Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Microorganisms ; 12(6)2024 May 24.
Article in English | MEDLINE | ID: mdl-38930439

ABSTRACT

Periodontitis is a destructive inflammatory response triggered by dysbiosis. Lactobacillus acidophilus LA5 (LA5) may impair microbial colonization and alter the host. Thus, we evaluated the effect of LA5 on alveolar bone loss in a periodontitis murine model and investigated its effect on the oral and gut microbiomes. Porphyromonas gingivalis, Prevotella intermedia, Fusobacterium nucleatum, and Streptococcus gordonii were inoculated in C57BL/6 mice (P+), with LA5 (L+). SHAM infected controls (P- and/or L- groups) were also evaluated. After 45 days, alveolar bone loss in the maxilla and oral and gut microbiomes were determined. The administration of LA5 controlled the microbial consortium-induced alveolar bone loss. Periodontopathogens infection resulted in shifts in the oral and gut microbiomes consistent with dysbiosis, and LA5 reshaped these changes. The oral microbiome of P+L- group showed the increased abundance of Enterococaccea, Streptoccocaceae, Staphylococcaceae, Moraxellaceae, and Pseudomonadaceae, which were attenuated by the administration of LA5 to the infected group (P+L+). The administration of LA5 to otherwise non-infected mice resulted in the increased abundance of the superphylum Patescibacteria and the family Saccharamonadaceae in the gut. These data indicate L. acidophilus LA5 as a candidate probiotic for the control of periodontitis.

2.
Microorganisms ; 12(4)2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38674780

ABSTRACT

The benefits of probiotics on dysbiotic microbiomes and inflammation are dependent on the tested strain, host factors, and the resident microbiome. There is limited knowledge on the effects of probiotics in A. actinomycetemcomitans-associated periodontitis. Thus, Lactobacillus acidophilus LA5 (LA5) was orally inoculated for 30 days in C57Bl/6 mice infected with A. actinomycetemcomitans JP2 (Aa) and S. gordonii (Sg). Alveolar bone loss, gingival gene expression, and oral and gut microbiomes were determined. LA5 controlled bone loss in Aa+Sg-infected mice, downregulated the expression of Il-1ß and upregulated Il-10 in gingival tissues, and altered the oral and gut microbiomes. LA5 increased the diversity of the oral microbiome of Aa+Sg infected mice, and Aa+Sg and Aa+Sg+LA5 oral or gut microbiomes clustered apart. LA5 induced shifts in Aa+Sg infected mice by increasing the abundance of Muribaculaceae and decreasing Bifidobacteriaceae in the oral cavity and increasing the abundance of Verrucomicrobiae and Eggerthellales in the gut. In conclusion, LA5 oral administration controls experimental Aa-associated periodontitis by altering inflammatory gene expression and the oral and gut microbiomes.

3.
J Periodontal Res ; 58(5): 1006-1019, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37482954

ABSTRACT

OBJECTIVE: To determine whether Bifidobacterium animalis subspecies lactis HN019 (B. lactis HN019) can reduce the sequelae of experimental periodontitis (EP) in rats modulating systemic parameters. BACKGROUND: This study evaluated the effects of probiotic therapy (PROB) in the prevention of local and systemic damage resulting from EP. METHODS: Forty-eight rats were allocated into four groups: C (control), PROB, EP, and EP-PROB. PROB (1 × 1010 CFU/mL) administration lasted 8 weeks and PE was induced on the 7th week by placing ligature on the animals' lower first molars. All animals were euthanized in the 9th week of the experiment. Biomolecular analyses, RT-PCR, and histomorphometric analyses were performed. The data obtained were analyzed statistically (ANOVA, Tukey, p < .05). RESULTS: The EP group had higher dyslipidemia when compared to the C group, as well as higher levels of insulin resistance, proteinuria levels, percentages of systolic blood pressure, percentage of fatty hepatocytes in the liver, and expression of adipokines was up-regulated (LEPR, NAMPT, and FABP4). All these parameters (except insulin resistance, systolic blood pressure, LEPR and FABP4 gene expression) were reduced in the EP-PROB group when compared to the EP group. The EP group had lower villus height and crypt depth, as well as a greater reduction in Bacteroidetes and a greater increase in Firmicutes when compared to the EP-PROB group. Greater alveolar bone loss was observed in the EP group when compared to the EP-PROB group. CONCLUSION: Bifidobacterium lactis HN019 can reduce the sequelae of EP in rats modulating intestinal parameters, attenuating expression of lipogenic genes and hepatic steatosis.


