eISSN: 1644-4124
ISSN: 1426-3912
Central European Journal of Immunology
Current issue Archive Manuscripts accepted About the journal Special Issues Editorial board Abstracting and indexing Subscription Contact Instructions for authors Ethical standards and procedures
Editorial System
Submit your Manuscript
SCImago Journal & Country Rank
1/2011
vol. 36
 
Share:
Share:

Review paper
Biological functions of exopolysaccharides from probiotic bacteria

Marta Ciszek-Lenda

(Centr Eur J Immunol 2011; 36 (1): 51-55)
Online publish date: 2011/03/31
Article file
- Biological functions.pdf  [0.07 MB]
Get citation
 
 

Introduction

Probiotic bacteria have the ability to produce exocellular polymers called exopolysaccharides (EPS). It has been suggested that the health benefit of probiotic bacteria can be attributed to the production of EPS. However, the composition, structure and biological functions of EPS may vary depending on the type of microorganism and environmental conditions. Some data suggest that EPS production is under control of quorum sensing (QS) through regulation of gene expression for proteins involved in EPS biosynthesis [1].

Eps is the main substance involved in biofilm formation and may achieve 50-90% of the total organic substances such as proteins, lipids and nucleic acids [1]. Bacteria develop biofilms to protect the microbial community against environmental stress. It has been established that both pathogenic, as well as commensal bacteria, generate biofilms in human mucosas. Biofilm formation is associated with bacterial infection but it may also play a protective role. For example, biofilm-like communities of the gastrointestinal and female urogenital tracts contain beneficial lactic acid bacteria. It has been shown that the cell wall components of probiotic bacteria, such as peptidoglycans or teichoic acids play an important role in activation of immune cells. By contrast, the role of EPS in modulation of the immune system is still unclear.

Probiotic bacteria

Probiotics are live microorganisms which exert a beneficial effects on the host. The Food and Agriculture Organization of the United Nations defines them as “live microorganisms which when administered in adequate amounts confer a health benefit on the host” [2, 3]. Members of genera Lactobacillus and Bifidobacterium are the most common probiotics used not only for human consumption [4] but also in pharmaceutical preparations or in biomedicine [5]. One of the main criteria for selection of oral administration of probiotics is their ability to adhere to the intestinal mucosa allowing a transitory colonization of the gastrointestinal tract [6]. Probiotics maintain the balance within such complex ecosystem as human intestine in many ways: inhibition of the proliferation of pathogens by competition between bacteria and pathogens for adhesion, suppression of production of virulent factors by pathogens secreting bacteriocins, or modulation of the host immune system via interaction between probiotic bacteria and intestinal epithelial cells [7-9]. However, the effectiveness of probiotics is strain-specific and each strain may affect the host health trough different mechanisms [10-13]. For instance, lactic acid bacteria (LAB) diminish symptoms of lactose intolerance, reduce serum cholesterol, prevent diarrhea, enhance immune responses and anti­carcinogenic activities, alleviate allergies [14-17]. Lactic acid bacteria can even prevent or inhibit growth of pathogenic bacteria [18]. All these effects depend on adhesion of bacteria and their survival in specific regions of the gastrointestinal tract, competition with pathogens, presence of harmful antigens in the environment and mucosal barrier function.

The idea of using LAB to cure diseases and to promote health has been existing for at least 100 years, but the mechanisms of these actions are still poorly understood. Nevertheless, recent studies show that the probiotic effects of LAB result not only from whole microorganisms and cell wall components but also from peptides, nucleic acids and extracellular polysaccharides produced during growth of these bacteria [19-21]. It is postulated that this effect is related to diversity of exopolysaccharide polymers produced by bacteria [22].

