1887

Abstract

The use of bacterial spores in probiotics over viable loads of bacteria has many advantages, including the durability of spores, which allows spore-based probiotics to effectively traverse the various biochemical barriers present in the gastrointestinal tract. However, the majority of spore-based probiotics developed currently aim to treat adults, and there is a litany of differences between the adult and infant intestinal systems, including the immaturity and low microbial species diversity observed within the intestines of infants. These differences are only further exacerbated in premature infants with necrotizing enterocolitis (NEC) and indicates that what may be appropriate for an adult or even a healthy full-term infant may not be suited for an unhealthy premature infant. Complications from using spore-based probiotics for premature infants with NEC may involve the spores remaining dormant and adhering to the intestinal epithelia, the out-competing of commensal bacteria by spores, and most importantly the innate antibiotic resistance of spores. Also, the ability of to produce spores under duress may result in less perishing within the intestines and releasing membrane branched-chain fatty acids. The isolate BG01-4 is a proprietary strain developed by Vernx Biotechnology through accumulating mutations within the BG01-4 genome in a serial batch culture. Strain BG01-4 was provided as a non-spore-forming , but a positive sporulation status for BG01-4 was confirmed through testing and suggested that selection for the sporulation defective genes could occur within an environment that would select against sporulation. The durability of key sporulation genes was ratified in this study, as the ability of BG01-4 to produce spores was not eliminated by the attempts to select against sporulation genes in BG01-4 by the epigenetic factors of high glucose and low pH. However, a variation in the genes in isolate BG01-4-8 involved in the regulation of sporulation is believed to have occurred during the mutation selection from the parent strain BG01-4. An alteration in selected sporulation regulation genes is expected to have occurred from BG01-4 to BG01-4-8, with BG01-4-8 producing spores within 24 h, ~48 h quicker than BG01-4.

Funding
This study was supported by the:
  • Vernx Biotechnology Pty & La Trobe University (Award N/A)
    • Principle Award Recipient: LukeM Bosnar
  • This is an open-access article distributed under the terms of the Creative Commons Attribution License. This article was made open access via a Publish and Read agreement between the Microbiology Society and the corresponding author’s institution.
Loading

Article metrics loading...

/content/journal/acmi/10.1099/acmi.0.000419
2023-05-26
2024-04-23
Loading full text...

Full text loading...

/deliver/fulltext/acmi/5/5/acmi000419.html?itemId=/content/journal/acmi/10.1099/acmi.0.000419&mimeType=html&fmt=ahah

References

  1. Ran-Ressler RR, Khailova L, Arganbright KM, Adkins-Rieck CK, Jouni ZE et al. Branched chain fatty acids reduce the incidence of necrotizing enterocolitis and alter gastrointestinal microbial ecology in a neonatal rat model. PLoS One 2011; 6:e29032 [View Article] [PubMed]
    [Google Scholar]
  2. Lin PW, Stoll BJ. Necrotising enterocolitis. Lancet 2006; 368:1271–1283 [View Article] [PubMed]
    [Google Scholar]
  3. Bell MJ, Ternberg JL, Feigin RD, Keating JP, Marshall RI et al. Neonatal necrotizing enterocolitis. Therapeutic decisions based upon clinical staging. Ann Surg 1978; 187:1–7 [View Article] [PubMed]
    [Google Scholar]
  4. Aagaard K, Ma J, Antony KM, Ganu R, Petrosino J et al. The placenta harbors a unique microbiome. Sci Transl Med 2014; 6:237ra65 [View Article] [PubMed]
    [Google Scholar]
  5. He Q, Kwok LY, Xi X, Zhong Z, Ma T et al. The meconium microbiota shares more features with the amniotic fluid microbiota than the maternal fecal and vaginal microbiota. Gut Microbes 2020; 12:1794266 [View Article] [PubMed]
    [Google Scholar]