Subject(s)
Bifidobacterium animalis , Fatty Liver , Insulin Resistance , Periodontitis , Probiotics , Rats , Animals , Bifidobacterium animalis/physiology , Probiotics/therapeutic use , Periodontitis/prevention & control , Intestinal Mucosa
4.
Heliyon ; 9(6): e16429, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37484240

ABSTRACT

Background: /Aims: Epidemiological data show that there is an important relationship between respiratory and intestinal diseases. To improve our understanding on the interconnectedness between the lung and intestinal mucosa and the overlap between respiratory and intestinal diseases, our aim was to investigate the influence of ovalbumin (OVA)-induced allergic airway inflammation on gut homeostasis. Methods: A/J mice were sensitized and challenged with OVA. The animals were euthanized 24 h after the last challenge, lung inflammation was determined by evaluating cells in Bronchoalveolar lavage fluid, serum anti-OVA IgG titers and colon morphology, inflammation and integrity of the intestinal mucosa were investigated. IL-4 and IL-13 levels and myeloperoxidase activity were determined in the colon samples. The expression of genes involved in inflammation and mucin production at the gut mucosa was also evaluated. Results: OVA challenge resulted not only in lung inflammation but also in macroscopic alterations in the gut such as colon shortening, increased myeloperoxidase activity and loss of integrity in the colonic mucosal. Neutral mucin intensity was lower in the OVA group, which was followed by down-regulation of transcription of ATOH1 and up-regulation of TJP1 and MUC2. In addition, the OVA group had higher levels of IL-13 and IL-4 in the colon. Ova-specific IgG1 and OVA-specific IgG2a titers were higher in the serum of the OVA group than in controls. Conclusions: Our data using the OVA experimental model suggested that challenges in the respiratory system may result not only in allergic airway inflammation but also in the loss of gut homeostasis.

5.
J Periodontol ; 94(11): 1363-1375, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37057371

ABSTRACT

BACKGROUND: This study evaluated the systemic (intestine and adipose tissue) and local (periodontal tissues) impact of probiotic therapy in rats with metabolic syndrome (MS) associated or not with periodontitis (PE). METHODS: Forty-eight rats received a high-fat diet for induction of MS for 16 weeks. They were subdivided into groups with (+) and without (-) PE, receiving (*) or not (**) receiving probiotics (PROB): MS (-**), MSP (-*), MSPE (+**), and MSPEP (+*). PROB administration (Bifidobacterium animalis subsp. lactis HN019) started on the 8th week of the study and PE was induced on the 14th week by placing ligature on the animals' lower first molars. Euthanasia occurred in the 16th week. Biomolecular, immunoenzymatic assays, and histomorphometric analyses were performed. The data obtained were statistically analyzed (ANOVA, Tukey, p < 0.05). RESULTS: The MSPEP group exhibited reduced alveolar bone loss when compared with the MSPE group, as well as lower levels of hepatic steatosis and proteinuria (p < 0.05). In the intestinal environment, the MSPE group exhibited significantly lower villus height and crypt depth, as well as a greater increase in Bacillota when compared with the MSPEP group (p < 0.05). The MSPEP group showed lower adipokine gene expression (LEPR, NAMPT, and FABP4) in adipose tissue than the MSPE group (p < 0.05). CONCLUSION: The probiotic B. lactis HN019 reduced the severity of experimental periodontitis and modulated the expression of lipogenic genes and intestinal morphological and microbiological parameters in rats with MS.


