Journal Home > Volume 12 , Issue 5

In this study, yak bone collagen hydrolysate (YBCH) was produced by mixed proteases and provided to standard-diet mice at a different dose (low dose (LD), medium dose (MD), and high dose (HD)) to investigate its effects on the composition of gut microbiota and short-chain fatty acids (SCFA) production. It was found that YBCH was mainly composed of small molecular peptides whose molecular weight below 2000 Da. Notably, supplementation with different doses of YBCH could significantly downregulate the ratio of Firmicutes to Bacteroidetes in the fecal microbiota. At the family level, the Lachnospiraceae abundance was significantly reduced in the YBCH gavage groups (mean reduction ratio 41.7%, 35.2%, and 36.4% for LD, MD, and HD group, respectively). The predicted functions of gut microbes in the MD group were significantly increased at "lipid metabolism" and "glycan biosynthesis and metabolism". Moreover, the SCFA production in the YBCH groups was elevated. Especially, the concentration of acetic acid, propionic acid, and butyric acid in the MD group was separately increased 79.7%, 89.2%, and 78.8% than that in the NC group. These results indicated that YBCH might be applied in the development of functional food for intestinal microecological regulation.


menu
Abstract
Full text
Outline
About this article

Supplementation with yak (Bos grunniens) bone collagen hydrolysate altered the structure of gut microbiota and elevated short-chain fatty acid production in mice

Show Author's information Zitao Guoa,bDalong YicBo HudLingyu Zhua,bJi Zhanga,bYuliang Yanga,bChunyu Liua,bYi Shia,bZhenghua Gua,bYu Xina,bHuaigao LiucLiang Zhanga,b( )
National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi 214122, China
Jiangsu Provincial Research Center for Bioactive Product Processing Technology, Jiangnan University, Wuxi 214122, China
Anhui Guotai Biotechnology Co., Ltd, Xuancheng 242100, China
National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China

Peer review under responsibility of KeAi Communications Co., Ltd.

Abstract

In this study, yak bone collagen hydrolysate (YBCH) was produced by mixed proteases and provided to standard-diet mice at a different dose (low dose (LD), medium dose (MD), and high dose (HD)) to investigate its effects on the composition of gut microbiota and short-chain fatty acids (SCFA) production. It was found that YBCH was mainly composed of small molecular peptides whose molecular weight below 2000 Da. Notably, supplementation with different doses of YBCH could significantly downregulate the ratio of Firmicutes to Bacteroidetes in the fecal microbiota. At the family level, the Lachnospiraceae abundance was significantly reduced in the YBCH gavage groups (mean reduction ratio 41.7%, 35.2%, and 36.4% for LD, MD, and HD group, respectively). The predicted functions of gut microbes in the MD group were significantly increased at "lipid metabolism" and "glycan biosynthesis and metabolism". Moreover, the SCFA production in the YBCH groups was elevated. Especially, the concentration of acetic acid, propionic acid, and butyric acid in the MD group was separately increased 79.7%, 89.2%, and 78.8% than that in the NC group. These results indicated that YBCH might be applied in the development of functional food for intestinal microecological regulation.

Keywords: Gut microbiota, Peptides, Short-chain fatty acid, Yak bone collagen hydrolysate, Lachnospiraceae

References(35)

[1]

M.M. Or-Rashid, N.E. Odongo, B. Subedi, et al., Fatty acid composition of yak (Bos grunniens) cheese including conjugated linoleic acid and trans-18:1 fatty acids, J. Agric. Food Chem. 56(5) (2008) 1654-1660. https://doi.org/10.1021/jf0725225.

[2]

X.Y. Mao, H.Y. Yang, J.P. Song, et al., Effect of yak milk casein hydrolysate on Th1/Th2 cytokines production by murine spleen lymphocytes in vitro, J. Agric. Food Chem. 55(3) (2007) 638-642. https://doi.org/10.1021/jf062452m.

[3]

Q. Qiu, G. Zhang, T. Ma, et al., The yak genome and adaptation to life at high altitude, Nat. Genet. 44(8) (2012) 946-949. https://doi.org/10.1038/ng.2343.

[4]

S. Gao, H. Hong, C. Zhang, et al., Immunomodulatory effects of collagen hydrolysates from yak (Bos grunniens) bone on cyclophosphamide-induced immunosuppression in BALB/c mice, J. Funct. Foods 60 (2019) 103420. https://doi.org/10.1016/j.jff.2019.103420.

