydig cells. Food Chem. Toxicol., 65: 252-259. https://doi.org/10.1016/j.fct.2013.12.027Alm K., Dahlbom M., Saynajarvi M., Anderson M.A., Salkinoja-Salonen M.S., Anderson M.C. 2002. Impaired semen quality of AI bulls fed with moldy hay: a case report. Theriogenology, 58: 1497-1502. https://doi.org/10.1016/S0093-691X(02)01079-8Asare G.A., Bronz M., Naidoo V., Kew M.C. 2007. Interactions between aflatoxin B1 and dietary iron overload in hepatic mutagenesis. Toxicol., 234: 157-166. https://doi.org/10.1016/j.tox.2007.02.009Ataman M.B., Dönmez H.H., Dönmez N., Sur E., Bucak M.N., Çoyan, K. 2014. Protective effect of esterified glucomannan on aflatoxin-induced changes in testicular function, sperm quality, and seminal plasma biochemistry in rams. Theriogenology, 81: 373-380. https://doi.org/10.1016/j.theriogenology.2013.10.007Clarke R.N., Doerr J.A., Ottinger M.A. 1986. Relative importance of dietary aflatoxin and feed restriction on reproductive changes associated with aflatoxicosis in the maturing white leghorn male. Poul. Sci., 65: 2239-2245. https://doi.org/10.3382/ps.0652239Cotty P.J., Jaime-Garcia R. 2007. Influences of climate on aflatoxin producing fungi and aflatoxin contamination. Int. J. Food Microbiol., 199: 109-115. https://doi.org/10.1016/j.ijfoodmicro.2007.07.060David S.S., O'Shea V.L., Kundu S. 2007. Base-excision repair of oxidative DNA damage. Nature, 447: 941-950. https://doi.org/10.1038/nature05978Dönmez N., Keskin E. 2008. The effects of aflatoxin and glucomannan on some antioxidants and biochemical parameters in rabbits. Acta Vet. Beograd., 58: 307-313. https://doi.org/10.2298/AVB0804307DEgbunike G.N. 1982. Steroidogenic and spermatogenic potentials of the male rat after acute treatment with Aflatoxin B1. Andrologia, 14: 440-446. https://doi.org/10.1111/j.1439-0272.1982.tb02291.xEisa A.M.A., Metwally A.Y. 2011. Effect of glucomannan on haematological, coagulation and biochemical parameters in male rabbits fed aflatoxin-contaminated ration. World Mycotoxin J., 4: 183-188. https://doi.org/10.3920/WMJ2010.1273Ellis O., Smith J.P., Simpson B.K. 1991. Aflatoxins in food: occurrence, biosynthesis, effects on organisms, detection, and methods of control. Crit. Rev. Food. Sci., 30: 403-439. https://doi.org/10.1080/10408399109527551European Commission. 2003. Commission directive 2003/100/EC of 31 October 2003 amending Annex I to Directive 2002/32/EC of the European Parliament and of the Council on undesirable substances in animal feed. No. 2003/100/EC, 31 October 2003. Off. J. Eur. Comm., 1 November 2003, L 285, 33-37.Ewuola E.O. 2008. Organ traits and histopathology of rabbits fed varied levels of dietary fumonisin B1. J. Anim. Physiol. An. N., 93: 726-731. https://doi.org/10.1111/j.1439-0396.2008.00862.xEwuola E.O., Egbunike G.N. 2010. Effects of dietary fumonisin B1 on the onset of puberty, semen quality, fertility rates and testicular morphology in male rabbits. Reproduction, 139: 439-445. https://doi.org/10.1530/REP-09-0077Ewuola E.O., Jimoh O.A., Bello A.D., Bolarinwa A.O. 2014. Testicular biochemicals, sperm reserves and daily sperm production of West African dwarf bucks fed varied levels of dietary aflatoxin. Anim. Reprod. Sci., 148: 182-187. https://doi.org/10.1016/j.anireprosci.2014.05.010Fan Y., Li X., Zhao L., Jia Y., Ji C., Ma Q., Chen Y., Wang L. 2012. Investigation on contamination situation of aflatoxin in detected feeds and feedstuffs in Beijing area. Scientia Agricultura Sinica, 45: 5102-5109.Fan Y., Zhao L., Ma Q., Li X., Shi H., Zhou T., Zhang J., Ji C. 2013. Effects of Bacillus subtilis ANSB060 on growth performance, meat quality and aflatoxin residues in broilers fed moldy peanut meal naturally contaminated with aflatoxins. Food Chem. Toxicol., 59: 748-753. https://doi.org/10.1016/j.fct.2013.07.010Faridha