Subject(s)
Bifidobacterium animalis , Metabolic Syndrome , Periodontitis , Probiotics , Rats , Animals , Metabolic Syndrome/complications , Periodontitis/therapy , Periodontitis/metabolism , Intestines/microbiology , Probiotics/therapeutic use , Probiotics/pharmacology
6.
Heliyon, v. 9, n. 6, e16429, jun. 2023
Article in English | Sec. Est. Saúde SP, SESSP-IBPROD, Sec. Est. Saúde SP | ID: bud-4971

ABSTRACT

Background Aims: Epidemiological data show that there is an important relationship between respiratory and intestinal diseases. To improve our understanding on the interconnectedness between the lung and intestinal mucosa and the overlap between respiratory and intestinal diseases, our aim was to investigate the influence of ovalbumin (OVA)-induced allergic airway inflammation on gut homeostasis. Methods A/J mice were sensitized and challenged with OVA. The animals were euthanized 24 h after the last challenge, lung inflammation was determined by evaluating cells in Bronchoalveolar lavage fluid, serum anti-OVA IgG titers and colon morphology, inflammation and integrity of the intestinal mucosa were investigated. IL-4 and IL-13 levels and myeloperoxidase activity were determined in the colon samples. The expression of genes involved in inflammation and mucin production at the gut mucosa was also evaluated. Results OVA challenge resulted not only in lung inflammation but also in macroscopic alterations in the gut such as colon shortening, increased myeloperoxidase activity and loss of integrity in the colonic mucosal. Neutral mucin intensity was lower in the OVA group, which was followed by down-regulation of transcription of ATOH1 and up-regulation of TJP1 and MUC2. In addition, the OVA group had higher levels of IL-13 and IL-4 in the colon. Ova-specific IgG1 and OVA-specific IgG2a titers were higher in the serum of the OVA group than in controls. Conclusions Our data using the OVA experimental model suggested that challenges in the respiratory system may result not only in allergic airway inflammation but also in the loss of gut homeostasis.

7.
Adv Exp Med Biol ; 1373: 353-375, 2022.
Article in English | MEDLINE | ID: mdl-35612808

ABSTRACT

Scaling and root planing is the gold standard for the treatment of periodontitis, but administration of systemic antibiotics may be needed especially for sites with deep probing depths, or in the presence of comorbidities. However, treated sites are subject to recolonization with a microbiota similar to that present before therapy, and supportive periodontal therapy is employed after the treatment of active disease. The use of beneficial organisms, known as probiotics, seems an attractive proposal to promote a healthy associated subgingival microbiome and to control inflammation for the management of periodontitis. The mechanisms underlying the benefits promoted by probiotics involve interference on periodontopathogens, modulation of the exacerbated immune host response and the ability to restore the integrity of the epithelial barrier on mucosa surfaces. This review examines the scientific data related to the effects of probiotics on the treatment of periodontal diseases and addresses the future approaches necessary for their implementation.


Subject(s)
Periodontitis , Probiotics , Anti-Bacterial Agents/therapeutic use , Humans , Periodontitis/drug therapy , Probiotics/therapeutic use , Root Planing
8.
Front Microbiol ; 13: 846192, 2022.
Article in English | MEDLINE | ID: mdl-35602018

