Probiotic effect on a gastrointestinal microbiocenosis of calves

Keywords: Key words: probiotic spore-forming strains of Bacillus sp., сalves, gastrointestinal microflora, cut-chewing, digestion

Abstract

The article presents studies about probiotic strains for calves from birth to one month of age. Probiotics are used in dairy farming to increase the immunity of newborn calves and prevent gastrointestinal disorders, increase average daily gain, reduce production stress, normalize the microbiota. The aim of the study was to determine the effect of probiotic strains of calves gastrointestinal microbiota. The research was carried out in the farm «Hlibodar» (village Golovashivka of Sumy district, Sumy region) with different technological groups of cattle management. In an experiment were formed five experimental groups of five animals each and one control group. Experimental groups were fed 5 grams of probiotic strains in the following composition per animal: Bacillus сoagulans, Bacillus mucilaginosus, Bacillus megaterium, Bacillus pumilus, Bacillus amyloliquefaciense.

B. сoagulans did not promote the growth of lactobacilli 7×104 (volumetric datum), but well reduces the number of opportunistic pathogens of the genus Clostridium 101, compared with the control group of calves without probiotic 3×101.

B. mucilaginosus contributed to the reproduction of Lactobacillus sp. up to 8×106, compared to the control group 1×105. The probiotic inhibited the growth of the genus Clostridium below the average 101. Feeding of B. mucilaginosus reduced the amount of Escherichia coli to 2×104.

B. megaterium has a positive effect on the growth of Lactobacillus sp. 5х106, but increases the amount of Clostridium 2×102 and Escherichia coli 6×104, compared with the control group 3×101 and 3×104 respectively.

B. pumilus promotes the growth and reproduction of lactobacilli 7×105, compared to the control - 1×105. The probiotic does not significantly inhibit the growth of Clostridium and Enterobacteriaceae. The probiotic does not destroy Escherichia coli with hemolytic activity.

B. amyloliquefaciense is not an antagonist for Clostridium 2×102and Escherichia coli 6×104. B. amyloliquefaciense destroys hemolytic Escherichia coli, but increases the growth of yeasts to 1×102, staphylococci to 2×104 compared with the control group 7×101 and 6×103, respectively.

Bacteria Salmonella and Pseudomonas were not detected in the calves fecal masses of the experimental and control groups, it indicates the well-being of the farm as regards bacterial gastrointestinal diseases. The number of bifidobacteria in all experimental and control groups were detected up to 105. Studies of Bacillus sp. probiotic strains  prove the partial effectiveness of each of them and give grounds for the creation of a complex probiotic to have an optimum effect for calves.