A., Faisal K., Akbarsha M.A. 2006. Duration-dependent histopathological and histometric changes in the testis of aflatoxin-treated mice. J. Endocrin. Reprod., 10: 117-133.Gholami-Ahangaran M., Zia-Jahromi N. 2013. Nanosilver effects on growth parameters in experimental aflatoxicosis in broiler chickens. Toxicol. Ind. Health, 29: 121-125. https://doi.org/10.1177/0748233711425078Gong Y., Hounsa A., Egal S., Turner P.C., Sutcliffe A.E., Hall A.J., Cardwell K., Wild C.P. 2004. Post-weaning exposure to aflatoxin results in impaired child growth: a longitudinal study in Benin. Environ. Health Perspect., 112: 1334-1338. https://doi.org/10.1289/ehp.6954Guerre P., Eeckhoutte C., Larrieu G., Burgat V., Galtier P. 1996. Dose-related effect of aflatoxin B1 on liver drug metabolizing enzymes in rabbit. Toxicology, 108: 39-48.https://doi.org/10.1016/S0300-483X(95)03269-LGuerre P., Larrieu G., Burgat V., Galtier P. 1999. Cytochrome P450 decreases are correlated to increased microsomal oxidative damage in rabbit liver and primary cultures of rabbit hepatocytes exposed to AFB1. Toxicol. Lett., 104: 117-125.https://doi.org/10.1016/S0378-4274(98)00352-XGuindon-Kezis K.A., Mulder J.E., Massey T.E. 2014. In vivo treatment with aflatoxin B1 increases DNA oxidation, base excision repair activity and 8-oxoguanine DNA glycosylase 1 levels in mouse lung. Toxicology, 321: 21-26. https://doi.org/10.1016/j.tox.2014.03.004Hancock K.D., Coleman E.S., Tao Y.X., Morrison E.E., Braden T.D., Kemppainen B.W., Akingbemi B.T. 2009. Genistein decreases androgen biosynthesis in rat Leydig cells by interference with luteinizing hormone-dependent signaling. Toxicol. Lett., 184: 169-175. https://doi.org/10.1016/j.toxlet.2008.11.005Issac A.A., Manjunatha K.N., Ebenezer O.F., Benson T.A. 2014. Aflatoxin B1 disrupts the androgen biosynthetic pathway in rat Leydig cells. Food Chem. Toxicol., 65: 252-259. https://doi.org/10.1016/j.fct.2013.12.027Kaneko J.J., Harvey J.W., Bruss M. 1997. Serum protein and the dysproteinemias. In clinical biochemistry of domestic animals, 5th ed.; Academic press: San Diego, CA, USA; pp. 117-137. https://doi.org/10.1016/B978-012396305-5/50006-3KĂ€renlampi S.O. 1987. Mechanism of cytotoxicity of aflatoxin B1: role of cytochrome P1-450. Biochem. Bioph. Res. Co., 145: 845-860. https://doi.org/10.1016/0006-291X(87)91043-6Kecici T., Demet Ö., Oguz H. 1995. Single and combined effects of dietary aflatoxin and adsorbent (Mycofix plus) on some hematological and serum biochemical parameters of broiler chickens. J. Vet. Sci., 11: 95-101.KovĂĄcs M., Tornyos G., Matics Zs., Kametler L., Rajli V., BodnĂĄr Zs., KulcsĂĄr M., Huszenicza Gy., Keresztes Zs., Cseh S. 2011. Subsequent effect of subacute T-2 toxicosis on spermatozoa, seminal plasma and testosterone production in rabbits. Animal, 5: 1563-1569. https://doi.org/10.1017/S1751731111000644Manafi M., Murthy H.N.N., Mohan K., Narayana H.D.S. 2012. Evaluation of different mycotoxin binders on broiler breeders induced with aflatoxin B1: effects on fertility, hatchability, embryonic mortality, residues in egg and semen quality. Global Vet., 8: 643-648.Marin D.E., TaranuI., Bunaciu R.P., Pascale F., Tudor D.S., Avram N., Sarca M., Cureu I., Criste R.D., Suta V., Oswald I.P. 2002. Changes in performance, blood parameters, humoral and cellular immune responses in weanling piglets exposed to low doses of aflatoxin. J. Anim. Sci., 80: 1250-1257. https://doi.org/10.2527/2002.8051250xMohanamba T., Rao M.R., Habibi S.M.M. 2007. Aflatoxin contamination in animal feeds. Ind. Vet. J., 84:416.3Morton D. 1988. The use of rabbits in male reproductive toxicology. Environ. Health Persp., 77: 5-9. https://doi.org/10.1289/ehp.88775Oguz H., Kurtoglu V. 2000. Effect of clinoptilolite on performance of broiler chickens