ABSTRACT

Probiotics may be considered as an additional strategy to achieve a balanced microbiome in periodontitis. However, the mechanisms underlying the use of probiotics in the prevention or control of periodontitis are still not fully elucidated. This in vitro study aimed to evaluate the effect of two commercially available strains of lactobacilli on gingival epithelial cells (GECs) challenged by Aggregatibacter actinomycetemcomitans. OBA-9 GECs were infected with A. actinomycetemcomitans strain JP2 at an MOI of 1:100 and/or co-infected with Lactobacillus acidophilus La5 (La5) or Lacticaseibacillus rhamnosus Lr32 (Lr32) at an MOI of 1:10 for 2 and 24 h. The number of adherent/internalized bacteria to GECs was determined by qPCR. Production of inflammatory mediators (CXCL-8, IL-1ß, GM-CSF, and IL-10) by GECs was determined by ELISA, and the expression of genes encoding cell receptors and involved in apoptosis was determined by RT-qPCR. Apoptosis was also analyzed by Annexin V staining. There was a slight loss in OBA-9 cell viability after infection with A. actinomycetemcomitans or the tested probiotics after 2 h, which was magnified after 24-h co-infection. Adherence of A. actinomycetemcomitans to GECs was 1.8 × 107 (± 1.2 × 106) cells/well in the mono-infection but reduced to 1.2 × 107 (± 1.5 × 106) in the co-infection with Lr32 and to 6 × 106 (± 1 × 106) in the co-infection with La5 (p < 0.05). GECs mono-infected with A. actinomycetemcomitans produced CXCL-8, GM-CSF, and IL-1ß, and the co-infection with both probiotic strains altered this profile. While the co-infection of A. actinomycetemcomitans with La5 resulted in reduced levels of all mediators, the co-infection with Lr32 promoted reduced levels of CXCL-8 and GM-CSF but increased the production of IL-1ß. The probiotics upregulated the expression of TLR2 and downregulated TLR4 in cells co-infected with A. actinomycetemcomitans. A. actinomycetemcomitans-induced the upregulation of NRLP3 was attenuated by La5 but increased by Lr32. Furthermore, the transcription of the anti-apoptotic gene BCL-2 was upregulated, whereas the pro-apoptotic BAX was downregulated in cells co-infected with A. actinomycetemcomitans and the probiotics. Infection with A. actinomycetemcomitans induced apoptosis in GECs, whereas the co-infection with lactobacilli attenuated the apoptotic phenotype. Both tested lactobacilli may interfere in A. actinomycetemcomitans colonization of the oral cavity by reducing its ability to interact with gingival epithelial cells and modulating cells response. However, L. acidophilus La5 properties suggest that this strain has a higher potential to control A. actinomycetemcomitans-associated periodontitis than L. rhamnosus Lr32.

9.
Front Pharmacol ; 12: 713595, 2021.
Article in English | MEDLINE | ID: mdl-34630089

ABSTRACT

Periodontitis is an inflammatory disease induced by a dysbiotic oral microbiome. Probiotics of the genus Bifidobacterium may restore the symbiotic microbiome and modulate the immune response, leading to periodontitis control. We evaluated the effect of two strains of Bifidobacterium able to inhibit Porphyromonas gingivalis interaction with host cells and biofilm formation, but with distinct immunomodulatory properties, in a mice periodontitis model. Experimental periodontitis (P+) was induced in C57Bl/6 mice by a microbial consortium of human oral organisms. B. bifidum 1622A [B+ (1622)] and B. breve 1101A [B+ (1101)] were orally inoculated for 45 days. Alveolar bone loss and inflammatory response in gingival tissues were determined. The microbial consortium induced alveolar bone loss in positive control (P + B-), as demonstrated by microtomography analysis, although P. gingivalis was undetected in oral biofilms at the end of the experimental period. TNF-α and IL-10 serum levels, and Treg and Th17 populations in gingiva of SHAM and P + B- groups did not differ. B. bifidum 1622A, but not B. breve 1101A, controlled bone destruction in P+ mice. B. breve 1101A upregulated transcription of Il-1ß, Tnf-α, Tlr2, Tlr4, and Nlrp3 in P-B+(1101), which was attenuated by the microbial consortium [P + B+(1101)]. All treatments downregulated transcription of Il-17, although treatment with B. breve 1101A did not yield such low levels of transcripts as seen for the other groups. B. breve 1101A increased Th17 population in gingival tissues [P-B+ (1101) and P + B+ (1101)] compared to SHAM and P + B-. Administration of both bifidobacteria resulted in serum IL-10 decreased levels. Our data indicated that the beneficial effect of Bifidobacterium is not a common trait of this genus, since B. breve 1101A induced an inflammatory profile in gingival tissues and did not prevent alveolar bone loss. However, the properties of B. bifidum 1622A suggest its potential to control periodontitis.