[5]

F. Li, D. Jia, K. Yao, Amino acid composition and functional properties of collagen polypeptide from yak (Bos grunniens) bone, LWT-Food Sci. Technol. 42(5) (2009) 945-949. https://doi.org/10.1016/j.lwt.2008.12.005.

[6]

G. Chen, L. Cheng, H. Xu, et al., Functions of different yak bone peptides, Int. J. Food Prop.14(5) (2011) 1136-1141. https://doi.org/10.1080/10942911003592753.

[7]

M. Ye, W. Jia, C. Zhang, et al., Valorization of yak (bos grunniens) bones as sources of functional ingredients, Waste Biomass Valorization 12 (2020) 1553-1564. https://doi.org/10.1007/s12649-020-01078-2.

[8]

J.M. Pickard, M.Y. Zeng, R. Caruso, et al., Gut microbiota: role in pathogen colonization, immune responses, and inflammatory disease, Immunol. Rev. 279(1) (2017) 70-89. https://doi.org/10.1111/imr.12567.

[9]

F. Backhed, R.E. Ley, J.L. Sonnenburg, et al., Host-bacterial mutualism in the human intestine, Science 307(5717) (2005) 1915-1920. https://doi.org/10.1126/science.1104816.

[10]

I. Sekirov, S.L. Russell, L.C.M. Antunes, et al., Gut microbiota in health and disease, Physiol. Rev. 90(3) (2010) 859-904. https://doi.org/10.1152/physrev.00045.2009.

[11]

X. Liu, S. Cao, X. Zhang, Modulation of gut microbiota brain axis by probiotics, prebiotics, and diet, J. Agric. Food Chem. 63(36) (2015) 7885-7895. https://doi.org/10.1021/acs.jafc.5b02404.

[12]

Y.J. Yu, M. Amorim, C. Marques, et al., Effects of whey peptide extract on the growth of probiotics and gut microbiota, J. Funct. Foods 21 (2016) 507-516. https://doi.org/10.1016/j.jff.2015.10.035.

[13]

Z. Karami, B. Akbari-adergani, Bioactive food derived peptides: a review on correlation between structure of bioactive peptides and their functional properties, J. Food Sci. Technol. 56(2) (2019) 535-547. https://doi.org/10.1007/s13197-018-3549-4.

[14]

J.T.J. Visser, N.A. Bos, L.F. Harthoorn, et al., Potential mechanisms explaining why hydrolyzed casein-based diets outclass single amino acid-based diets in the prevention of autoimmune diabetes in diabetes-prone BB rats, Diabetes Metab. Res. Rev. 28(6) (2012) 505-513. https://doi.org/10.1002/dmrr.2311.

[15]

Z. Guo, B. Hu, H. Han, et al., Metagenomic insights into the effects of nanobubble water on the composition of gut microbiota in mice, Food Funct. 11(8) (2020) 7175-7182. https://doi.org/10.1039/d0fo01592j.

[16]

Z. Guo, B. Hu, H.X. Wang, et al., Supplementation with nanobubble water alleviates obesity-associated markers through modulation of gut microbiota in high-fat diet fed mice, J. Funct. Foods 67 (2020) 103820. https://doi.org/10.1016/j.jff.2020.103820.

[17]

M. Ye, W. Jia, C. Zhang, et al., Preparation, identification and molecular docking study of novel osteoblast proliferation-promoting peptides from yak (Bos grunniens) bones, RSC Adv. 9(26) (2019) 14627-14637. https://doi.org/10.1039/C9RA00945K.

[18]

I. Lagkouvardos, T.R. Lesker, T.C.A. Hitch, et al., Sequence and cultivation study of Muribaculaceae reveals novel species, host preference, and functional potential of this yet undescribed family, Microbiome 7 (2019) 28. https://doi.org/10.1186/s40168-019-0637-2.

[19]

M. Vacca, G. Celano, F.M. Calabrese, et al., The controversial role of human gut Lachnospiraceae, Microorganisms 8(4) (2020) 573. https://doi.org/10.3390/microorganisms8040573.

[20]

E.W. Sayers, J. Beck, E.E. Bolton, et al., Database resources of the National Center for Biotechnology Information, Nucleic Acids Res. 49(D1) (2021) D10-D17. https://doi.org/10.1093/nar/gkaa892.

[21]

P.O. Sheridan, J.C. Martin, T.D. Lawley, et al., Polysaccharide utilization loci and nutritional specialization in a dominant group of butyrate-producing human colonic Firmicutes, Microb. Genomics. 2(2) (2016) e000043. https://doi.org/10.1099/mgen.0.000043.

