Journal Home Online First Current Issue Archive For Authors Journal Information 中文版

Engineering >> 2022, Volume 17, Issue 10 doi: 10.1016/j.eng.2020.09.016

Phylogenetic and Comparative Genomic Analysis of Lactobacillus fermentum Strains and the Key Genes Related to Their Intestinal Anti-inflammatory Effects

a State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
b School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
c National Engineering Research Centre for Functional Food, Jiangnan University, Wuxi, 214122, China
d International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi, 214122, China
e Beijing Innovation Centre of Food Nutrition and Human Health, Beijing Technology and Business University, Beijing, 100048, China

Received: 2020-03-16 Revised: 2020-09-13 Accepted: 2020-09-24 Available online: 2021-02-05

Next Previous

Abstract

Highlights

•Pan-genomic and phylogenetic characterizations of 109 L. fermentum strains were performed.

•No co-evolutionary relationship exists between L. fermentum strains and the geographical origins of their host Immunomodulatory properties of L. fermentum were strain-dependent.

•Some specific genes may account for the anti-inflammatory and immunoregulation of L. fermentum strains.

Emerging evidence shows that some Lactobacillus fermentum strains can contribute to the prevention and treatment of ulcerative colitis (UC). In this study, 105 isolates of L. fermentum strains were separated from fecal samples of populations in different regions in China and their draft genomes were sequenced. Pan-genomic and phylogenetic characterizations of these strains and four model strains (L. fermentum 3872, CECT5716, IFO3956, and VRI003) were performed. Phylogenetic analysis indicated that there was no significant adaptive evolution between the genomes of L. fermentum strains and the geographical location, sex, ethnicity, and age of the hosts. Three L. fermentum strains (FWXBH115, FGDLZR121, and FXJCJ61) from different branches of the phylogenetic tree and strain type L. fermentum CECT5716 were selected and their anti-inflammatory and immune modulatory activities in a dextran sulphate sodium (DSS)-induced colitis mouse model were further investigated. Both L. fermentum FXJCJ61 and CECT5716 significantly alleviated UC by reducing all colitis-associated histological indices, maintaining mucosal integrity, and stimulating replenishment of short-chain fatty acids (SCFAs), while the other two strains failed to offer similar protection. The anti-inflammatory mechanisms of L. fermentum FXJCJ61 and CECT5716 were related to the inhibition of nuclear factor kappa-B (NF-κB) signaling pathway activation and enhancement of interleukin 10 (IL-10) production. Comparative genomic analysis of these strains identified candidate genes that may contribute to the anti-inflammatory effects of specific L. fermentum strains.

SupplementaryMaterials

Figures

Fig. 1

Fig. 2

Fig. 3

Fig. 4

Fig. 5

Fig. 6

Fig. 7

Fig. 8

Fig. 9

References

[ 1 ]

[ 2 ] Archer AC, Halami PMJAM. Probiotic attributes of Lactobacillus fermentum isolated from human feces and dairy products. Appl Microbiol Biotechnol 2015;99(19):8113–23.

[ 3 ] Lin WH, Yu B, Jang SH, Tsen HY. Different probiotic properties for Lactobacillus fermentum strains isolated from swine and poultry. Anaerobe 2007;13(3– 4):107–13.

[ 4 ] Morita H, Toh H, Fukuda S, Horikawa H, Oshima K, Suzuki T, et al. Comparative genome analysis of Lactobacillus reuteri and Lactobacillus fermentum reveal a genomic island for reuterin and cobalamin production. DNA Res 2008;15 (3):151–61.

[ 5 ] Jayashree S, Pooja S, Pushpanathan M, Rajendhran J, Gunasekaran P. Identification and characterization of bile salt hydrolase genes from the genome of Lactobacillus fermentum MTCC 8711. Appl Biochem Biotechnol 2014;174(2):855–66.

[ 6 ] Dan T, Fukuda K, Sugai-Bannai M, Takakuwa N, Motoshima H, Urashima T. Characterization and expression analysis of the exopolysaccharide gene cluster in Lactobacillus fermentum TDS030603. Biosci Biotechnol Biochem 2009;73 (12):2656–64.