10.
Anaerobe ; 72: 102458, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34547426

ABSTRACT

Porphyromonas gingivalis inhibits the release of CXCL8 by gingival epithelial cells and reduces their proliferation. We previously reported that Bifidocaterium sp. and Lactobacillus sp. immunomodulate gingival epithelial cells response to this periodontal pathogen, but their effects on re-epithelialization properties are still unknown. Herein we explored these activities of potential probiotics on gingival epithelial cells and clarified their mechanisms. The immortalized OBA-9 lineage was used to perform in vitro scratches. Twelve clinical isolates and commercially available strains of Bifidobacterium sp. and Lactobacillus sp. were screened. L. casei 324 m and B. pseudolongum 1191A were selected to perform mechanistic assays with P. gingivalis W83 infection and the following parameters were measured: percentage of re-epithelialization by DAPI immunofluorescence area measurement; cell number by Trypan Blue exclusion assay; CXCL8 regulation by ELISA and RT-qPCR; and expression of CXCL8 cognate receptors-CXCR1 and CXCR2 by Flow Cytometry. Complementary mechanistic assays were performed with CXCL8, in the presence or absence of the CXCR1/CXCR2 inhibitor-reparixin. L. casei 324 m and B. pseudolongum 1191A enhanced re-epithelialization/cell proliferation as well as inhibited the harmful effects of P. gingivalis W83 on these activities through an increase in the expression and release of CXCL8 and in the number of cells positive for CXCR1/CXCR2. Further, we revealed that the beneficial effects of these potential probiotics were dependent on activation of the CXCL8-CXCR1/CXCR2 axis. The current findings indicate that these potential probiotics strains may improve wound healing in the context of the periodontal tissues by a CXCL8 dependent mechanism.


Subject(s)
Bacteroidaceae Infections/metabolism , Bacteroidaceae Infections/microbiology , Host-Pathogen Interactions , Microbial Interactions , Porphyromonas gingivalis , Probiotics/administration & dosage , Re-Epithelialization , Biomarkers , Cell Line , Gene Expression Regulation , Humans , Interleukin-8/genetics , Interleukin-8/metabolism , Receptors, Interleukin-8A/antagonists & inhibitors , Receptors, Interleukin-8A/genetics , Receptors, Interleukin-8A/metabolism , Receptors, Interleukin-8B/antagonists & inhibitors , Receptors, Interleukin-8B/genetics , Receptors, Interleukin-8B/metabolism , Signal Transduction , Wound Healing
11.
Mol Oral Microbiol ; 36(1): 92-102, 2021 02.
Article in English | MEDLINE | ID: mdl-33372378