References

1. Cheng, G., Hao, H., Xie, S., Wang, X., Dai, M., Huang, L. and Yuan, Z. (2014) Antibiotic alternatives: the substitution of antibiotics in animal husbandry? Front Microbiol 5, 69– 83. https://doi.org/10.3389/fmicb.2014.00217
2. Hao, H., Cheng, G., Iqbal, Z., Ai, X., Hussain, H. I., Huang, L., Dai, M., Wang, Y., Liu, Z., & Yuan, Z. (2014). Benefits and risks of antimicrobial use in food-producing animals. Frontiers in microbiology, 5, 288. https://doi.org/10.3389/fmicb.2014.00288.
3. Basso, F.C., Adesogan, A.T., Lara E.C., Rabelo, C. H. S., Berchielli, T. T., Teixeira, I. A. M. A., Siqueira, G. R., Reis, R. A. (2014). Effects of feeding corn silage inoculated with microbial additives on the ruminal fermentation, microbial protein yield, and growth performance of lambs. J Anim Sci. 92(12),5640‐5650. https://doi.org/10.2527/jas.2014-8258
4. Tan, J., McKenzie, C., Potamitis, M., Thorburn, A.N., Mackay, C.R., Macia, L. (2014). The role of short-chain fatty acids in health and disease. Adv Immunol. 121, 91–119. https://doi.org/10.1016/B978-0-12-800100-4.00003-9
5. Shkromada, O., Palii, A., Palii, A., Skliar, O., Dudchenko, Y., & Necherya, T. (2019). Improvement of milk quality for micro-climate formation on cattle farms. Bulletin of Sumy National Agrarian University. The Series: Veterinary Medicine, (4 (47), 43-49. https://doi.org/10.32845/bsnau.vet.2019.4.7
6. Kong, L., Yang, C., Dong, L., Diao, Q., Si, B., Ma, J., & Tu, Y. (2019). Rumen Fermentation Characteristics in Pre- and Post-Weaning Calves upon Feeding with Mulberry Leaf Flavonoids and Candida tropicalis Individually or in Combination as a Supplement. Animals : an open access journal from MDPI, 9(11), 990. doi.org/10.3390/ani9110990
7. Liu, X., Zhao, W., Yu, D., Cheng, J. G., Luo, Y., Wang, Y., Yang, Z. X., Yao, X. P., Wu, S. S., Wang, W. Y., Yang, W., Li, D. Q., & Wu, Y. M. (2019). Effects of compound probiotics on the weight, immunity performance and fecal microbiota of forest musk deer. Scientific reports, 9(1), 19146. doi.org/10.1038/s41598-019-55731-5
8. Rybachuk, Z., Shkromada, O., Predko, A., & Dudchenko, Y. (2020). Influence of probiotics “Immunobacterin-D” on biocenoses and development of the gastrointestinal tract of calves. Scientific Messenger of LNU of Veterinary Medicine and Biotechnologies. Series: Veterinary Sciences, 22(98), 22-27. https://doi.org/10.32718/nvlvet9804
9. Ruiz, L., Ruas-Madiedo, P., Gueimonde, M., de Los Reyes-Gavilán, C. G., Margolles, A., & Sánchez, B. (2011). How do bifidobacteria counteract environmental challenges? Mechanisms involved and physiological consequences. Genes & nutrition, 6(3), 307–318. https://doi.org/10.1007/s12263-010-0207-5
10. Govender, M., Choonara, Y. E., Kumar, P., du Toit, L. C., van Vuuren, S., & Pillay, V. (2014). A review of the advancements in probiotic delivery: Conventional vs. non-conventional formulations for intestinal flora supplementation. AAPS PharmSciTech, 15(1), 29–43. https://doi.org/10.1208/s12249-013-0027-1
11. Mingmongkolchai, S., & Panbangred, W. (2018). Bacillus probiotics: an alternative to antibiotics for livestock production. Journal of applied microbiology, 124(6), 1334–1346. https://doi.org/10.1111/jam.13690
12. Kapse, N. G., Engineer, A. S., Gowdaman, V., Wagh, S., & Dhakephalkar, P. K. (2019). Functional annotation of the genome unravels probiotic potential of Bacillus сoagulans HS243. Genomics, 111(4), 921–929. https://doi.org/10.1016/j.ygeno.2018.05.022
13. Fijan S. (2014). Microorganisms with claimed probiotic properties: an overview of recent literature. International journal of environmental research and public health, 11(5), 4745–4767. https://doi.org/10.3390/ijerph110504745
14. Wu, H.J., Sun, L.B., Li, C.B., Li, Z.Z., Zhang, Z., Wen, X.B., Hu, Z., Zhang, Y.L. (2014) Enhancement of the immune response and protection against Vibrio parahaemolyticus by indigenous probiotic Bacillus strains in mud crab (Scylla paramamosain). Fish Shellfish Immunol 41, 156– 162. https://doi.org/10.1016/j.fsi.2014.08.027
15. EFSA (2014) Guidance on the assessment of the toxigenic potential of Bacillus species used in animal nutrition. EFSA J 12, 3665. https://doi.org/10.2903/j.efsa.2014.3665
16. Zhou, Y., Zeng, Z., Xu, Y., Ying, J., Wang, B., Majeed, M., Majeed, S., Pande, A., & Li, W. (2020). Application of Bacillus сoagulans in Animal Husbandry and Its Underlying Mechanisms. Animals : an open access journal from MDPI, 10(3), 454. https://doi.org/10.3390/ani10030454
17. Izuddin, W. I., Humam, A. M., Loh, T. C., Foo, H. L., & Samsudin, A. A. (2020). Dietary Postbiotic Lactobacillus plantarum Improves Serum and Ruminal Antioxidant Activity and Upregulates Hepatic Antioxidant Enzymes and Ruminal Barrier Function in Post-Weaning Lambs. Antioxidants (Basel, Switzerland), 9(3), 250. doi.org/10.3390/antiox9030250
18. Silva, L.D.D., Pereira, O.G., Silva, T.C.D., Valadares Filho, S.C., Ribeiro, K.G. (2016) Effects of silage crop and dietary crude protein levels on digestibility ruminal fermentation, nitrogen use efficiency, and performance of finishing beef cattle. Anim Feed Sci Technol. 220, 22–33. https://doi: 10.1016/j.anifeedsci.2016.07.008.
19. Soares, M.S., Oliveira, P.S., Debom, G.N., DaSilveira, M.B., Polachini, C.R., Baldissarelli, J., et al. (2017). Chronic administration of methionine and/or methionine sulfoxide alters oxidative stress parameters and ALA-D activity in liver and kidney of young rats. Amino Acids. 49(1), 129–138. https://doi.org/10.1007/s00726-016-2340-y.
20. Fan, P., Li, L., Rezaei, A., Eslamfam, S., Che, D., Ma, X. (2015). Metabolites of dietary protein and peptides by intestinal microbes and their impacts on the gut. Curr Protein Pept Sci. 16, 646–654. https://doi.org/10.2174/1389203716666150630133657.
21. Aikman, P.C., Henning, P.H., Humphries, D.J., Horn, C.H. (2010). Rumen pH and fermentation characteristics in dairy cows supplemented with Megasphaera elsdenii NCIMB 41125 in early lactation. J. Dairy Sci. 94, 2840–2849. https://doi.org/10.3168/jds.2010-3783
22. Stover, P.J., Durga, J., Field, M.S. (2017). Folate nutrition and blood–brain barrier dysfunction. Curr Opin Biotechnol. 44, 146–152. https://doi.org/10.1016/j.copbio.2017.01.006.
23. Diao, Q., Zhang, R., Tu, Y. (2017). Current research progresses on calf rearing and nutrition in China. J. Integr. 16, 2805–2814. https://doi.org/10.1016/S2095-3119(17)61767-2
24. Sun, P., Wang, J.Q., Zhang, H.T. (2010). Effects of Bacillus subtilis natto on performance and immune function of preweaning calves. J Dairy Sci. 93(12), 5851‐5855. https://doi.org/10.3168/jds.2010-3263
25. Shinde, T., Vemuri, R., Shastri, M.D., Perera, A.P., Tristram, S., Stanley, R., Eri, R. (2019). Probiotic Bacillus сoagulans MTCC 5856 spores exhibit excellent in-vitro functional efficacy in simulated gastric survival, mucosal adhesion and immunomodulation. J. Funct. Foods, 52, 100–108. https://doi.org/10.1016/j.jff.2018.10.031.
How to Cite
Shkromada, O., Dudchenko, J., & Udovenko, Y. (1). Probiotic effect on a gastrointestinal microbiocenosis of calves. Bulletin of Sumy National Agrarian University. The Series: Veterinary Medicine, (1 (48), 3-8. https://doi.org/10.32845/bsnau.vet.2020.1.1