Journal Home > Volume 11 , Issue 2

This study was performed to determine the effects of bacteriocin-producing and non-bacteriocin-producing Pediococcus acidilactici strains on the immune system and intestinal flora of normal mice. Two P. acidilactici strains with antibacterial activity (P. acidilactici CCFM28 and CCFM18) were obtained based on the inhibition-zone assay. The produced components were identified as bacteriocins through protease treatment, pH adjustment and hydrogen peroxide treatment. Bacteriocin-producing and non-bacteriocin-producing P. acidilactici strains (P. acidilactici CCFM28, CCFM18 and NT17-3) caused significant changes in serum immune factors and intestinal flora of normal mice. After 14 days of intervention, the relative abundance of Firmicutes was significantly decreased, but that of Proteobacteria was significantly increased at the phylum level. At the genus level, the administration of three P. acidilactici strains resulted in the downregulation of Blautia and the upregulation of Ruminococcus and Lactobacillus. Furthermore, there were also different regulations on some probiotic strains, such as Bifidobacterium, Coprococcus and Akkermansia, which were closely related to the antibacterial ability of the bacteriocin and the type of strain. The results indicated that the intervention of different P. acidilactici strains could differently change the structure of intestinal flora in normal mice, which provided theoretical guidance for the selective use of bacteriocin-producing strains for health regulation in the future.


menu
Abstract
Full text
Outline
About this article

Effect of bacteriocin-producing Pediococcus acidilactici strains on the immune system and intestinal flora of normal mice

Show Author's information Yiteng Qiaoa,bZhichang QiucFengwei Tiana,bLeilei Yua,bJianxin Zhaoa,bHao Zhanga,b,d,eQixiao Zhaia,bWei Chena,b,d( )
State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
College of Food Science and Engineering, Shandong Agricultural University, Tai'an, Shandong 271018, China
National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
Wuxi Translational Medicine Research Center, Jiangsu Translational Medicine, Research Institute Wuxi Branch, Wuxi 214122, China

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

Abstract

This study was performed to determine the effects of bacteriocin-producing and non-bacteriocin-producing Pediococcus acidilactici strains on the immune system and intestinal flora of normal mice. Two P. acidilactici strains with antibacterial activity (P. acidilactici CCFM28 and CCFM18) were obtained based on the inhibition-zone assay. The produced components were identified as bacteriocins through protease treatment, pH adjustment and hydrogen peroxide treatment. Bacteriocin-producing and non-bacteriocin-producing P. acidilactici strains (P. acidilactici CCFM28, CCFM18 and NT17-3) caused significant changes in serum immune factors and intestinal flora of normal mice. After 14 days of intervention, the relative abundance of Firmicutes was significantly decreased, but that of Proteobacteria was significantly increased at the phylum level. At the genus level, the administration of three P. acidilactici strains resulted in the downregulation of Blautia and the upregulation of Ruminococcus and Lactobacillus. Furthermore, there were also different regulations on some probiotic strains, such as Bifidobacterium, Coprococcus and Akkermansia, which were closely related to the antibacterial ability of the bacteriocin and the type of strain. The results indicated that the intervention of different P. acidilactici strains could differently change the structure of intestinal flora in normal mice, which provided theoretical guidance for the selective use of bacteriocin-producing strains for health regulation in the future.

Keywords: Regulation, Pediococcus acidilactici, Bacteriocin, Immune system, Intestinal flora

References(28)

[1]

L.H. Deegan, P.D. Cotter, C. Hill, et al., Bacteriocins: biological tools for bio-preservation and shelf-life extension, Int. Dairy J. 16(9) (2006) 1058–1071. https://doi.org/10.1016/j.idairyj.2005.10.026.

[2]

S. Messaoudi, G. Kergourlay, M. Dalgalarrondo, et al., Purification and characterization of a new bacteriocin active against Campylobacter produced by Lactobacillus salivarius SMXD51, Food Microbiol. 32(1) (2012) 129–134. https://doi.org/10.1016/j.fm.2012.05.002.