Structure of lactic acid bacteria derived exopolysaccharides

Almost all LAB have ability to secrete polysaccharide polymers, which are called exopolysaccharides (EPS). Lactobacillus, Streptococcus and Lactococcus prevail mostly among these genera [21-23]. Some strains of Bifidobacterium are also able to produce EPS [24]. EPS is secreted in two forms: as a capsular exopolysaccharide which is associated with the cell surface or as slime exopolysaccharide secreted as free polymers to the environment [25, 26]. Chemical structure of EPS has been studied in details [27-30]. There are over 50 different EPSes derived from LAB described and they are mostly composed of repeated units of a certain number of diverse sugar residues or sugar derivatives [28, 30]. Two main groups of EPSes are described: homopolysaccharides and hete­ropolysaccharides (Fig. 1). Structure of EPS depends on chemical composition. Homopolysaccharides contain a single type of monosaccharide and it is usually glucose or fructose. Thus, they are called -glucans (dextrans) or fructans. Repeated units consist often of three to eight monosaccharides. EPS of Streptococcus thermophilus was the first heteropolysaccharide studied and described in details [31]. Heteropolysaccharides are composed of at least two different sugars out of glucose, galactose and rhamnose at different ratios. The molecular mass of these polymers ranges between 4.0 × 104 and 6.0 × 106 Da [32]. The composition and nature of EPS is affected by environmental conditions, biosynthetic pathways or rate of microbial growth. Residues such as sn-glycerol-3-phosphate, N-acetyl-aminosugars, phosphate and acetyl groups can also be found [30, 33]. This and the molecular weight determines the functional properties of EPS [22, 34]. However, not only LAB can produce EPS; some pathogenic bacteria can synthesize these molecules as well [35, 36].

Biosynthesis and genes of exopolysaccharides

All homopolysaccharides are synthesized by extra­cellular specific enzyme – glycosyl transferase and energy for this synthesis comes from hydrolysis of sucrose [33]. Heteropolysaccharides are polymers of sugar precursors in the cytoplasm and several enzymes or proteins are involved in their synthesis and secretion [37]. The genes for these enzymes and proteins have been revealed in several strains of LAB. Genes for EPS synthesis in Lactococcus lactis and Lactobacillus casei are located in the plasmids [38] in contrast to all thermophilic LAB, genes of which are located in a bacterial chromosome [33]. There have been described sequences of genes for Streptococcus thermophilus Sfi6 [39, 40], S. thermophilus NCFB 2393 [41] and S. thermophilus MR-1C [42]. Organization of these genes appears to be highly conserved [33]. There is no gene cluster found in Lactobacillus delbrueckii ssp. bulgaricus so far despite its importance for the production of fermented products such as yogurts [43].

Biofilm formation – the role of exopolysaccharides

There is an increasing interest among researchers concerning EPS, but the physiological role of these molecules is still not clear [22, 44, 45]. Most of this research relate to biofilm formation and its role in bacterial ecology [46, 47]. The term ‘biofilm’ was used for the first time in 1978 by Costerton et al. [48]. Studies on the role of EPS in biofilm formation are generally focused on pathogenic bacteria which are mostly Gram-negative species [49, 50]. Less is known about EPS in Gram-positive species. EPS fills intracellular space between bacteria and together with proteins, nucleic acids and lipids composes the structure of the biofilm matrix. EPS in biofilm protects bacterial cells from desiccation, phage attack, antimicrobial compounds, osmotic stress and predatory attack from protozoa [46, 51-53]. It helps bacteria to survive in detrimental conditions such as too low or too high temperature or pH. Capsular polysaccharides can promote the adherence of bacteria to biological surfaces, thereby facilitating the colonization of various ecological niches [25, 30]. EPS also can enable probiotics to survive in gastric acid and bile salts [19]. Biofilm produced by pathogenic bacteria makes them less susceptible to antibiotics and attacks by innate host defense. It plays an important role in many chronic bacterial infections [54, 55]. Biofilm formation and EPS production is under control of regulatory pathway of QS. It has been suggested that QS allows bacteria to communicate and regulate the expression of genes which are required for synthesis of EPS in response to changes in bacteria density [1].

LAB derived exopolysaccharides and the immune system

Immunomodulating mechanism of LAB is obscure. However, it has been shown that the cell wall components of these bacteria, such as peptidoglycan or teichoic acids may play an important role in activating immune system cells in the gut [56]. An extract (without peptidoglycan) of cell walls from Lactobacillus rhamnosus KLC37 containing EPS was tested in vitro in our laboratory for immunomodulating capacity. It was compared with lipopolysaccharide (LPS) from Escherichia coli. It turned out that it stimulated production of proinflammatory cytokines by mice macrophages in a dose-dependent manner and this stimulation depended on p38 and ERK kinase activity. However, participation of this extract in immunological response of macrophages was slightly comparable to that of whole bacteria. Only LPS, but not the extract, could induce hyporesponsiveness to a subsequent stimulation with LPS. Interestingly, extract-primed macrophages increased their ability to bind LPS in studies with atomic force microscopy [our data, unpublished].

