Effect of probiotic Lactobacillus rhamnosus by-products on gingival epithelial cells challenged with Porphyromonas gingivalis

https://doi.org/10.1016/j.archoralbio.2021.105174Get rights and content

Highlights

  • L. rhamnosus Lr-32 by-products rather than live probiotic alter the response driven by P. gingivalis in gingival epithelial cells.

  • Secreted products by the probiotic in the spent medium decreased cell response to P. gingivalis.

  • Cell lysate of probiotic increased transcription of inflammatory genes in P. gingivalis infected gingival epithelial cells.

Abstract

Objective

Probiotics are usually given as living cells, but their effects may be also achieved by postbiotics. We hypothesized that probiotics products (spent media and lysate) altered the response induced by P. gingivalis in gingival epithelial cells (GECS).

Methods

Immortalized human OBA-9 GECs (∼2,5 × 105cells/well) were challenged with P. gingivalis ATCC33277, and co-infected with L. rhamnosus Lr-32 for 4 h. L. rhamnosus Lr-32 spent medium or cells lysate was added to GECs co-infected with P. gingivalis. Another set of OBA-9 GECs were first exposed to P. gingivalis ATCC 33277 and then to the living probiotic or probiotic products. Transcription of genes encoding inflammatory mediators (IL-1β, TNF-α, IL-6, and CXCL-8) and receptors (TLR2 and TLR4) were evaluated by RT-qPCR. P. gingivalis growth under L. rhamnosus Lr-32 postbiotics was also evaluated.

Results

L. rhamnosus Lr-32 spent media decreased cell viability, while living cells and cell lysates did not. L. rhamnosus Lr-32 lysate, but not spent media, upregulated transcription of inflammatory mediators (IL-1β, TNF-α, IL-6, and CXCL-8) in GECs infected with P. gingivalis. Transcription of TRL2 was upregulated in all experimental groups compared to control, whereas TLR4 was upregulated by the probiotic or its postbiotics in P. gingivalis infected cells. Spent media and lysates reduced the growth of P. gingivalis.

Conclusion

L. rhamnosus Lr-32 cell components rather than live probiotic enhanced the expression of inflammatory mediators in P. gingivalis infected gingival epithelial cells. The increased potential of Lr-32 cell lysates to promote immune response to the periodontopathogen may favor pathogen elimination but may also lead to additional deleterious effects of the exacerbated inflammation.

Introduction

Probiotics improve health potentially through many mechanisms (Rolfe, 2000), including increasing epithelial barrier function (Ewaschuk et al., 2008) and modulation of the immune response (Isolauri, Sütas, Kankaanpää, Arvilommi, & Salminen, 2001; Lai et al., 2010). There has been increasing interest in probiotics control of periodontal diseases, and clinical trials have been conducted to elucidate their possible impact on oral health (Matsubara, Bandara, Ishikawa, Mayer, & Samaranayake, 2016). Plausible mechanisms of action for probiotics in periodontal diseases are based on modifications of the pathogenic potential of biofilm and include interference on growth and development of periodontal pathogens, the replacement of pathogenic microorganisms by beneficial bacteria, and prevention of colonization by periodontopathogens (Yanine et al., 2013).

The use of probiotics could be an alternative to the overuse of antibiotics, with no concerns regarding drug resistance (Bonifait, Chandad, & Grenier, 2009). Probiotic lactobacilli are very heterogeneous and we have shown that they can elicit distinct response in P. gingivalis infected gingival epithelial cells. Several tested lactobacilli were able to inhibit the adhesion to and invasion of P. gingivalis in gingival epithelial cells (GECs), but unexpectedly, the strain L. rhamnosus Lr-32 triggered GECs leading to increased production of IL-1β by P. gingivalis infected cells (Albuquerque‐Souza et al., 2019).