[ 7 ] Oloyede AR, Albert OM, Arowosegbe OA. Detection and molecular characterization of butyrate-producing genes in probiotic lactic acid bacteria for use in livestock. Niger J Biotechnol 2017;33(1):58–65.

[ 8 ] Makarova K, Slesarev A, Wolf Y, Sorokin A, Mirkin B, Koonin E, et al. Comparative genomics of the lactic acid bacteria. Proc Natl Acad Sci USA 2006;103(42):15611–6.

[ 9 ] Mendes-Soares H, Suzuki H, Hickey RJ, Forney LJ. Comparative functional genomics of Lactobacillus spp. reveals possible mechanisms for specialization of vaginal lactobacilli to their environment. J Bacteriol 2014;196(7):1458–70.

[10] O’Sullivan O, O’Callaghan J, Sangrador-Vegas A, Mcauliffe O, Slattery L, Kaleta P, et al. Comparative genomics of lactic acid bacteria reveals a niche-specific gene set. BMC Microbiol 2009;9(1):50.

[11] Siezen RJ, Tzeneva VA, Castioni A, Wels M, Phan HTK, Rademaker JLW, et al. Phenotypic and genomic diversity of Lactobacillus plantarum strains isolated from various environmental niches. Environ Microbiol Rep 2010;12 (3):758–73.

[12] Zheng J, Zhao X, Lin XB, Ganzle M. Comparative genomics Lactobacillus reuteri from sourdough reveals adaptation of an intestinal symbiont to food fermentations. Sci Rep 2015;5(1):18234.

[13] Kant R, Rintahaka J, Yu X, Sigvart-Mattila P, Paulin L, Mecklin JP, et al. A comparative pan-genome perspective of niche-adaptable cell-surface protein phenotypes in Lactobacillus rhamnosus. PLoS ONE 2014;9(7).

[14] Cai H, Thompson R, Budinich MF, Broadbent JR, Steele JLJGB. Genome sequence and comparative genome analysis of Lactobacillus casei: insights into their niche-associated evolution. Genome Biol Evol 2009;1:239–57.

[15] Oh PL, Benson AK, Peterson DA, Patil PB, Moriyama EN, Roos S, et al. Diversification of the gut symbiont Lactobacillus reuteri as a result of hostdriven evolution. ISME J 2010;4(3):377–87.

[16] Dan T, Liu W, Song Y, Xu H, Menghe B, Zhang H, et al. The evolution and population structure of Lactobacillus fermentum from different naturally fermented products as determined by multilocus sequence typing (MLST). BMC Microbiol 2015;15(1):107.

[17] Silvi S, Verdenelli MC, Orpianesi C, Cresci A. Cresci AJJoFE. EU project Crownalife: functional foods, gut microflora and healthy ageing:isolation and identification of Lactobacillus and Bifidobacterium strains from faecal samples of elderly subjects for a possible probiotic use in functional foods. J Food Eng 2003;56(2-3):195–200.

[18] Morandi S, Brasca M, Lodi R. Technological, phenotypic and genotypic characterisation of wild lactic acid bacteria involved in the production of Bitto PDO Italian cheese. Dairy Sci Technol 2011;91(3):341–59.

[19] Blomberg L, Henriksson A, Conway PL. Inhibition of adhesion of Escherichia coli K88 to piglet ileal mucus by Lactobacillus spp.. Appl Environ Microbiol 1993;59(1):34–9.

[20] Lehri B, Seddon AM, Karlyshev AV. Lactobacillus fermentum 3872 as a potential tool for combatting Campylobacter jejuni infections. Virulence 2017;8(8):1753–60.

[21] Kim BK, Lee IO, Tan PL, Eor JY, Hwang JK, Kim SH. Protective effect of Lactobacillus fermentum LA12 in an alcohol-induced rat model of alcoholic steatohepatitis. Korean J Food Sci Anim Resour 2017;37:931–9.

[22] Kullisaar T, Zilmer K, Salum T, Rehema A, Zilmer M. The use of probiotic L. fermentum ME-3 containing reg’activ cholesterol supplement for 4 weeks has a positive influence on blood lipoprotein profiles and inflammatory cytokines: an open-label preliminary study. Nutr J 2016;15(1):93.