  6. Collado MC, Rautava S, Aakko J, Isolauri E, Salminen S. Human gut colonisation may be initiated in utero by distinct microbial communities in the placenta and amniotic fluid. Sci Rep 2016; 6:23129 [View Article] [PubMed]
    [Google Scholar]
  7. Ferretti P, Pasolli E, Tett A, Asnicar F, Gorfer V et al. Mother-to-infant microbial transmission from different body sites shapes the developing infant gut microbiome. Cell Host Microbe 2018; 24:133–145 [View Article] [PubMed]
    [Google Scholar]
  8. Ríos-Covián D, Ruas-Madiedo P, Margolles A, Gueimonde M, de Los Reyes-Gavilán CG et al. Intestinal short chain fatty acids and their link with diet and human health. Front Microbiol 2016; 7:185 [View Article] [PubMed]
    [Google Scholar]
  9. Yan Y, Wang Z, Wang D, Lawrence P, Wang X et al. BCFA-enriched vernix-monoacylglycerol reduces LPS-induced inflammatory markers in human enterocytes in vitro. Pediatr Res 2018; 83:874–879 [View Article] [PubMed]
    [Google Scholar]
  10. Nishijima K, Yoneda M, Hirai T, Takakuwa K, Enomoto T. Biology of the vernix caseosa: A review. J Obstet Gynaecol Res 2019; 45:2145–2149 [View Article] [PubMed]
    [Google Scholar]
  11. Visscher MO, Hoath SB. Vernix caseosa and its substitutes: lipid composition and physicochemical properties. In Treatment of Dry Skin Syndrome Berlin, Heidelberg: Springer; 2012 pp 193–213 [View Article]
    [Google Scholar]
  12. Ran-Ressler RR, Glahn RP, Bae S, Brenna JT. Branched-chain fatty acids in the neonatal gut and estimated dietary intake in infancy and adulthood. Nestle Nutr Inst Workshop Ser 2013; 77:133–143 [View Article] [PubMed]
    [Google Scholar]
  13. Ran-Ressler RR, Devapatla S, Lawrence P, Brenna JT. Branched chain fatty acids are constituents of the normal healthy newborn gastrointestinal tract. Pediatr Res 2008; 64:605–609 [View Article] [PubMed]
    [Google Scholar]
  14. Cong X, Xu W, Janton S, Henderson WA, Matson A et al. Gut microbiome developmental patterns in early life of preterm infants: impacts of feeding and gender. PLoS One 2016; 11:e0152751 [View Article] [PubMed]
    [Google Scholar]
  15. Rinninella E, Raoul P, Cintoni M, Franceschi F, Miggiano GAD et al. What is the healthy gut microbiota composition? a changing ecosystem across age, environment, diet, and diseases. Microorganisms 2019; 7:14 [View Article] [PubMed]
    [Google Scholar]
  16. Stewart CJ, Embleton ND, Marrs ECL, Smith DP, Fofanova T et al. Longitudinal development of the gut microbiome and metabolome in preterm neonates with late onset sepsis and healthy controls. Microbiome 2017; 5:1–11 [View Article]
    [Google Scholar]
  17. Zhou Y, Shan G, Sodergren E, Weinstock G, Walker WA et al. Longitudinal analysis of the premature infant intestinal microbiome prior to necrotizing enterocolitis: a case-control study. PLoS One 2015; 10:e0118632 [View Article] [PubMed]
    [Google Scholar]
  18. Hagen PC, Skelley JW. Efficacy of Bifidobacterium species in prevention of necrotizing enterocolitis in very-low birth weight infants. A systematic review. J Pediatr Pharmacol Ther 2019; 24:10–15 [View Article] [PubMed]
    [Google Scholar]
  19. Morrow AL, Lagomarcino AJ, Schibler KR, Taft DH, Yu Z et al. Early microbial and metabolomic signatures predict later onset of necrotizing enterocolitis in preterm infants. Microbiome 2013; 1:1–16 [View Article]
    [Google Scholar]
  20. Coggins SA, Wynn JL, Weitkamp J-H. Infectious causes of necrotizing enterocolitis. Clin Perinatol 2015; 42:133–154 [View Article] [PubMed]
    [Google Scholar]
  21. Hoffmann KM, Deutschmann A, Weitzer C, Joainig M, Zechner E et al. Antibiotic-associated hemorrhagic colitis caused by cytotoxin-producing Klebsiella oxytoca. Pediatrics 2010; 125:e960–e963 [View Article]
    [Google Scholar]
  22. Denning N-L, Prince JM. Neonatal intestinal dysbiosis in necrotizing enterocolitis. Mol Med 2018; 24:1–10 [View Article]
    [Google Scholar]
  23. Paveglio S, Ledala N, Rezaul K, Lin Q, Zhou Y et al. Cytotoxin-producing Klebsiella oxytoca in the preterm gut and its association with necrotizing enterocolitis. Emerg Microbes Infect 2020; 9:1321–1329 [View Article] [PubMed]
    [Google Scholar]