[3]

T. Onda, F. Yanagida, M. Tsuji, et al., Production and purification of a bacteriocin peptide produced by Lactococcus sp. strain GM005, isolated from Miso-paste, Int. J. Food Microbiol. 87(1-2) (2003) 153–159. https://doi.org/10.1016/S0168-1605(03)00063-1.

[4]

S.D. Todorov, H. Prévost, M. Lebois, et al., Bacteriocinogenic Lactobacillus plantarum ST16Pa isolated from papaya (Carica papaya)—from isolation to application: characterization of a bacteriocin, Food Res. Int. 44(5) (2011) 1351–1363. https://doi.org/10.1016/j.foodres.2011.01.027.

[5]

S. Ennahar, T. Sashihara, K. Sonomoto, et al., Class Ⅱa bacteriocins: biosynthesis, structure and activity, FEMS Microbiol. Rev. 24(1) (2000) 85–106. https://doi.org/10.1016/S0168-6445(99)00031-5.

[6]

Gillor, A. Etzion, M.A. Riley, The dual role of bacteriocins as antiand probiotics, Appl. Microbiol. Biot. 81(4) (2008) 591–606. https://doi.org/10.1007/s00253-008-1726-5.

[7]

M.C. Walsh, G.E. Gardiner, O.M. Hart, et al., Predominance of a bacteriocin-producing Lactobacillus salivarius component of a five-strain probiotic in the porcine ileum and effects on host immune phenotype, FEMS Microbiol. Ecol. 64(2) (2008) 317–327. https://doi.org/10.1111/j.1574- 6941.2008.00454.x.

[8]

S.C. Corr, Y. Li, C.U. Riedel, et al., Bacteriocin production as a mechanism for the antiinfective activity of Lactobacillus salivarius UCC118, P. Natl. Acad. Sci. 104(18) (2007) 7617–7621. https://doi.org/10.1073/ pnas.0700440104.

[9]

M. Meijerink, S. Van Hemert, N. Taverne, et al., Identification of genetic loci in Lactobacillus plantarum that modulate the immune response of dendritic cells using comparative genome hybridization, PLoS One 5(5) (2010) e10632. https://doi.org/10.1371/journal.pone.0010632.

[10]

G. Cheng, H. Hao, S. Xie, et al., Antibiotic alternatives: the substitution of antibiotics in animal husbandry? Front. Microbiol. 5 (2014) 217. https://doi.org/10.3389/fmicb.2014.00217.

[11]

K. Skowron, J. Kwiecińska-Piróg, K. Grudlewska, et al., The occurrence, transmission, virulence and antibiotic resistance of Listeria monocytogenes in fish processing plant, Int. J. Food Microbiol. 282 (2018) 71–83. https://doi.org/10.1016/j.ijfoodmicro.2018.06.011.

[12]

K. Skowron, E. Wałecka-Zacharksa, K. Grudlewska, et al., Characteristics of Listeria monocytogenes strains isolated from milk and humans and the possibility of milk-borne strains transmission, Pol. J. Microbiol. 68(3) (2019) 353. https://doi.org/10.33073/pjm-2019-038.

[13]

A.B. Tahoun, R.M. Abou Elez, E.N. Abdelfatah, et al., Listeria monocytogenes in raw milk, milking equipment and dairy workers: molecular characterization and antimicrobial resistance patterns, J. Glob. Antimicrob. Res. 10 (2017) 264–270. https://doi.org/10.1016/j.jgar.2017.07.008.

[14]

S. Maher, S. McClean, Investigation of the cytotoxicity of eukaryotic and prokaryotic antimicrobial peptides in intestinal epithelial cells in vitro, Biochem. Pharmacol. 71(9) (2006) 1289–1298. https://doi.org/10.1016/ j.bcp.2006.01.012.

[15]

M.C. Rea, C.S. Sit, E. Clayton, et al., Thuricin CD, a posttranslationally modified bacteriocin with a narrow spectrum of activity against Clostridium difficile, P. Natl. Acad. Sci. 107(20) (2010) 9352–9357. https://doi.org/10.1073/pnas.0913554107.