during experimental aflatoxicosis. Brit. Poult. Sci., 41: 512-517. https://doi.org/10.1080/713654953Peters L.P., Teel R.W. 2003. Effect of high sucrose diet on liver enzyme content and activity and aflatoxin B1-induced mutagenesis. InVivo.,17: 205-210.Prabu P.C., Dwivedi P., Sharma A.K. 2013. Toxico pathological studies on the effects of aflatoxin B1, ochratoxin A and their interaction in New Zealand White rabbits. Exp. Toxicol. Pathol., 65: 277-286. https://doi.org/10.1016/j.etp.2011.09.003Richard J.L., Thurston J.R. 1975. Effect of aflatoxin on phagocytosis of Aspergillus fumigatus spores by rabbit alveolar macrophages. Appl. Microbiol., 30: 44-47.Rustemeyer S.M., Lamberson W.R., Ledoux D.R., Rottinghaus G.E., Shaw D.P., Cockrum R.R., Kessler K.L., Austin K.J., Cammack K.M. 2014. Effects of dietary aflatoxin on the health and performance of growing barrows. J. Anim. Sci., 88: 3624-3630.https://doi.org/10.2527/jas.2009-2663Salem M.H., Kamel K.I., Yousef M.I., Hassan G.A., ELNouty F.D. 2001. Protective role of ascorbic acid to enhance semen quality of rabbits treated with sublethal doses of aflatoxin B1. Toxicology, 162: 209-218.https://doi.org/10.1016/S0300-483X(01)00366-3Sherrill J.D., Sparks M., Dennis J., Mansour M., Kemppainen B.W., Bartol F.F., Morrison E.E., Akingbemi B.T. 2010. Developmental exposures of male rats to soy isoflavones impact Leydig cell differentiation. Biol. Reprod., 83: 488-501. https://doi.org/10.1095/biolreprod.109.082685Soliman K.M., El-Faramawy A.A., Zakaria S.M., Mekkawy S.H. 2001. Monitoring the preventive effect of hydrogen peroxide and Îł-radiation of aflatoxicosis in growing rabbits and the effect of cooking on aflatoxin residues. J. Agric. Food Chem., 49: 3291-3295. https://doi.org/10.1021/jf0010735Tung H.T., Donaldson W.E., Hamilton P.B. 1972. Altered lipid transport during aflatoxicosis. Toxicol. Appl. Pharmacol., 22: 97-104. https://doi.org/10.1016/0041-008X(72)90229-3Verma R.J., Mathuria N. 2010. Curcumin ameliorates aflatoxininduced changes in caput and cauda epididymis of mice. Int. J. Fertil. Steril., 4: 17-22.Waal Malefyt R., Abrams J., Bennett B., Figdor C.G., de Vries J.E. 1991. Interleukin 10 (IL-10) inhibits cytokine synthesis: An auto regulatory role of IL-10 produced by monocytes. J. Exp. Med. 174: 1209-1220. https://doi.org/10.1084/jem.174.5.1209Weaver A.C., See M.T., Hansen J.A., Kim Y.B, De Souza A.L.P., Teena F.M., Kim S.W. 2013. The use of feed additives to reduce the effects of aflatoxin and deoxynivalenol on pig growth, organ health and immune status during chronic exposure. Toxins, 5: 1261-1281. https://doi.org/10.3390/toxins5071261Williams J.H., Phillips T.D., Jolly P.E., Stiles J.K., Jolly C.M. Aggarwal D. 2004. Human aflatoxicosis in developing countries; a review of toxicology, exposure, potential health consequences, and interventions. Am. J. Clin. Nutr., 80: 1106-1122. https://doi.org/10.1093/ajcn/80.5.1106Yang J., Bai F., Zhang K., Bai S., Peng X., Ding X., Li Y., Zhang J., Zhao L. 2012. Effects of feeding corn naturally contaminated with aflatoxin B1 and B2 on hepatic functions of broilers. Poul. Sci., 91: 2792-2801. https://doi.org/10.3382/ps.2012-02544Yegani M., Smith T.K., Leeson S., Boermans H.J. 2006. Effects of feeding grains naturally contaminated with Fusarium mycotoxins on performance and metabolism of broiler breeders. Poult. Sci., 85: 1541-1549. https://doi.org/10.1093/ps/85.9.1541Yu F.L. 1982. Studies on the mechanism of aflatoxin B1 inhibition of the rat liver nucleolar RNA synthesis. J. Biol. Chem., 256: 3292-3297.Yunus A.W., Razzazi-Fazeli E., Bohm J. 2011. Aflatoxin B1 in affecting broiler's performance, immunity, and gastrointestinal tract: a review of history and contemporary issues. Toxins, 3: 566-590. https://doi.org/10.3390/toxins306056