ABSTRACT

Periodontitis is characterized by a dysbiotic microbial community and treatment strategies include the reestablishment of symbiosis by reducing pathogens abundance. Aggregatibacter actinomycetemcomitans (Aa) is frequently associated with rapidly progressing periodontitis. Since the oral ecosystem may be affected by metabolic end-products of bacteria, we evaluated the effect of soluble compounds released by probiotic lactobacilli, known as postbiotics, on Aa biofilm and expression of virulence-associated genes. Cell-free pH-neutralized supernatants (CFS) of Lactobacillus rhamnosus Lr32, L. rhamnosus HN001, Lactobacillus acidophilus LA5, and L. acidophilus NCFM were tested against a fimbriated clinical isolate of Aa JP2 genotype (1 × 107  CFU/well) on biofilm formation for 24 hr, and early and mature preformed biofilms (2 and 24 hr). Lactobacilli CFS partially reduced Aa viable counts and biofilms biomass, but did not affect the number of viable non-adherent bacteria, except for LA5 CFS. Furthermore, LA5 CFS and, in a lesser extent HN001 CFS, influenced Aa preformed biofilms. Lactobacilli postbiotics altered expression profile of Aa in a strain-specific fashion. Transcription of cytolethal distending toxin (cdtB) and leukotoxin (ltxA) was downregulated by CFS of LA5 and LR32 CFS. Although all probiotics produced detectable peroxide, transcription of katA was downregulated by lactobacilli CFS. Transcription of dspB was abrogated by LR32 and NCFM CFS, but increased by HN001, whereas expression of pgA was not affected by any postbiotic. Our data indicated the potential of postbiotics from lactobacilli, especially LA5, to reduce colonization levels of Aa and to modulate the expression of virulence factors implicated in evasion of host defenses.


Subject(s)
Lactobacillus , Probiotics , Aggregatibacter actinomycetemcomitans/genetics , Biofilms , Ecosystem , Lactobacillus/genetics , Virulence
12.
J Oral Microbiol ; 12(1): 1805553, 2020 Aug 20.
Article in English | MEDLINE | ID: mdl-32944156

ABSTRACT

BACKGROUND AND OBJECTIVE: The potential of probiotics on the prevention and control of periodontitis and other chronic inflammatory conditions has been suggested. Lactobacillus and Bifidobacterium species influence P. gingivalis interaction with gingival epithelial cells (GECs) but may not act in a unique way. In order to select the most appropriate probiotic against P. gingivalis, we aimed to evaluate the effect of several strains on Porphyromonas gingivalis biofilm formation and transcription virulence-associated factors (PgVAFs). METHODS: Cell-free pH neutralized supernatants (CFS) and living Lactobacillus spp. and Bifidobacterium spp. were tested against P. gingivalis ATCC 33277 and W83, in mono- and multi-species (with Streptococcus oralis and S. gordonii) biofilms. Relative transcription of P. gingivalis genes (fimA, mfa1, kgp, rgp, ftsH and luxS) was determined in biofilms and under GECs co-infection. RESULTS: Probiotics CFS reduced P. gingivalis ATCC 33277 levels in mono-species biofilms and living probiotics reduced P. gingivalis abundance in multi-species biofilms. L. acidophilus LA5 down-regulated transcription of most PgVAFs in biofilms and GECs. CONCLUSIONS: Probiotics affect P. gingivalis biofilm formation by down-regulating overall PgVAFs with the most pronounced effect observed for L. acidophilus LA5.

13.
J Periodontol ; 91(6): 775-783, 2020 06.
Article in English | MEDLINE | ID: mdl-31799694

ABSTRACT

BACKGROUND: The combination of probiotics and prebiotics might be useful to treat oral halitosis. The aim of this study was to assess the effect of Lactobacillus salivarius G60 (LS) and inulin on oral halitosis and tongue coating. METHODS: In this double-masked, randomized, phase II clinical trial, 45 patients (aged 35 ± 15 years, 66% female) with oral halitosis and tongue coating were allocated to three treatment groups (n = 15) using gums of oral dissolution (one gum every 12 hours) for 10 days. Each gum contained LS (1 billion colony forming units [CFUs]) + inulin (1 g), LS (1 billion CFU) or placebo. Primary outcomes were organoleptic test, Halimeter, and tongue coating, whereas secondary outcomes were quality of life (QOL) and treatment safety. Generalized linear models were used, adjusting for age and sex. In vitro tests were performed to verify whether LS interacts with inulin and whether LS inhibits the growth of Porphyromonas gingivalis and Prevotella intermedia. RESULTS: Forty-four patients (97%) completed the study. Patients treated with LS + inulin showed greater reduction in halitosis measured by Halimeter compared with placebo (adjusted post-intervention average: 96.7 versus 142.5 ppb; P = 0.003), whereas LS and placebo did not differ (115.7 versus 142.5 ppb; P = 0.097). Organoleptic measurements and coating index showed a similar decrease for all groups. QOL improved in patients treated with LS + inulin compared with placebo (P = 0.029). Side effects were mild and transient in all groups. LS did not metabolize inulin but inhibited the growth of P. gingivalis and P. intermedia after 72 hours. CONCLUSIONS: Treatment with L. salivarius G60 combined or not with inulin showed significant decrease in the outcomes organoleptic test, Halimeter, and coating index, improving oral halitosis. However, no significant difference was obtained between the groups.