[22]

M. De Angelis, I. Ferrocino, F.M. Calabrese, et al., Diet influences the functions of the human intestinal microbiome, Sci. Rep. 10(1) (2020) 4247. https://doi.org/10.1038/s41598-020-61192-y.

[23]

B.R. Di Iorio, M.T. Rocchetti, M. De Angelis, et al., Nutritional therapy modulates intestinal microbiota and reduces serum levels of total and free indoxyl sulfate and p-cresyl sulfate in chronic kidney disease (medika study), J. Clin. Med. 8(9) (2019) 1424. https://doi.org/10.3390/jcm8091424.

[24]

Z. Huawei, S.L. Ishaq, F.Q. Zhao, et al., Colonic inflammation accompanies an increase of beta-catenin signaling and Lachnospiraceae/Streptococcaceae bacteria in the hind gut of high-fat diet-fed mice, J. Nutr. Biochem. 35 (2016) 30-36. https://doi.org/10.1016/j.jnutbio.2016.05.015.

[25]

J. Qin, Y. Li, Z. Cai, et al., A metagenome-wide association study of gut microbiota in type 2 diabetes, Nature 490(7418) (2012) 55-60. https://doi.org/10.1038/nature11450.

[26]

A.D. Kostic, D. Gevers, H. Siljander, et al., The dynamics of the human infant gut microbiome in development and in progression toward type 1 diabetes, Cell Host Microbe. 17(2) (2015) 260-273. https://doi.org/10.1016/j.chom.2015.01.001.

[27]

F. Shen, R.D. Zheng, X.Q. Sun, et al., Gut microbiota dysbiosis in patients with non-alcoholic fatty liver disease, Hepatobiliary Pancreatic Dis. Int. 16(4) (2017) 375-381. https://doi.org/10.1016/S1499-3872(17)60019-5.

[28]

K.L. Ormerod, D.L.A. Wood, N. Lachner, et al., Genomic characterization of the uncultured Bacteroidales family S24-7 inhabiting the guts of homeothermic animals, Microbiome 4(1) (2016) 17. https://doi.org/10.1186/s40168-016-0181-2.

[29]

P.J. Turnbaugh, R.E. Ley, M.A. Mahowald, et al., An obesity-associated gut microbiome with increased capacity for energy harvest, Nature 444(7122) (2006) 1027-1031. https://doi.org/10.1038/nature05414.

[30]

J. Fernandes, W. Su, S. Rahat-Rozenbloom, et al., Adiposity, gut microbiota and faecal short chain fatty acids are linked in adult humans, Nutr. Diabetes 4(6) (2014) e121. https://doi.org/10.1038/nutd.2014.23.

[31]

B. Dalile, L. Van Oudenhove, B. Vervliet, et al., The role of short-chain fatty acids in microbiota-gut-brain communication, Nat. Rev. Gastroenterol. Hepatol. 16(8) (2019) 461-478. https://doi.org/10.1038/s41575-019-0157-3.

[32]

J.M. Laparra, Y. Sanz, Interactions of gut microbiota with functional food components and nutraceuticals, Pharmacol. Res. 61(3) (2010) 219-225. https://doi.org/10.1016/j.phrs.2009.11.001.

[33]

J. Zhao, X. Zhang, H. Liu, et al., Dietary protein and gut microbiota composition and function, Curr. Protein Pept. Sci. 20(2) (2019) 145-154. https://doi.org/10.2174/1389203719666180514145437.

[34]

U. Hynonen, P. Rasinkangas, R. Satokari, et al., Isolation and whole genome sequencing of a Ruminococcus-like bacterium, associated with irritable bowel syndrome, Anaerobe 39 (2016) 60-67. https://doi.org/10.1016/j.anaerobe.2016.03.001.

[35]

E.Y. Hsiao, S.W. McBride, S. Hsien, et al., Microbiota modulate behavioral and physiological abnormalities associated with neurodevelopmental disorders, Cell 155(7) (2013) 1451-1463. https://doi.org/10.1016/j.cell.2013.11.024.

Publication history
Copyright
Acknowledgements
Rights and permissions

Publication history

Received: 12 April 2021
Revised: 07 July 2021
Accepted: 01 August 2021
Published: 21 March 2023
Issue date: September 2023

Copyright

© 2023 Beijing Academy of Food Sciences.

Acknowledgements

Acknowledgements

The authors are very grateful for the technical support from the staff of the National Engineering Research Center for Functional Food, Jiangnan University. This work was supported by the Postdoctoral Research Funding of Jiangsu Province (2021K269B) and National Key Research & Developmental Program of China (2018YFA0900300).

Rights and permissions

This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Return