  24. Olm MR, Bhattacharya N, Crits-Christoph A, Firek BA, Baker R et al. Necrotizing enterocolitis is preceded by increased gut bacterial replication, Klebsiella, and fimbriae-encoding bacteria. Sci Adv 2019; 5:eaax5727 [View Article] [PubMed]
    [Google Scholar]
  25. Cassir N, Simeoni U, La Scola B. Gut microbiota and the pathogenesis of necrotizing enterocolitis in preterm neonates. Future Microbiol 2016; 11:273–292 [View Article] [PubMed]
    [Google Scholar]
  26. Kiu R, Hall LJ. An update on the human and animal enteric pathogen Clostridium perfringens. Emerg Microbes Infect 2018; 7:141 [View Article] [PubMed]
    [Google Scholar]
  27. Laursen MF. Gut microbiota development: influence of diet from infancy to toddlerhood. Ann Nutr Metab 2021; 77:21–34 [View Article]
    [Google Scholar]
  28. Cho SX, Rudloff I, Lao JC, Pang MA, Goldberg R et al. Characterization of the pathoimmunology of necrotizing enterocolitis reveals novel therapeutic opportunities. Nat Commun 2020; 11:1–9 [View Article]
    [Google Scholar]
  29. Zheng N, Gao Y, Zhu W, Meng D, Walker WA. Short chain fatty acids produced by colonizing intestinal commensal bacterial interaction with expressed breast milk are anti-inflammatory in human immature enterocytes. PLoS One 2020; 15:e0229283 [View Article] [PubMed]
    [Google Scholar]
  30. Schreurs RRCE, Baumdick ME, Sagebiel AF, Kaufmann M, Mokry M et al. Human fetal TNF-α-cytokine-producing CD4+ effector memory T cells promote intestinal development and mediate inflammation early in life. Immunity 2019; 50:462–476 [View Article] [PubMed]
    [Google Scholar]
  31. Werts AD, Fulton WB, Ladd MR, Saad-Eldin A, Chen YX et al. A novel role for necroptosis in the pathogenesis of necrotizing enterocolitis. Cell Mol Gastroenterol Hepatol 2020; 9:403–423 [View Article] [PubMed]
    [Google Scholar]
  32. Arboleya S, Binetti A, Salazar N, Fernández N, Solís G et al. Establishment and development of intestinal microbiota in preterm neonates. FEMS Microbiol Ecol 2012; 79:763–772 [View Article] [PubMed]
    [Google Scholar]
  33. Warner BB, Deych E, Zhou Y, Hall-Moore C, Weinstock GM et al. Gut bacteria dysbiosis and necrotising enterocolitis in very low birthweight infants: a prospective case-control study. Lancet 2016; 387:1928–1936 [View Article] [PubMed]
    [Google Scholar]
  34. Mai V, Young CM, Ukhanova M, Wang X, Sun Y et al. Fecal microbiota in premature infants prior to necrotizing enterocolitis. PLoS One 2011; 6:e20647 [View Article] [PubMed]
    [Google Scholar]
  35. Dardas M, Gill SR, Grier A, Pryhuber GS, Gill AL et al. The impact of postnatal antibiotics on the preterm intestinal microbiome. Pediatr Res 2014; 76:150–158 [View Article] [PubMed]
    [Google Scholar]
  36. Bury RG, Tudehope D. Enteral antibiotics for preventing necrotizing enterocolitis in low birthweight or preterm infants. Cochrane Database Syst Rev 2001; 2001:CD000405 [View Article] [PubMed]
    [Google Scholar]
  37. Pammi M, Cope J, Tarr PI, Warner BB, Morrow AL et al. Intestinal dysbiosis in preterm infants preceding necrotizing enterocolitis: a systematic review and meta-analysis. Microbiome 2017; 5:31 [View Article] [PubMed]
    [Google Scholar]
  38. Li Y, Shen RL, Ayede AI, Berrington J, Bloomfield FH et al. Early use of antibiotics is associated with a lower incidence of necrotizing enterocolitis in preterm, very low birth weight infants: the NEOMUNE-NeoNutriNet cohort study. J Pediatr 2020; 227:128–134 [View Article] [PubMed]
    [Google Scholar]
  39. Maier L, Goemans CV, Wirbel J, Kuhn M, Eberl C et al. Unravelling the collateral damage of antibiotics on gut bacteria. Nature 2021; 599:120–124 [View Article] [PubMed]
    [Google Scholar]
  40. Alexander VN, Northrup V, Bizzarro MJ. Antibiotic exposure in the newborn intensive care unit and the risk of necrotizing enterocolitis. J Pediatr 2011; 159:392–397 [View Article] [PubMed]
    [Google Scholar]
  41. Hartantyo SHP, Chau ML, Koh TH, Yap M, Yi T et al. Foodborne Klebsiella pneumoniae: virulence potential, antibiotic resistance, and risks to food safety. J Food Prot 2020; 83:1096–1103 [View Article] [PubMed]
    [Google Scholar]