The health benefit of LAB have been attributed to the production of EPS [25]. LAB EPSes have been claimed to have immunostimulatory activity [57, 58], antitumor effects [59, 60] or blood pressure and cholesterol lowering activity [61, 62]. EPS reduces symptoms of lactose intolerance and prevents diarrhea [14]. There have been reports that sugar polymers have antimicrobial properties and help to heal wounds [63, 64]. It has been also shown that some EPSes induce cytokine production, act like lymphocytes B mitogens or change functions of splenocytes [65-67]. EPS can reduce the symptoms of collagen-induced arthritis or diminish arteriosclerosis in mice (our research, unpublished). Orally administrated EPS-producing LAB attenuate severity of colitis and may be a promising agent in therapy of inflam­matory bowel disease [7, 68].

In our opinion, such wide diversity of EPS effects on the immune system results not only from strain specificity, but also from microenvironmental impact on the EPS metabolism of probiotic bacteria. However, it is still not clear whether EPS can be the ligand for pattern recognition receptors and how the immune system can differentiate pathogenic bacteria from commensal flora. It is possible that EPS plays a role of signaling molecule in the mucosal immune system.

Conclusions

EPS is produced by many probiotic bacteria and it is a key molecule of the biofilm matrix. However, due to the extreme heterogeneity of EPS, strain specificity and unpredictable enzymatic modifications, its immuno­modulatory potential should be established individually for each isolated molecule separately. Moreover, the role of EPS in QS regulation remains to be explained.

Acknowledgements

I am very grateful to Prof. Janusz Marcinkiewicz for critical reading the manuscript and valuable advices. This work was supported by the grants of Jagiellonian University College of Medicine: No. K/ZDS/000684 and K/PBW/000559.

References

 1. Vu B, Chen M, Crawford RJ, Ivanova EP (2009): Bacterial extracellular polysaccharides involved in biofilm formation. Molecules 14: 2535-2554.  

2. Salminen S, von Wright A, Morelli L, et al. (1998): Demonstration of safety of probiotics – a review. Int J Food Microbiol 44: 93-106.  

3. Fuller R (1989): Probiotics in man and animals. J Appl Bacteriol 66: 365-378.  

4. Crittenden RG, Martinez NR, Playne MJ (2003): Synthesis and utilisation of folate by yoghurt starter cultures and probiotic bacteria. Int J Food Microbiol 80: 217-222.  

5. Simmering R, Breves R (2009): Pre- and probiotic cosmetics. Hautarzt 60: 809-814.  

6. Dunne C (2001): Adaptation of bacteria to the intestinal niche: probiotics and gut disorder. Inflamm Bowel Dis 7: 136-145.  

7. Sengül N, IºIk S, AslIm B, et al. (2010): The effect of exopolysaccharide-producing probiotic strains on gut oxidativ damage in experimental colitis. Dig Dis Sci [Epub ahead of print].  

8. Servin AL, Coconnier MH (2003): Adhesion of probiotic strains to the intestinal mucosa and interaction with pathogens. Best Pract Res Clin Gastroenterol 17: 741-754.  

9. Gill HS (2003): Probiotics to enhance anti-infective defenses in the gastrointestinal tract. Best Pract Res Clin Gastroenterol 17: 755-773.

10. Marcinkiewicz J, Ciszek M, Bobek M, et al. (2007): Differential inflammatory mediator response in vitro from murine macrophages to lactobacilli and pathogenic intestinal bacteria. Int J Exp Pathol 88: 155-164.

11. Matsumoto S, Hara T, Hori T, et al. (2005): Probiotic Lactobacillus-induced improvement in murine chronic inflammatory bowel disease is associated with the down-regulation of pro-inflammatory cytokines in lamina propria mononuclear cells. Clin Exp Immunol 140: 417-426.