L. rhamnosus is frequently isolated from dairy products and humans, and most strains, especially the human isolates, display anti-microbial activity against pathogens (Douillard et al., 2013). The probiotic strain L. rhamnosus Lr-32 has been studied for human gastrointestinal affections and is considered safe for human consumption, inhibits pathogens such as Staphylococcus aureus and E. coli in vitro, and is able to modulate immune function (Memorandum, 2008). L. rhamnosus Lr-32 beneficial effect may be extended to the control of candidiasis since its administration as a cheese additive decreased Candida albicans colonization levels in dental wearers (Miyazima, Ishikawa, Mayer, Saad, & Nakamae, 2017). Although the mechanisms underlying the in vivo effect were not completely understood, in vitro data indicated that L. rhamnosus Lr-32 increased IL-1β and IL-10 production, and decreased IL-12 levels in THP1-macrophages with or without Candida albicans/and or lipopolysaccharide (LPS) challenge, with concomitant decreased expression of the receptors dectin-1 and TLR4 (tool-like-receptor 4) (Matsubara et al., 2017). Furthermore, L. rhamnosus Lr-32 reduced the transition of C. albicans from the commensal yeast form to the invasive hypha form in a biofilm in vitro model (Matsubara, Wang, Bandara, Mayer, & Samaranayake, 2016).

Considering L. rhamnosus Lr-32 potential use, and its intriguingly excessive proinflammatory profile, we evaluated the effects of its products on GECs infected with P. gingivalis. Published evidence suggests that products isolated from probiotics, termed postbiotics, would be as effective as probiotics (Adams, 2010; Kataria, Li, Wynn, & Neu, 2009; Zakostelska et al., 2011). Postbiotics comprise enzymes, teichoic acids, peptides derived from peptidoglycan, exopolysaccharides, surface and secreted proteins and bacteriocins generated by a probiotic organism (Konstantinov, Kuipers, & Peppelenbosch, 2013; Tsilingiri et al., 2012). These metabolites and signaling molecules exhibit a broad spectrum of antibacterial and immunomodulatory activities (Cicenia et al., 2014). Despite the proven benefits of probiotics, there are many concerns about their side effects such as the spread of antibiotic resistance genes, virulence factors of a given strain, translocation to tissues or blood, risk of sepsis in premature babies and impairment of normal microbiota colonization (Kataria et al., 2009). Postbiotics may also be advantageous over probiotics due to their chemical structure, safety dose parameters and longer shelf life (Shenderov, 2013). Thus, we hypothesized that products of L. rhamnosus Lr-32 (spent media and lysate) altered the gingival epithelial cells response induced by P. gingivalis.

Section snippets

Bacterial strains and growth conditions

The probiotic strain L. rhamnosus Lr-32™(Danisco, Madison, WI, USA) was cultivated under microaerophilic conditions (5% CO2, 37 °C) in broth and agar MRS (Man Rogosa and Sharp, Difco Laboratories, Detroit, MI, USA). P. gingivalis ATCC33277 was grown under anaerobic conditions (90 % N2, 5 % CO2 and 5 % H2, 37 °C) on blood agar plates [Brain Heart agar, Difco Laboratories) enriched with 5 % defibrinated sheep blood, 0.5 mg mL−1 hemin, and 1 mg mL−1menadione] and in BHHM broth [Brain Heart broth

Results

Both P. gingivalis and living probiotic L. rhamnosus Lr-32 did not alter the OBA-9 cell viability compared to control (p > 0.05). The spent media significantly reduced OBA-9 cell viability when compared to control (OBA-9 in KSFM without antibiotic) either used alone or together with P. gingivalis (in both situations: in co-culture with P. gingivalis for 4 h and after prior infection with P. gingivalis for 2 h) (p < 0.05). Cell viability of the other experimental groups did not differ from

Discussion

We investigated the effect of live probiotic L. rhamnosus Lr-32, probiotic lysate and spent culture medium on P. gingivalis-infected OBA-9 cells. Only spent culture medium significantly affected the viability of gingival epithelial cells either alone or in co-culture with P. gingivalis, possibly caused by some byproduct released by the bacteria in the spent media, such as organic acids produced during the probiotic metabolism. Although this is the first study that addresses the by-products of a

Funding information

This project was funded by the São Paulo Research Foundation (FAPESP), grant # 2015/18273-9. GV was supported by a FAPESP scholarship #2017/10829-3.

Data availability

All data generated or analysed during this study are included in this published article.

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Declaration of Competing Interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

References (59)

  • B.R. Aguilar - Uscanga et al.