  42. Baghani A, Ghourchian S, Aliramezani A, Yaseri M, Mesdaghinia A et al. Highly antibiotic-resistant Clostridium difficile isolates from Iranian patients. J Appl Microbiol 2018; 125:1518–1525 [View Article] [PubMed]
    [Google Scholar]
  43. Paredes-Sabja D, Shen A, Sorg JA. Clostridium difficile spore biology: sporulation, germination, and spore structural proteins. Trends Microbiol 2014; 22:406–416 [View Article] [PubMed]
    [Google Scholar]
  44. Jiang YN, Muk T, Stensballe A, Nguyen DN, Sangild PT et al. Early protein markers of necrotizing enterocolitis in plasma of preterm pigs exposed to antibiotics. Front Immunol 2020; 11:565862 [View Article] [PubMed]
    [Google Scholar]
  45. Schanler RJ. Probiotics and necrotising enterocolitis in premature infants. Arch Dis Child Fetal Neonatal Ed 2006; 91:F395–7 [View Article] [PubMed]
    [Google Scholar]
  46. Silverman MA, Konnikova L, Gerber JS. Impact of antibiotics on necrotizing enterocolitis and antibiotic-associated diarrhea. Gastroenterol Clin North Am 2017; 46:61–76 [View Article] [PubMed]
    [Google Scholar]
  47. Schwiertz A, Gruhl B, Löbnitz M, Michel P, Radke M et al. Development of the intestinal bacterial composition in hospitalized preterm infants in comparison with breast-fed, full-term infants. Pediatr Res 2003; 54:393–399 [View Article] [PubMed]
    [Google Scholar]
  48. Wang S, Yu J, Suvira M, Setlow P, Li Y. Uptake of and resistance to the antibiotic berberine by individual dormant, germinating and outgrowing Bacillus spores as monitored by laser tweezers raman spectroscopy. PLoS One 2015; 10:e0144183 [View Article] [PubMed]
    [Google Scholar]
  49. Wauters L, Slaets H, De Paepe K, Ceulemans M, Wetzels S et al. Efficacy and safety of spore-forming probiotics in the treatment of functional dyspepsia: a pilot randomised, double-blind, placebo-controlled trial. Lancet Gastroenterol Hepatol 2021; 6:784–792 [View Article] [PubMed]
    [Google Scholar]
  50. Hong HA, Huang JM, Khaneja R, Hiep LV, Urdaci MC et al. The safety of Bacillus subtilis and Bacillus indicus as food probiotics. J Appl Microbiol 2008; 105:510–520 [View Article] [PubMed]
    [Google Scholar]
  51. Duc LH, Hong HA, Barbosa TM, Henriques AO, Cutting SM. Characterization of Bacillus probiotics available for human use. Appl Environ Microbiol 2004; 70:2161–2171 [View Article] [PubMed]
    [Google Scholar]
  52. Tavares Batista M, Souza RD, Paccez JD, Luiz WB, Ferreira EL et al. Gut adhesive Bacillus subtilis spores as a platform for mucosal delivery of antigens. Infect Immun 2014; 82:1414–1423 [View Article] [PubMed]
    [Google Scholar]
  53. Angioi A, Zanetti S, Sanna A, Delogu G, Fadda G. Adhesiveness of Bacillus subtilis Strains to Epithelial Cells Cultured in vitro. Microb Ecol Health Dis 1995; 8:71–77 [View Article]
    [Google Scholar]
  54. Alebouyeh M, Behzadian-Nejad Q, Soleimani M, Hassan ZM, Salmanian AH et al. Characterization of the interaction of undomesticated Bacillus subtilis spores with Caco-2 cell line. Ann Microbiol 2009; 59:273–277 [View Article]
    [Google Scholar]
  55. Kaneda T. Fatty acids of the genus Bacillus: an example of branched-chain preference. Bacteriol Rev 1977; 41:391–418 [View Article] [PubMed]
    [Google Scholar]
  56. Diomandé SE, Nguyen-The C, Guinebretière M-H, Broussolle V, Brillard J. Role of fatty acids in Bacillus environmental adaptation. Front Microbiol 2015; 6:813 [View Article] [PubMed]
    [Google Scholar]
  57. Ríos-Covián D, Ruas-Madiedo P, Margolles A, Gueimonde M, de Los Reyes-Gavilán CG et al. Intestinal short chain fatty acids and their link with diet and human health. Front Microbiol 2016; 7:185 [View Article] [PubMed]
    [Google Scholar]
  58. Hong HA, Khaneja R, Tam NMK, Cazzato A, Tan S et al. Bacillus subtilis isolated from the human gastrointestinal tract. Res Microbiol 2009; 160:134–143 [View Article] [PubMed]
    [Google Scholar]
  59. Higgins D, Dworkin J. Recent progress in Bacillus subtilis sporulation. FEMS Microbiol Rev 2012; 36:131–148 [View Article] [PubMed]
    [Google Scholar]
  60. Galbraith H, Miller TB. Physicochemical effects of long chain fatty acids on bacterial cells and their protoplasts. J Appl Bacteriol 1973; 36:647–658 [View Article] [PubMed]