12. Cross ML, Ganner A, Teilab D, et al. (2004): Patterns of cytokine induction by gram-positive and gram-negative probiotic bacteria. FEMS Immunol Med Microbiol 42: 173-180.

13. Christensen HR, Fro/kiaer H, Pestka JJ (2002): Lactobacilli differentially modulate expression of cytokines and maturation surface markers in murine dendritic cells. J Immunol 168: 171-178.

14. Grandy G, Medina M, Soria R, et al. (2010): Probiotics in the treatment of acute rotavirus diarrhoea. A randomized, double-blind, controlled trial using two different probiotic preparations in Bolivian children. BMC Infect Dis 10: 253.

15. Lee J, Kim Y, Yun HS, et al. (2010): Genetic and proteomic analysis of factors affecting serum cholesterol reduction by Lactobacillus acidophilus A4. Appl Environ Microbiol 76: 4829-4835.

16. Burns AJ, Rowland IR (2000): Anti-carcinogenicity of probiotics and prebiotics. Curr Issues Intest Microbiol 1: 13-24.

17. Savilahti E, Kuitunen M, Vaarala O (2008): Pre and probiotics in the prevention and treatment of food allergy. Curr Opin Allergy Clin Immunol 8: 243-248.

18. Imase K, Tanaka A, Tokunaga K, et al. (2007): Lactobacillus reuteri tablets suppress Helicobacter pylori infection – a double-blind randomised placebo-controlled cross-over clinical study. Kansenshogaku Zasshi 81: 387-393.

19. Sengül N, Aslím B, Uçar G, et al. (2006): Effects of exopolysaccharide-producing probiotic strains on experimental colitis in rats. Dis Colon Rectum 49: 250-258.

20. Shanahan F (2002): Probiotics and inflammatory bowel disease: from fads and fantasy to facts and future. Br J Nutr 88: 5-9.

21. Nishimura-Uemura J, Kitazawa H, Kawai Y, et al. (2003): Functional alteration of murine macrophages stimulated with extracellular polysaccharides from Lactobacillus delbrueckii ssp. bulgaricus OLL1073R-1. Food Microbiol 20: 267-273.

22. Makino S, Ikegami S, Kano H, et al. (2006): Immuno­modulatory effects of polysaccharides produced by Lactobacillus delbrueckii ssp. bulgaricus OLL1073R-1. J Diary Sci 89: 2873-2881.

23. Ruas-Madiedo P, de los Reyes-Gavilán CG (2005): Invited review: methods for the screening, isolation, and characterization of exopolysaccharides produced by lactic acid bacteria. J Dairy Sci 88: 843-856.

24. Salazar N, Ruas-Madiedo P, Kolida S, et al. (2009): Exopoly­saccharides produced by Bifidobacterium longum IPLA E44 and Bifidobacterium animalis subsp. lactis IPLA R1 modify the composition and metabolic activity of human faecal microbiota in pH-controlled batch cultures. Int J Food Microbiol 135: 260-267.

25. Ruas-Madiedo P, Gueimonde M, Margolles A (2006): Exopolysaccharides produced by probiotic strains modify the adhesion of probiotics and enteropathogens to human intestinal mucus. Food Prot 69: 2011-2015.

26. Whitfield C (1998): Bacterial extracellular polysaccharides. Can J Microbiol 34: 415-420.

27. Robijn GW, Wienk HL, van den Berg DJ, et al. (1996): Structural studies of the exopolysaccharide produced by Lactobacillus paracasei 34-1. Carbohydr Res 285: 129-139.

28. Górska S, Jachymek W, Rybka J, et al. (2010): Structural and immunochemical studies of neutral exopolysaccharide produced by Lactobacillus johnsonii 142. Carbohydr Res 345: 108-114.

29. Lipiński T, Jones C, Lemercinier X, et al. (2003): Structural analysis of the Lactobacillus rhamnosus strain KL37C exopolysaccharide. Carbohydr Res 338: 605-609.

30. Landersjö C, Yang Z, Huttunen E, Widmalm G (2002): Structural studies of the exopolysaccharide produced by Lactobacillus rhamnosus strain GG (ATCC 53103). Biomacromolecules 3: 880-884.