[23] Chen X, Zhao X, Wang H, Yang Z, Li J, Suo H. Prevent effects of Lactobacillus fermentum HY01 on dextran sulfate sodium-induced colitis in mice. Nutrients 2017;9(6):545.

[24] Zhao Y, Hong K, Zhao J, Zhang H, Zhai Q, Chen W. Lactobacillus fermentum and its potential immunomodulatory properties. J Funct Foods 2019;56:21–32.

[25] Rodríguez-Nogales A, Algieri F, Garrido-Mesa J, Vezza T, Utrilla MP, Chueca N, et al. Differential intestinal anti-inflammatory effects of Lactobacillus fermentum and Lactobacillus salivarius in DSS mouse colitis: impact on microRNAs expression and microbiota composition. Mol Nutr Food Res 2017;61(11):1700144.

[26] Chery J, Dvoskin D, Morato FP, Fahoum B. Lactobacillus fermentum, a pathogen in documented cholecystitis. Int J Surg Case Rep 2013;4(8):662–4.

[27] Anderson RC, Ulluwishewa D, Young W, Ryan LJ, Henderson G, Meijerink M, et al. Human oral isolate Lactobacillus fermentum AGR1487 induces a proinflammatory response in germ-free rat colons. Sci Rep 2016;6(1):20318.

[28] Yao L, Seaton SC, Ndousse-Fetter S, Adhikari AA, DiBenedetto N, Mina AI. A selective gut bacterial bile salt hydrolase alters host metabolism. eLife 2018;7: e37182.

[29] Guo CJ, Allen BM, Hiam KJ, Dodd D, Van Treuren W, Higginbottom S, et al. Depletion of microbiome-derived molecules in the host using Clostridium genetics. Science 2019;366(6471):eaav1282.

[30] Kumar R, Rajkumar H, Kumar M, Varikuti SR, Athimamula R, Shujauddin M, et al. Molecular cloning, characterization and heterologous expression of bile salt hydrolase (Bsh) from Lactobacillus fermentum NCDO394. Mol Biol Rep 2013;40(8):5057–66.

[31] Hartemink R, Domenech VR, Rombouts FM. LAMVAB—a new selective medium for the isolation of lactobacilli from faeces. J Microbiol Methods 1997;29 (2):77–84.

[32] Zheng J, Ruan L, Sun M, Ganzle M. A genomic view of Lactobacilli and Pediococci demonstrates that phylogeny matches ecology and physiology. Appl Environ Microbiol 2015;81(20):7233–43.

[33] Verma R, Lee C, Jeun EJ, Yi J, Kim KS, Ghosh A. Cell surface polysaccharides of Bifidobacterium bifidum induce the generation of Foxp3+ regulatory T cells. Sci Immunol 2018;3(28):eaat6975.

[34] Liu HL, Zhang YL, Yang N, Zhang YX, Liu XQ, Li CG, et al. A functionalized single-walled carbon nanotube-induced autophagic cell death in human lung cells through Akt-TSC2-mTOR signaling. Cell Death Dis 2011;2(5).

[35] Zhang Z, Wu X, Cao S, Wang L, Wang D, Yang H, et al. Caffeic acid ameliorates colitis in association with increased Akkermansia population in the gut microbiota of mice. Oncotarget 2016;7(22):31790–9.

[36] Zhai Q, Wang G, Zhao J, Liu X, Tian F, Zhang H, et al. Protective effects of Lactobacillus plantarum CCFM8610 against acute cadmium toxicity in mice. Appl Environ Microbiol 2013;79(5):1508–15.

[37] Tan H, Zhao J, Zhang H, Zhai Q, Chen WJAM. Novel strains of Bacteroides fragilis and Bacteroides ovatus alleviate the LPS-induced inflammation in mice. Appl Microbiol Biotechnol 2019;103(5):2353–65.

[38] Xu Q, Li X, Wang E, He Y, Yin B, Fang D, et al. A cellular model for screening of lactobacilli that can enhance tight junctions. RSC Adv 2016;6 (113):111812–21.

[39] Mao B, Li D, Ai C, Zhao J, Zhang H, Chen W. Lactulose differently modulates the composition of luminal and mucosal microbiota in C57BL/6J mice. J Agric Food Chem 2016;64(31):6240–7.