    Synthesis and structure of m-terphenyl thio-, seleno-, and telluroethers

    Get PDF
    Several routes for the synthesis of m-terphenyl thio-, seleno-, and telluroethers were investigated. m-Terphenyl iodides react with diphenyl diselenides or ditellurides (CsOH·H₂O, DMSO, 110 °C) to give the desired compounds in 19−84% yield which significantly extends the previously reported such reactions because o-benzyne cannot be an intermediate as previously suggested. However, the most general synthetic route was that involving reaction of 2,6-diaryl Grignard reagents with sulfur, selenium, or tellurium electrophiles. The m-terphenyl thio-, seleno-, and telluroethers were characterized spectroscopically and, in one case, by single-crystal X-ray analysis. Certain of these compounds showed atropisomerism and barriers for interconversion of isomers were determined by variable-temperature NMR spectroscopy. The barriers for interconverting the syn and anti atropisomers increase on going from the analogous S to Se to Te compounds. Calculations on this isomerization revealed that the barriers are due to rotation about the aryl−aryl bond and that the barriers for rotation about the aryl−chalcogen bond are much lower

    Synthesis and rotation barriers in 2, 6-Di-(o-anisyl) anisole

    Get PDF
    Variable temperature ÂčH NMR spectroscopic studies of 2, 6-di(o-anisyl) anisole show syn and anti atropisomers at low temperature. The barrier for interconverting these isomers by rotation about the aryl-aryl bond, found by fitting the experimental data, is 41.2 kJ/mol