    [Google Scholar]
  61. Rice KC, Bayles KW. Molecular control of bacterial death and lysis. Microbiol Mol Biol Rev 2008; 72:85–109 [View Article] [PubMed]
    [Google Scholar]
  62. Casula G, Cutting SM. Bacillus probiotics: spore germination in the gastrointestinal tract. Appl Environ Microbiol 2002; 68:2344–2352 [View Article] [PubMed]
    [Google Scholar]
  63. Frewer L, Lassen J, Kettlitz B, Scholderer J, Beekman V et al. Societal aspects of genetically modified foods. Food Chem Toxicol 2004; 42:1181–1193 [View Article] [PubMed]
    [Google Scholar]
  64. Kamaldeen S, Powell DA. Public perceptions of biotechnology. In Food Safety Network Technical Report vol 17 2000 pp 1–6
    [Google Scholar]
  65. Husby J. Definitions of GMO/LMO and modern biotechnology. Biosafety first Trondheim: Tapir Academic Publishers; 2007
    [Google Scholar]
  66. Kanchiswamy CN, Malnoy M, Velasco R, Kim JS, Viola R. Non-GMO genetically edited crop plants. Trends Biotechnol 2015; 33:489–491 [View Article] [PubMed]
    [Google Scholar]
  67. Gramelsberger G. Continuous culture techniques as simulators for standard cells: Jacques Monod’s, Aron Novick’s and Leo Szilard’s quantitative approach to microbiology. Hist Philos Life Sci 2018; 40:1–23 [View Article] [PubMed]
    [Google Scholar]
  68. James TW. Continuous culture of microorganisms. Annu Rev Microbiol 1961; 15:27–46 [View Article]
    [Google Scholar]
  69. Esteller M. Epigenetics in evolution and disease. The Lancet 2008; 372:S90–S96 [View Article]
    [Google Scholar]
  70. Berger SL, Kouzarides T, Shiekhattar R, Shilatifard A. An operational definition of epigenetics. Genes Dev 2009; 23:781–783 [View Article] [PubMed]
    [Google Scholar]
  71. Veening JW, Murray H, Errington J. A mechanism for cell cycle regulation of sporulation initiation in Bacillus subtilis. Genes Dev 2009; 23:1959–1970 [View Article] [PubMed]
    [Google Scholar]
  72. Walsh C, Wright G. Introduction: antibiotic resistance. Chem Rev 2005; 105:391–394 [View Article] [PubMed]
    [Google Scholar]
  73. Papadopoulos D, Schneider D, Meier-Eiss J, Arber W, Lenski RE et al. Genomic evolution during a 10,000-generation experiment with bacteria. Proc Natl Acad Sci U S A 1999; 96:3807–3812 [View Article] [PubMed]
    [Google Scholar]
  74. Han TH, Lee JH, Cho MH, Wood TK, Lee J. Environmental factors affecting indole production in Escherichia coli. Res Microbiol 2011; 162:108–116 [View Article] [PubMed]
    [Google Scholar]
  75. Takahashi K. Influence of bacteria on epigenetic gene control. Cell Mol Life Sci 2014; 71:1045–1054 [View Article] [PubMed]
    [Google Scholar]
  76. Katju V, Bergthorsson U. Old trade, new tricks: insights into the spontaneous mutation process from the partnering of classical mutation accumulation experiments with high-throughput genomic approaches. Genome Biol Evol 2019; 11:136–165 [View Article] [PubMed]
    [Google Scholar]
  77. Heilbron K, Toll-Riera M, Kojadinovic M, MacLean RC. Fitness is strongly influenced by rare mutations of large effect in a microbial mutation accumulation experiment. Genetics 2014; 197:981–990 [View Article] [PubMed]
    [Google Scholar]
  78. Halligan DL, Keightley PD. Spontaneous mutation accumulation studies in evolutionary genetics. Annu Rev Ecol Evol Syst 2009; 40:151–172 [View Article]
    [Google Scholar]
  79. Mohammad B. HN, Pezeshki P. Bacterial mutation; types, mechanisms and mutant detection methods: a review. Eur Sci J 2013
    [Google Scholar]
  80. Roth JR, Benson N, Galitski T, Haack K, Lawrence JG et al. Rearrangements of the bacterial chromosome: formation and applications. Escherichia coli and Salmonella: Cell Mol Biol 1996; 2:2256–2276
    [Google Scholar]
  81. Sun S, Ke R, Hughes D, Nilsson M, Andersson DI et al. Genome-wide detection of spontaneous chromosomal rearrangements in bacteria. PLoS One 2012; 7:e42639 [View Article]
    [Google Scholar]
  82. Cao S, Brandis G, Huseby DL, Hughes D. Positive selection during niche adaptation results in large-scale and irreversible rearrangement of chromosomal gene order in bacteria. Mol Biol Evol 2022; 39:msac069 [View Article] [PubMed]