[40] Sun Y, Jin C, Zhang X, Jia W, Le J, Ye J. Restoration of GLP-1 secretion by Berberine is associated with protection of colon enterocytes from mitochondrial overheating in diet-induced obese mice. Nutr Diabetes 2018;8 (1):53.

[41] Song YL, Kato N, Liu CX, Matsumiya Y, Kato H, Watanabe KJFML. Rapid identification of 11 human intestinal Lactobacillus species by multiplex PCR assays using group- and species-specific primers derived from the 16S–23S rRNA intergenic spacer region and its flanking 23S rRNA. FEMS Microbiol Lett 2000;187(2):167–73.

[42] Bahl MI, Bergstrom A, Licht TR. Freezing fecal samples prior to DNA extraction affects the Firmicutes to Bacteroidetes ratio determined by downstream quantitative PCR analysis. FEMS Microbiol Lett 2012;329(2):193–7.

[43] Li P, Gu Q, Yang L, Yu Y, Wang Y. Characterization of extracellular vitamin B12 producing Lactobacillus plantarum strains and assessment of the probiotic potentials. Food Chem 2017;234:494–501.

[44] Sulemankhil I, Parent M, Jones ML, Feng Z, Labbe A, Prakash S. In vitro and in vivo characterization and strain safety of Lactobacillus reuteri NCIMB 30253 for probiotic applications. Can J Microbiol 2012;58(6):776–87.

[45] Moran NA, Sloan DB. The hologenome concept: helpful or hollow? PLoS Biol 2015;13(12).

[46] Groussin M, Mazel F, Sanders JG, Smillie CS, Lavergne S, Thuiller W, et al. Unraveling the processes shaping mammalian gut microbiomes over evolutionary time. Nat Commun 2017;8(1):14319.

[47] Duar RM, Lin XB, Zheng J, Martino ME, Grenier T, Perez-Munoz ME, et al. Lifestyles in transition: evolution and natural history of the genus Lactobacillus. FEMS Microbiol Rev 2017;41(Supp 1):S27–48.

[48] Guo X, Li S, Zhang J, Wu F, Li X, Wu D, et al. Genome sequencing of 39 Akkermansia muciniphila isolates reveals its population structure, genomic and functional diversity, and global distribution in mammalian gut microbiotas. BMC Genomics 2017;18(1):800.

[49] Ottman N, Reunanen J, Meijerink M, Pietila TE, Kainulainen V, Klievink J, et al. Pili-like proteins of Akkermansia muciniphila modulate host immune responses and gut barrier function. PLoS ONE 2017;12(3).

[50] Pandurangan AK, Ismail S, Saadatdoust Z, Esa NM. Allicin alleviates dextran sodium sulfate- (DSS-) induced ulcerative colitis in BALB/c mice. Oxid Med Cell Longev 2015;2015.

[51] Cario E. Toll-like receptors in inflammatory bowel diseases: a decade later. Inflamm Bowel Dis 2010;16(9):1583–97.

[52] Takagawa T, Kitani A, Fuss I, Levine B, Brant SR, Peter I, et al. An increase in LRRK2 suppresses autophagy and enhances Dectin-1-induced immunity in a mouse model of colitis. Sci Transl Med 2018;10(444):eaan8162.

[53] Mittal SK, Roche PA. Suppression of antigen presentation by IL-10. Curr Opin Immunol 2015;34:22–7.

[54] Onizawa M, Nagaishi T, Kanai T, Nagano K, Oshima S, Nemoto Y, et al. Signaling pathway via TNF-alpha/NF-kappaB in intestinal epithelial cells may be directly involved in colitis-associated carcinogenesis. Am J Physiol Gastrointest Liver Physiol 2009;296(4):G850–9.

[55] Price MN, Wetmore KM, Waters RJ, Callaghan M, Ray J, Liu H, et al. Mutant phenotypes for thousands of bacterial genes of unknown function. Nature 2018;557(7706):503–9.

[56] Spaulding CN, Klein RD, Ruer S, Kau AL, Schreiber HL, Cusumano ZT, et al. Selective depletion of uropathogenic E. coli from the gut by a FimH antagonist. Nature 2017;546(7659):528–32.