[16]

L. Settanni, O. Massitti, D. Van Sinderen, et al., In situ activity of a bacteriocin-producing Lactococcus lactis strain. Influence on the interactions between lactic acid bacteria during sourdough fermentation, J. Appl. Microbiol. 99(3) (2005) 670–681. https://doi.org/10.1111/j.1365- 2672.2005.02647.x.

[17]

P.D. Cotter, Bioengineering: a bacteriocin perspective, Bioengineered 3(6) (2012) 313–319. https://doi.org/10.4161/bioe.21601.

[18]

M.R. Levengood, P.J. Knerr, T.J. Oman, et al., In vitro mutasynthesis of lantibiotic analogues containing nonproteinogenic amino acids, J. Am. Chem. Soc. 131(34) (2009) 12024–12025. https://doi.org/10.1021/ja903239s.

[19]

A.C. Ross, S.M. McKinnie, J.C. Vederas, The synthesis of active and stable diaminopimelate analogues of the lantibiotic peptide lactocin S, J. Am. Chem. Soc. 134(4) (2012) 2008–2011. https://doi.org/10.1021/ja211088m.

[20]

Y. Nagakura, Y. Naitoh, T. Kamato, et al., Compounds possessing 5-HT3 receptor antagonistic activity inhibit intestinal propulsion in mice, Eur. J. Pharmacol. 311 (1996) 67–72. https://doi.org/10.1016/0014-2999(96)00403-7.

[21]

Z. Qiu, N. Li, X. Lu, et al., Characterization of microbial community structure and metabolic potential using Illumina MiSeq platform during the black garlic processing, Food Res. Int. 106 (2018) 428–438. https://doi.org/10.1016/j.foodres.2017.12.081.

[22]

L. Zhao, G. Wang, P. Siegel, et al., Quantitative genetic background of the host influences gut microbiomes in chickens, Sci. Rep. 3(1) (2013) 1–6. https://doi.org/10.1038/srep01163.

[23]

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.

[24]

R.E. Ley, F. Bäckhed, P. Turnbaugh, et al., Obesity alters gut microbial ecology, P. Natl. Acad. Sci. 102(31) (2005) 11070–11075. https://doi.org/10.1073/pnas.0504978102.

[25]

N. Dabour, A. Zihler, E. Kheadr, et al., In vivo study on the effectiveness of pediocin PA-1 and Pediococcus acidilactici UL5 at inhibiting Listeria monocytogenes, Int. J. Food Microbiol. 133(3) (2009) 225–233. https://doi.org/10.1016/j.ijfoodmicro.2009.05.005.

[26]

H. Plovier, A. Everard, C. Druart, et al., A purified membrane protein from Akkermansia muciniphila or the pasteurized bacterium improves metabolism in obese and diabetic mice, Nat. Med. 23(1) (2017) 107–113. https://doi.org/10.1038/nm.4236.

[27]

P. Alvarez-Sieiro, M. Montalbán-López, D. Mu, et al., Bacteriocins of lactic acid bacteria: extending the family, Appl. Microbiol. Biot. 100(7) (2016) 2939–2951. https://doi.org/10.1007/s00253-016-7343-9.

[28]

Ö.C. Umu, C. Bäuerl, M. Oostindjer, et al., The potential of class Ⅱ bacteriocins to modify gut microbiota to improve host health, PLoS One 11(10) (2016) e0164036. https://doi.org/10.1371/journal.pone.0164036.

Publication history
Copyright
Acknowledgements
Rights and permissions

Publication history

Received: 26 March 2021
Revised: 02 April 2021
Accepted: 04 April 2021
Published: 25 November 2021
Issue date: March 2022

Copyright

© 2022 Beijing Academy of Food Sciences. Publishing services by Elsevier B.V. on behalf of KeAi Communications Co., Ltd.

Acknowledgements

Acknowledgements

This work was supported by the National Natural Science Foundation of China Program (32021005); the Natural Science Foundation of Jiangsu Province (BK20200084); Projects of Innovation and Development Pillar Program for Key Industries in Southern Xinjiang of Xinjiang Production and Construction Corps (2018DB002); National First Class Discipline Program of Food Science and Technology (JUFSTR20180102); the Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province.

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