    Effect of culture medium on bacteriocin production by Lactobacillus rhamnosus HN001 and lactobacillus reuteri ATCC 53608

    Journal of Microbiology, Biotechnology and Food Sciences

    (2013)
  • E. Albuquerque‐Souza et al.

    Probiotics alter the immune response of gingival epithelial cells challenged by Porphyromonas gingivalis

    Journal of Periodontal Research

    (2019)
  • T. Asahara et al.

    Protective effect of Lactobacillus casei strain Shirota against lethal infection with multi-drug resistant Salmonella enterica serovar Typhimurium DT104 in mice

    Journal of Applied Microbiology

    (2011)
  • S.P. Barros et al.

    Gingival crevicular fluid as a source of biomarkers for periodontitis

    Periodontology

    (2016)
  • A. Bascones et al.

    Tissue destruction in periodontitis: Bacteria or cytokines fault?

    Quintessence International (Berlin, Germany: 1985)

    (2005)
  • L. Bonifait et al.

    Probiotics for oral health: Myth or reality?

    Journal (Canadian Dental Association)

    (2009)
  • J. Cai et al.

    Recombinant fimbriae protein of Porphyromonas gingivalis induces an inflammatory response via the TLR4/NF-κB signaling pathway in human peripheral blood mononuclear cells

    International Journal of Molecular Medicine

    (2019)
  • A. Cicenia et al.

    Postbiotic activities of lactobacilli-derived factors

    Journal of Clinical Gastroenterology

    (2014)
  • R.P. Darveau et al.

    Porphyromonas gingivalis lipopolysaccharide contains multiple lipid a species that functionally interact with both toll-like receptors 2 and 4

    Infection and Immunity

    (2004)
  • M. Davey et al.

    Bacterial fimbriae stimulate proinflammatory activation in the endothelium through distinct TLRs

    The Journal of Immunology

    (2008)
  • R. Dimitrijević et al.

    The identification of a low molecular mass bacteriocin, rhamnosin A, produced by Lactobacillus rhamnosus strain 68

    Journal of Applied Microbiology

    (2009)
  • S. Doron et al.

    Risk and safety of probiotics

    Clinical Infectious Diseases

    (2015)
  • F.P. Douillard et al.

    Comparative genomic and functional analysis of 100 Lactobacillus rhamnosus strains and their comparison with strain GG

    PLoS Genetics

    (2013)
  • N. Dutzan et al.

    Interleukin-21 expression and its association with proinflammatory cytokines in untreated chronic periodontitis patients

    Journal of Periodontology

    (2012)
  • J.B. Ewaschuk et al.

    Secreted bioactive factors from Bifidobacterium infantis enhance epithelial cell barrier function

    American Journal of Physiology. Gastrointestinal and Liver Physiology

    (2008)
  • J.E. Fitzgerald et al.

    Localization of interleukin-8 in human gingival tissues

    Oral Microbiology and Immunology

    (1995)
  • J.A. Holden et al.

    Porphyromonas gingivalis lipopolysaccharide weakly activates M1 and M2 polarized mouse macrophages but induces inflammatory cytokines

    Infection and Immunity

    (2014)
  • E. Isolauri et al.

    Probiotics: Effects on immunity

    The American Journal of Clinical Nutrition

    (2001)
  • N. Kantrong et al.

    Gingival epithelial cell recognition of lipopolysaccharide

    Advances in Experimental Medicine and Biology

    (2019)
  • Cited by (32)

    • The effects of Lactobacillus reuteri on the inflammation and periodontal tissue repair in rats: A pilot study

      2022, Saudi Dental Journal
      Citation Excerpt :

      According to the World Health Organization (WHO) probiotics are described as ‘live microorganisms which, when administered in adequate amounts confer a health benefit on the host’ (WHO, 2002; Varela-López et al., 2018; Vale and Mayer, 2021). Compounds such as paraprobiotics (inactivated probiotic microorganisms) (Butera et al., 2021) and postbiotics (concentrated bacterial active metabolites) (Vale and Mayer, 2021; Butera et al., 2022) have showed promising results in in vitro and clinical studies in the field of dentistry. However, future research is required in order to improve current knowledge about all these treatment possibilities.

    View all citing articles on Scopus
    View full text