[57] Yang J, Li Q, Henning SM, Zhong J, Hsu M, Lee R, et al. Effects of prebiotic fiber xylooligosaccharide in adenine-induced nephropathy in mice. Mol Nutr Food Res 2018;62(15):1800014.

[58] Adachi O, Fujii Y, Ano Y, Moonmangmee D, Toyama H, Shinagawa E, et al. Membrane-bound sugar alcohol dehydrogenase in acetic acid bacteria catalyzes L-ribulose formation and NAD-dependent ribitol dehydrogenase is independent of the oxidative fermentation. Biosci Biotechnol Biochem 2001;65(1):115–25.

[59] Wen L, Wong FS. Dietary short-chain fatty acids protect against type 1 diabetes. Nat Immunol 2017;18(5):484–6.

[60] Zagato E, Pozzi C, Bertocchi A, Schioppa T, Saccheri F, et al. Endogenous murine microbiota member Faecalibaculum rodentium and its human homologue protect from intestinal tumour growth. Nat Microbiol 2020;5(3):511–24.

[61] Smith PM, Howitt MR, Panikov N, Michaud M, Gallini CA, Bohlooly-Y M, et al. the microbial metabolites, short-chain fatty acids, regulate colonic treg cell homeostasis. Science 2013;341(6145):569–73.

[62] Mishiro T, Kusunoki R, Otani A, Ansary MM, Tongu M, Harashima N, et al. Butyric acid attenuates intestinal inflammation in murine DSS-induced colitis model via milk fat globule-EGF factor 8. Lab Invest 2013;93(7):834–43.

[63] Laffin M, Fedorak R, Zalasky A, Park H, Gill A, Agrawal A, et al. A high-sugar diet rapidly enhances susceptibility to colitis via depletion of luminal short-chain fatty acids in mice. Sci Rep 2019;9(1):12294.

[64] Wang M, Wichienchot S, He X, Fu X, Huang Q, Zhang B. In vitro colonic fermentation of dietary fibers: fermentation rate, short-chain fatty acid production and changes in microbiota. Trends Food Sci Technol 2019;88:1–9.

[65] Tajik N, Frech M, Schulz O, Schälter F, Lucas S, Azizov V, et al. Targeting zonulin and intestinal epithelial barrier function to prevent onset of arthritis. Nat Commun 2020;11(1):1995.

[66] Yang C, Gao J, Zhang J, Luo AL. Enterochromaffin cells in the gut: a distant regulator of brain function? Gut 2018;67(8):1557–8.

[67] Lee YS, Kim TY, Kim Y, Lee SH, Kim S, Kang SW, et al. Microbiota-derived lactate accelerates intestinal stem-cell-mediated epithelial development. Cell Host Microbe 2018;24(6):833–46.

[68] Iraporda C, Errea A, Romanin DE, Cayet D, Pereyra E, Pignataro O, et al. Lactate and short chain fatty acids produced by microbial fermentation downregulate proinflammatory responses in intestinal epithelial cells and myeloid cells. Immunobiology 2015;220(10):1161–9.

[69] Witkin SS, Linhares IM. Why do lactobacilli dominate the human vaginal microbiota? BJOG 2017;124(4):606–11.

[70] Wulandari AP, Miyazaki J, Kobashi N, Nishiyama M, Hoshino T, Yamane H. Characterization of bacterial homocitrate synthase involved in lysine biosynthesis. FEBS Lett 2002;522(1–3):35–40.

[71] Wu R, Sun Z, Wu J, Meng H, Zhang H. Effect of bile salts stress on protein synthesis of Lactobacillus casei Zhang revealed by 2-dimensional gel electrophoresis. J Dairy Sci 2010;93(8):3858–68.

[72] Blount KF, Wang JX, Lim J, Sudarsan N, Breaker RR. Antibacterial lysine analogs that target lysine riboswitches. Nat Chem Biol 2007;3(1):44–9.

[73] Goh YJ, Barrangou R. Harnessing CRISPR-Cas systems for precision engineering of designer probiotic lactobacilli. Curr Opin Biotechnol 2019;56:163–71.

Related Research