    [Google Scholar]
  83. Watford S, Steven J W. Bacterial DNA mutations. In StatPearls 2021
    [Google Scholar]
  84. Maughan H, Masel J, Birky CW Jr, Nicholson WL. The roles of mutation accumulation and selection in loss of sporulation in experimental populations of Bacillus subtilis. Genetics 2007; 177:937–948 [View Article] [PubMed]
    [Google Scholar]
  85. Barreto HC, Cordeiro TN, Henriques AO, Gordo I. Rampant loss of social traits during domestication of a Bacillus subtilis natural isolate. Sci Rep 2020; 10:1–16 [View Article]
    [Google Scholar]
  86. Maughan H, Callicotte V, Hancock A, Birky CW Jr, Nicholson WL et al. The population genetics of phenotypic deterioration in experimental populations of Bacillus subtilis. Evolution 2006; 60:686–695 [View Article] [PubMed]
    [Google Scholar]
  87. Siranosian KJ, Grossman AD. Activation of spo0A transcription by sigma H is necessary for sporulation but not for competence in Bacillus subtilis. J Bacteriol 1994; 176:3812–3815 [View Article] [PubMed]
    [Google Scholar]
  88. Siebring J, Elema MJH, Drubi Vega F, Kovács AT, Haccou P et al. Repeated triggering of sporulation in Bacillus subtilis selects against a protein that affects the timing of cell division. ISME J 2014; 8:77–87 [View Article] [PubMed]
    [Google Scholar]
  89. Grossman AD. Genetic networks controlling the initiation of sporulation and the development of genetic competence in Bacillus subtilis. Annu Rev Genet 1995; 29:477–508 [View Article] [PubMed]
    [Google Scholar]
  90. Carvalho ALU de, Oliveira FHPC de, Mariano R de LR, Gouveia ER, Souto-Maior AM. Growth, sporulation and production of bioactive compounds by Bacillus subtilis R14. Braz arch biol technol 2010; 53:643–652 [View Article]
    [Google Scholar]
  91. Cosby WM, Zuber P. Regulation of Bacillus subtilis sigmaH (spo0H) and AbrB in response to changes in external pH. J Bacteriol 1997; 179:6778–6787 [View Article] [PubMed]
    [Google Scholar]
  92. Gauvry E, Mathot AG, Couvert O, Leguérinel I, Coroller L. Effects of temperature, pH and water activity on the growth and the sporulation abilities of Bacillus subtilis BSB1. Int J Food Microbiol 2021; 337:108915 [View Article] [PubMed]
    [Google Scholar]
  93. Dempfle L. Conservation, creation, and utilization of genetic variation. JDS 1990; 73:2593–2600 [View Article]
    [Google Scholar]
  94. Koskiniemi S, Sun S, Berg OG, Andersson DI. Selection-driven gene loss in bacteria. PLoS Genet 2012; 8:e1002787 [View Article] [PubMed]
    [Google Scholar]
  95. Leitch J, Collier PJ. A new chemically-defined medium for Bacillus subtilis (168) NCIMB 12900. Lett Appl Microbiol 1996; 22:18–20 [View Article]
    [Google Scholar]
  96. Wood DE, Salzberg SL. Kraken: ultrafast metagenomic sequence classification using exact alignments. Genome Biol 2014; 15:1–12 [View Article]
    [Google Scholar]
  97. Ondov BD, Bergman NH, Phillippy AM. Interactive metagenomic visualization in a Web browser. BMC Bioinformatics 2011; 12:1–10 [View Article]
    [Google Scholar]
  98. Dervaux J, Magniez JC, Libchaber A. On growth and form of Bacillus subtilis biofilms. Interface Focus 2014; 4:20130051 [View Article] [PubMed]
    [Google Scholar]
  99. Manabe K, Kageyama Y, Tohata M, Ara K, Ozaki K et al. High external pH enables more efficient secretion of alkaline α-amylase AmyK38 by Bacillus subtilis. Microb Cell Fact 2012; 11:1–3 [View Article]
    [Google Scholar]
  100. Romero S, Merino E, Bolívar F, Gosset G, Martinez A. Metabolic engineering of Bacillus subtilis for ethanol production: lactate dehydrogenase plays a key role in fermentative metabolism. Appl Environ Microbiol 2007; 73:5190–5198 [View Article] [PubMed]
    [Google Scholar]
  101. Arnaouteli S, Bamford NC, Stanley-Wall NR, Kovács ÁT. Bacillus subtilis biofilm formation and social interactions. Nat Rev Microbiol 2021; 19:600–614 [View Article] [PubMed]
    [Google Scholar]
  102. López D, Vlamakis H, Kolter R. Biofilms. Cold Spring Harb Perspect Biol 2010; 2:a000398 [View Article] [PubMed]
    [Google Scholar]