31. Doco T, Wieruszeski JM, Fournet B, et al. (1990): Structure of an exocellular polysaccharide produced by Streptococcus thermophilus. Carbohydr Res 198: 313-321.

32. Welman AD, Maddox IS (2003): Exopolysaccharides from lactic acid bacteria: perspectives and challenges. Trends Biotechnol 21: 269-274.

33. De Vuyst L, Degeest B (1999): Heteropolysaccharides from lactic acid bacteria. FEMS Microbiol Rev 23: 153-177.

34. Ruas-Madiedo P, Hugenholtz J, Zoon P (2002): An overview of the functionality of exopolysaccharides produced by lactic acid bacteria. Int Dairy J 12: 163-171.

35. Watnick PI, Kolter R (1999): Steps in the development of a Vibrio cholerae El Tor biofilm. Mol Microbiol 34: 586-595.

36. Sutherland IW, Vu B, Chen M, et al. (2001): Microbial polysaccharides from Gram-negative bacteria involved in biofilm formation. Int Dairy J 11: 663-674.

37. De Vuyst L, De Vin F, Vaningelgem F, Degeest B (2001): Recent developments in the biosynthesis and applications of heteropolysaccharides from lactic acid bacteria. Int Diary J 11: 687-707.

38. van Kranenburg R, Marugg JD, van Swam II, et al. (1997): Molecular characterization of the plasmid-encoded eps gene cluster essential for exopolysaccharide biosynthesis in Lactococcus lactis. Mol Microbiol 24: 387-397.

39. Stingele F, Neeser JR, Mollet BJ (1996): Identification and characterization of the eps (Exopolysaccharide) gene cluster from Streptococcus thermophilus Sfi6. Bacteriol 178: 1680-1690.

40. Stingele F, Mollet B (1995): Homologous integration and transposition to identify genes involved in the production of exopolysaccharides in Streptococcus thermophilus. Dev Biol Stand 85: 487-493.

41. Griffin AM, Morris VJ, Gasson MJ (1996): The cpsABCDE genes involved in polysaccharide production in Streptococcus salivarius ssp. thermophilus strain NCBF 2393. Gene 183: 23-27.

42. Low D, Ahlgren JA, Horne D, et al. (1998): Role of Streptococcus thermophilus MR-1C capsular exopolysaccharide in cheese moisture retention. Appl Environ Microbiol 64: 2147-2151.

43. Lamothe GT, Jolly L, Mollet B, Stingele F (2002): Genetic and biochemical characterization of exopolysaccharide biosynthesis by Lactobacillus delbrueckii subsp. bulgaricus. Arch Microbiol 178: 218-228.

44. Badel S, Bernardi T, Michaud P (2011): New perspectives for Lactobacilli exopolysaccharides. Biotechnol Adv 29: 54-66.

45. Laws A, Gu Y, Marshall V (2001): Biosynthesis, characteriza­tion, and design of bacterial exopolysaccharides from lactic acid bacteria. Biotechnol Adv 19: 597-625.

46. Vuong C, Kocianova S, Voyich JM, et al. (2004): A crucial role for exopolysaccharide modification in bacterial biofilm formation, immune evasion, and virulence. J Biol Chem 279: 54881-54886.

47. Looijesteijn PJ, Trapet L, De Vries E, et al. (2001): Physiological function of exopolysaccharides produced by Lactococcus lactis. Int J Food Microbiol 64: 71-80.

48. Costerton JW, Geesey GG, Cheng KJ (1978): How bacteria stick. Sci Am 238: 86-95.

49. Watnick P, Kolter R (2000): Biofilm, City of Microbes. J Bacteriol 182: 2675-2679.

50. Byrd MS, Pang B, Mishra M, et al. (2010): The Pseudomonas aeruginosa exopolysaccharide psl facilitates surface adherence and NF-kappaB activation in A549 cells. MBio 1: e00140-10.

51. Herasimenka Y, Cescutti P, Impallomeni G, Rizzo R (2007): Exopolysaccharides produced by Inquilinus limosus, a new pathogen of cystic fibrosis patients: novel structures with usual components. Carbohydr Res 342: 2404-2415.

52. Forde A, Fitzgerald GF (1999): Bacteriophage defence systems in lactic acid bacteria. Antonie Van Leeuwenhoek 76: 89-113.