    Finishing the euchromatic sequence of the human genome

    Get PDF
    The sequence of the human genome encodes the genetic instructions for human physiology, as well as rich information about human evolution. In 2001, the International Human Genome Sequencing Consortium reported a draft sequence of the euchromatic portion of the human genome. Since then, the international collaboration has worked to convert this draft into a genome sequence with high accuracy and nearly complete coverage. Here, we report the result of this finishing process. The current genome sequence (Build 35) contains 2.85 billion nucleotides interrupted by only 341 gaps. It covers ∌99% of the euchromatic genome and is accurate to an error rate of ∌1 event per 100,000 bases. Many of the remaining euchromatic gaps are associated with segmental duplications and will require focused work with new methods. The near-complete sequence, the first for a vertebrate, greatly improves the precision of biological analyses of the human genome including studies of gene number, birth and death. Notably, the human enome seems to encode only 20,000-25,000 protein-coding genes. The genome sequence reported here should serve as a firm foundation for biomedical research in the decades ahead

    Assessment of genetic variation among four populations of Small East African goats using microsatellite markers

    Get PDF
    The majority of goats in Tanzania belong to the Small East African (SEA) breed, which exhibits large phenotypic variation. This study aimed to determine the genetic structure of, and relationships among four populations (Sukuma, Gogo, Sonjo, and Pare) of the SEA breed that have not been studied adequately. A total of 120 individuals (24 from each population) were analysed at eight microsatellite loci. In addition, 24 goats of the South African Boer breed were used as reference. Observed heterozygosity (Ho) ranged from 0.583 ± 0.04 for Sukuma to 0.659 ± 0.030 for Gogo, while expected heterozygosity (He) ranged from 0.632 ± 0.16 for Sukuma to 0.716 ± 0.16 for Boer. Five loci deviated from Hardy–Weinberg equilibrium (HWE) across populations. The mean number of alleles ranged from 4.75 ± 1.58 for Pare to 6.88 ± 3.00 for Sukuma. The mean inbreeding coefficient (FIS) ranged from 0.003 in Sonjo to 0.148 in Sukuma. The differentiation coefficient (FST) was highest (0.085) between Boer and Sukuma and lowest (0.008) between Gogo and Sonjo. The largest genetic distance (0.456) was found between Sukuma and Boer, while the smallest (0.031) was between Gogo and Sonjo populations. Pare, Gogo, and Sonjo populations, formed one cluster, while Sukuma and Boer populations formed two separate clusters. From the findings, it can be concluded that the SEA goats in this study showed high in population genetic variation, which implies that there is good scope for their further improvement through selection within populations. The Sukuma population, which has fairly high inbreeding, is moderately differentiated from Pare, Sonjo, and Gogo goat populations, which showed a high level of admixture. Conservation and improvement strategies of the goats should be designed with first priority being on Sukuma goats.Keywords: Conservation, genetic diversity, genetic markers, local genetic resource

    The Glycolytic Metabolite, Fructose-1,6-bisphosphate, Blocks Epileptiform Bursts by Attenuating Voltage-Activated Calcium Currents in Hippocampal Slices

    No full text
    Manipulation of metabolic pathways (e.g., ketogenic diet (KD), glycolytic inhibition) alters neural excitability and represents a novel strategy for treatment of drug-refractory seizures. We have previously shown that inhibition of glycolysis suppresses epileptiform activity in hippocampal slices. In the present study, we aimed to examine the role of a “branching” metabolic pathway stemming off glycolysis (i.e., the pentose-phosphate pathway, PPP) in regulating seizure activity, by using a potent PPP stimulator and glycolytic intermediate, fructose-1,6-bisphosphate (F1,6BP). Employing electrophysiological approaches, we investigated the action of F1,6BP on epileptiform population bursts, intrinsic neuronal firing, glutamatergic and GABAergic synaptic transmission and voltage-activated calcium currents (ICa) in the CA3 area of hippocampal slices. Bath application of F1,6BP (2.5–5 mM) blocked epileptiform population bursts induced in Mg2+-free medium containing 4-aminopyridine, in ~2/3 of the slices. The blockade occurred relatively rapidly (~4 min), suggesting an extracellular mechanism. However, F1,6BP did not block spontaneous intrinsic firing of the CA3 neurons (when synaptic transmission was eliminated with DNQX, AP-5 and SR95531), nor did it significantly reduce AMPA or NMDA receptor-mediated excitatory postsynaptic currents (EPSCAMPA and EPSCNMDA). In contrast, F1,6BP caused moderate reduction (~50%) in GABAA receptor-mediated current, suggesting it affects excitatory and inhibitory synapses differently. Finally and unexpectedly, F1,6BP consistently attenuated ICa by ~40% without altering channel activation or inactivation kinetics, which may explain its anticonvulsant action, at least in this in vitro seizure model. Consistent with these results, epileptiform population bursts in CA3 were readily blocked by the nonspecific Ca2+ channel blocker, CdCl2 (20 ÎŒM), suggesting that these bursts are calcium dependent. Altogether, these data demonstrate that the glycolytic metabolite, F1,6BP, blocks epileptiform activity via a previously unrecognized extracellular effect on ICa, which provides new insight into the metabolic control of neural excitability