  103. Hu P, Suh J, Yoshikawa A, Zhu J. The effects of glucose concentration on the growth rates of wild type and PDC1 mutant Saccharomyces cerevisiae. The Expedition 2014; 4:
    [Google Scholar]
  104. Duanis-Assaf D, Steinberg D, Shemesh M. Efficiency of Bacillus subtilis metabolism of sugar alcohols governs its probiotic effect against cariogenic Streptococcus mutans. Artif Cells Nanomed Biotechnol 2020; 48:1222–1230 [View Article] [PubMed]
    [Google Scholar]
  105. Powers EM. Method for obtaining free bacterial spores of Bacillus subtilis var. niger. Appl Microbiol 1968; 16:180–181 [View Article] [PubMed]
    [Google Scholar]
  106. Phillips ZEV, Strauch MA. Bacillus subtilis sporulation and stationary phase gene expression. Cell Mol Life Sci 2002; 59:392–402 [View Article] [PubMed]
    [Google Scholar]
  107. Saujet L, Monot M, Dupuy B, Soutourina O, Martin-Verstraete I. The key sigma factor of transition phase, SigH, controls sporulation, metabolism, and virulence factor expression in Clostridium difficile. J Bacteriol 2011; 193:3186–3196 [View Article] [PubMed]
    [Google Scholar]
  108. Ferraris RP, Yasharpour SA, Lloyd KC, Mirzayan RA, Diamond JM. Luminal glucose concentrations in the gut under normal conditions. Am J Physiol 1990; 259:G822–37 [View Article] [PubMed]
    [Google Scholar]
  109. Stümpel F, Burcelin R, Jungermann K, Thorens B. Normal kinetics of intestinal glucose absorption in the absence of GLUT2: evidence for a transport pathway requiring glucose phosphorylation and transfer into the endoplasmic reticulum. Proc Natl Acad Sci U S A 2001; 98:11330–11335 [View Article] [PubMed]
    [Google Scholar]
  110. Knopp JL, Signal M, Harris DL, Marics G, Weston P et al. Modelling intestinal glucose absorption in premature infants using continuous glucose monitoring data. Comput Methods Programs Biomed 2019; 171:41–51 [View Article] [PubMed]
    [Google Scholar]
  111. Fallingborg J. Intraluminal pH of the human gastrointestinal tract. Dan Med Bull 1999; 46:183–196 [PubMed]
    [Google Scholar]
  112. Evans DF, Pye G, Bramley R, Clark AG, Dyson TJ et al. Measurement of gastrointestinal pH profiles in normal ambulant human subjects. Gut 1988; 29:1035–1041 [View Article] [PubMed]
    [Google Scholar]
  113. Chen L, Tuo B, Dong H. Regulation of intestinal glucose absorption by ion channels and transporters. Nutrients 2016; 8:43 [View Article] [PubMed]
    [Google Scholar]
  114. Pácha J. Development of intestinal transport function in mammals. Physiol Rev 2000; 80:1633–1667 [View Article] [PubMed]
    [Google Scholar]
  115. Cavalli C, Teng C, Battaglia FC, Bevilacqua G. Free sugar and sugar alcohol concentrations in human breast milk. J Pediatr Gastroenterol Nutr 2006; 42:215–221 [View Article] [PubMed]
    [Google Scholar]
  116. Scano P, Murgia A, Demuru M, Consonni R, Caboni P. Metabolite profiles of formula milk compared to breast milk. Food Res Int 2016; 87:76–82 [View Article] [PubMed]
    [Google Scholar]
  117. Hove H, Nørgaard H, Mortensen PB. Lactic acid bacteria and the human gastrointestinal tract. Eur J Clin Nutr 1999; 53:339–350 [View Article] [PubMed]
    [Google Scholar]
  118. Rider AK, Schedl HP, Nokes G, Shining S. Small intestinal glucose transport. Proximal-distal kinetic gradients. J Gen Physiol 1967; 50:1173–1182 [View Article] [PubMed]
    [Google Scholar]
  119. Dound YA, Jadhav SS, Devale M, Bayne T, Krishnan K et al. The effect of probiotic Bacillus subtilis HU58 on immune function in healthy human. Indian Pract 2017; 70:15–20
    [Google Scholar]
  120. Cook AM, Lund BM. Total counts of bacterial spores using counting slides. J Gen Microbiol 1962; 29:97–104 [View Article] [PubMed]
    [Google Scholar]
  121. Morris S, Nicholls J. An evaluation of optical density to estimate fungal spore concentrations in water suspensions. Strain 1978; 1:1240–1242
    [Google Scholar]
  122. Galperin MY, Mekhedov SL, Puigbo P, Smirnov S, Wolf YI et al. Genomic determinants of sporulation in Bacilli and Clostridia: towards the minimal set of sporulation-specific genes. Environ Microbiol 2012; 14:2870–2890 [View Article] [PubMed]