53. Kumon H (2000): Management of biofilm infections in the urinary tract. World J Surg 24: 1193-1196.

54. Ho/iby N, Bjarnsholt T, Givskov M, et al. (2010): Antibiotic resistance of bacterial biofilms. Int J Antimicrob Agents 35: 322-332.

55. Burmo/lle M, Thomsen TR, Fazli M, et al. (2010): Biofilms in chronic infections - a matter of opportunity - monospecies biofilms in multispecies infections. FEMS Immunol Med Microbiol 59: 324-336.

56. Vidal K, Donnet-Hughes A, Granato D (2002): Lipoteichoic acids from Lactobacillus johnsonii strain La1 and Lactobacillus acidophilus strain La10 antagonize the responsiveness of human intestinal epithelial HT29 cells to lipopolysaccharide and gram-negative bacteria. Infect Immun 70: 2057-2064.

57. Vinderola G, Perdigón G, Duarte J et al. (2006): Effects of the oral administration of the exopolysaccharide produced by Lactobacillus kefiranofaciens on the gut mucosal immunity Cytokine 36: 254-260.

58. Hosono A, Lee J, Ametani A, et al. (1997): Characterization of a water-soluble polysaccharide fraction with immunopotentiating activity from Bifidobacterium adolescentis M101-4. Biosci Biotechnol Biochem 61: 312-316.

59. Furukawa N, Matsuoka A, Takahashi T, Yamanaka Y (2000): Anti-metastic effect of kefir grain components on Lewis lung carcinoma and highly metastic B16 melanoma in mice. J Agric Sci Tokyo Nogyo Daigaku 45: 62-70.

60. Kitazawa H, Harata T, Uemura J, et al. (1998): Phosphate group requirement for mitogenic activation of lymphocytes by an extracellular phosphopolysaccharide from Lactobacillus delbrueckii ssp. bulgaricus. Int J Food Microbiol 40: 169-175.

61. Maeda H, Zhu X, Omura K, et al. (2004): Effects of an exopolysaccharide (kefiran) on lipids, blood pressure, blood glucose, and constipation. Biofactors 22: 197-200.

62. Nakajima H, Suzuki Y, Hirota T, et al. (1992): Cholesterol lowering activity of ropy fermented milk. J Food Sci 57: 327-1329.

63. Rodrigues KL, Caputo LR, Carvalho JC, et al. (2005): Antimicrobial and healing activity of kefir and kefiran extract. Int J Antimicrob Agents 25: 404-408.

64. Wu MH, Pan TM, Wu YJ (2010): Exopolysaccharide activities from probiotic bifidobacterium: Immunomodulatory effects (on J774A.1 macrophages) and antimicrobial properties. Int J Food Microbiol 144: 104-110.

65. Chabot S, Yu H-Lig, De Léséleu L (2001): Exopolysaccharides from Lactobacillus rhamnosus RW-9595M stimulate TNF, IL-6 and IL-12 in human and mouse cultured immuno­com­petent cells, and IFN- in mouse splenocytes. Lait 81: 683-697.

66. Kitazawa H, Yamaguchi T, Fujimoto Y, Itoh T (1993): Comparative activity of B-cell mitogen, a phosphopolysaccharide produced by Lactococcus lactis ssp. cremoris on various lymphocytes. Anim Sci Technol 64: 604-607.

67. Bleau C, Monges A, Rashidan K (2010): Intermediate chains of exopolysaccharides from Lactobacillus rhamnosus RW-9595M increase IL-10 production by macrophages J Appl Microbiol 108: 666-675.

68. Lebeer S, Ceuppens J, Geboes K, et al. (2007): Mechanisms of probiotic-host interaction with IBD as a case study: a role for exopolysaccharides? Commun Agric Appl Biol Sci 72: 41-45.
Copyright: © 2011 Polish Society of Experimental and Clinical Immunology This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International (CC BY-NC-SA 4.0) License (http://creativecommons.org/licenses/by-nc-sa/4.0/), allowing third parties to copy and redistribute the material in any medium or format and to remix, transform, and build upon the material, provided the original work is properly cited and states its license.

Quick links
© 2024 Termedia Sp. z o.o.
Developed by Bentus.