    Comparative Transcriptomic Profiling in Ovarian Tissues of Lohmann Hens and Chengkou Mountain Chicken

    No full text
    Background: As a crucial economic characteristic and a major indicator of reproductive performance in layers, egg production is controlled by a series of complex regulatory heredity basis. In particular, the interacting regulatory function between noncoding RNAs (ncRNAs) and coding RNA plays important roles in regulating laying performance. Methods: In this study, the RNA sequencing (RNA-seq) of ovarian tissues from Lohmann hens (n = 3) and Chengkou Mountain chicken (n = 3) under the laying peak period was performed to identify RNA transcriptional differences among different laying-performance populations. Results: Results showed that the expression level of 303 mRNAs, 68 long ncRNAs (lncRNAs), 533 circular RNAs (circRNAs), and 79 microRNAs (miRNAs) was significantly different among the groups. Functional enrichment analysis of these differentially expressed (DE) mRNAs revealed that the laying process was implicated in numerous significantly enriched pathways (p < 0.05), such as the neuroactive ligand-receptor interaction, steroid hormone biosynthesis, and calcium-signaling pathway. Furthermore, the lncRNA/circRNA–miRNA–mRNA regulatory networks related to the regulation of laying performance were constructed. Some randomly selective DE RNAs were verified by Real Time Quantitative (RT-qRCR), indicating that the bioinformatics analysis results of RNA-seq data were credible. Conclusions: This study could increase our understanding of the heredity basis of transcriptome in the laying performance of chicken

    Hereditary Basis of Coat Color and Excellent Feed Conversion Rate of Red Angus Cattle by Next-Generation Sequencing Data

    No full text
    Angus cattle have made remarkable contributions to the livestock industry worldwide as a commercial meat-type breed. Some evidence supported that Angus cattle with different coat colors have different feed-to-meat ratios, and the genetic basis of their coat color is inconclusive. Here, genome-wide association study was performed to investigate the genetic divergence of black and red Angus cattle with 63 public genome sequencing data. General linear model analysis was used to identify genomic regions with potential candidate variant/genes that contribute to coat color and feed conversion rate. Results showed that six single nucleotide polymorphisms (SNPs) and two insertion&ndash;deletions, which were annotated in five genes (ZCCHC14, ANKRD11, FANCA, MC1R, and LOC532875 [AFG3-like protein 1]), considerably diverged between black and red Angus cattle. The strongest associated loci, namely, missense mutation CHIR18_14705671 (c.296T &gt; C) and frameshift mutation CHIR18_12999497 (c.310G&gt;-), were located in MC1R. Three consecutive strongly associated SNPs were also identified and located in FANCA, which is widely involved in the Fanconi anemia pathway. Several SNPs of highly associated SNPs was notably enriched in ZCCHC14 and ANKRD11, which are related to myofiber growth and muscle development. This study provides a basis for the use of potential genetic markers to be used in future breeding programs to improve cattle selection in terms of coat color and meat phenotype. This study is also helpful to understand the hereditary basis of different coat colors and meat phenotypes. However, the putative candidate genes or markers identified in this study require further investigation to confirm their phenotypic causality and potential effective genetic relationships
    • 

    corecore