    [Google Scholar]
  123. Das G, Prasad MP. Isolation, purification & mass production of protease enzyme from Bacillus subtilis. Int Res J Microbiol 2010; 1:26–31
    [Google Scholar]
  124. Gromova LV, Fetissov SO, Gruzdkov AA. Mechanisms of glucose absorption in the small intestine in health and metabolic diseases and their role in appetite regulation. Nutrients 2021; 13:2474 [View Article] [PubMed]
    [Google Scholar]
  125. Sonenshein AL, Hoch JA, Losick R. Bacillus Subtilis and Its Closest Relatives Washington, DC, USA: 2001 pp 519–525 [View Article]
    [Google Scholar]
  126. Onyenwoke RU, Brill JA, Farahi K, Wiegel J. Sporulation genes in members of the low G+C Gram-type-positive phylogenetic branch (Firmicutes). Arch Microbiol 2004; 182:182–192 [View Article] [PubMed]
    [Google Scholar]
  127. Traag BA, Pugliese A, Eisen JA, Losick R. Gene conservation among endospore-forming bacteria reveals additional sporulation genes in Bacillus subtilis. J Bacteriol 2013; 195:253–260 [View Article] [PubMed]
    [Google Scholar]
  128. Govin J, Berger SL. Genome reprogramming during sporulation. Int J Dev Biol 2009; 53:425–432 [View Article] [PubMed]
    [Google Scholar]
  129. Keijser BJF, Ter Beek A, Rauwerda H, Schuren F, Montijn R et al. Analysis of temporal gene expression during Bacillus subtilis spore germination and outgrowth. J Bacteriol 2007; 189:3624–3634 [View Article] [PubMed]
    [Google Scholar]
  130. Mutlu A, Kaspar C, Becker N, Bischofs IB. A spore quality-quantity tradeoff favors diverse sporulation strategies in Bacillus subtilis. ISME J 2020; 14:2703–2714 [View Article] [PubMed]
    [Google Scholar]
  131. González-Pastor JE. Cannibalism: a social behavior in sporulating Bacillus subtilis. FEMS Microbiol Rev 2011; 35:415–424 [View Article] [PubMed]
    [Google Scholar]
  132. Lamsa A, Liu WT, Dorrestein PC, Pogliano K. The Bacillus subtilis cannibalism toxin SDP collapses the proton motive force and induces autolysis. Mol Microbiol 2012; 84:486–500 [View Article] [PubMed]
    [Google Scholar]
  133. Höfler C, Heckmann J, Fritsch A, Popp P, Gebhard S et al. Cannibalism stress response in Bacillus subtilis. Microbiology 2016; 162:164–176 [View Article] [PubMed]
    [Google Scholar]
  134. Pérez Morales TG, Ho TD, Liu WT, Dorrestein PC, Ellermeier CD. Production of the cannibalism toxin SDP is a multistep process that requires SdpA and SdpB. J Bacteriol 2013; 195:3244–3251 [View Article] [PubMed]
    [Google Scholar]
  135. Figaro S, Durand S, Gilet L, Cayet N, Sachse M et al. Bacillus subtilis mutants with knockouts of the genes encoding ribonucleases RNase Y and RNase J1 are viable, with major defects in cell morphology, sporulation, and competence. J Bacteriol 2013; 195:2340–2348 [View Article] [PubMed]
    [Google Scholar]
  136. Wang M, Yu H, Li X, Shen Z. Single-gene regulated non-spore-forming Bacillus subtilis: Construction, transcriptome responses, and applications for producing enzymes and surfactin. Metab Eng 2020; 62:235–248 [View Article] [PubMed]
    [Google Scholar]
  137. Molle V, Fujita M, Jensen ST, Eichenberger P, González-Pastor JE et al. The Spo0A regulon of Bacillus subtilis. Mol Microbiol 2003; 50:1683–1701 [View Article] [PubMed]
    [Google Scholar]
  138. Fujita M, Losick R. The master regulator for entry into sporulation in Bacillus subtilis becomes a cell-specific transcription factor after asymmetric division. Genes Dev 2003; 17:1166–1174 [View Article] [PubMed]
    [Google Scholar]
  139. Shafikhani SH, Leighton T. AbrB and Spo0E control the proper timing of sporulation in Bacillus subtilis. Curr Microbiol 2004; 48:262–269 [View Article] [PubMed]
    [Google Scholar]
  140. Zuber PE, Losick RI. Role of AbrB in Spo0A- and Spo0B-dependent utilization of a sporulation promoter in Bacillus subtilis. J Bacteriol 1987; 169:2223–2230 [View Article] [PubMed]
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journal/acmi/10.1099/acmi.0.000419
Loading
/content/journal/acmi/10.1099/acmi.0.000419
Loading

Data & Media loading...

Supplements

Supplementary material 1

PDF
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error