Subject(s)
Halitosis , Ligilactobacillus salivarius , Probiotics , Adult , Female , Halitosis/drug therapy , Humans , Inulin/therapeutic use , Male , Middle Aged , Probiotics/therapeutic use , Quality of Life , Young Adult
14.
J Periodontal Res ; 54(2): 115-127, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30284741

ABSTRACT

BACKGROUND AND OBJECTIVE: Although previous studies revealed the potential use of probiotics in the control of periodontitis, little is known about their interactions with gingival epithelial cells (GECs). Since GECs comprise the first defense in the subgingival microenvironment, the aim of this study was to evaluate the effect of probiotic lactobacilli and bifidobacteria strains on OBA-9 cells challenged with Porphyromonas gingivalis. METHODS: Immortalized human GECs (OBA-9) were challenged with live P. gingivalis (strains W83 and ATCC33277) and co-infected with one of 12 tested probiotic strains at a multiplicity of infection (MOI) of 1:1000 for 2 hours. Bacterial adhesion and invasion were determined by antibiotic exclusion analysis and CFU counting. OBA-9 viability was assessed by MTT assay, and levels of inflammatory mediators (TNF-α, IL-1ß, and CXCL8) in the supernatants were determined by ELISA. The expression of genes encoding Toll-like receptors (TLR2, TLR4) was evaluated by RT-qPCR. RESULTS: Both strains of P. gingivalis were able to adhere and invade OBA-9 cells, with significant loss in cell viability, increase in the levels of TNF-α and IL-1ß, and upregulation of TLR4. However, co-infection with probiotics attenuated these effects in P. gingivalis challenged GECs. Most probiotics maintained OBA-9 viability and reduced pathogens adhesion and invasion. Furthermore, probiotics were able to adhere to GECs, which was enhanced for most strains in the presence of P. gingivalis. The synthesis of IL-1ß and TNF-α by P. gingivalis in challenged GECs was reduced in co-culture with most of the tested probiotics, whereas the secretion of CXCL8 increased, and TLR4 was downregulated. CONCLUSION: Probiotics can alter the interaction of GECs with P. gingivalis by modulating the pathogen's ability to adhere and invade these cells, as well as by regulating the innate immune response. Such properties are strain-specific and may indicate the most efficient probiotics to control periodontitis.


Subject(s)
Antibiosis/immunology , Bifidobacterium/physiology , Epithelial Cells/immunology , Epithelial Cells/microbiology , Gingiva/cytology , Gingiva/immunology , Immunity, Innate , Lactobacillus/physiology , Periodontitis/prevention & control , Periodontitis/therapy , Porphyromonas gingivalis/immunology , Porphyromonas gingivalis/pathogenicity , Probiotics , Cells, Cultured , Cellular Microenvironment/immunology , Humans , Interleukin-1beta/metabolism , Interleukin-8/metabolism , Periodontitis/immunology , Periodontitis/microbiology , Porphyromonas gingivalis/metabolism , Toll-Like Receptor 4/metabolism , Tumor Necrosis Factor-alpha/metabolism
15.
Front Microbiol ; 8: 2280, 2017.
Article in English | MEDLINE | ID: mdl-29238325

ABSTRACT

Probiotics are live microorganisms that confer benefits to the host health. The infection rate of potentially pathogenic organisms such as Candida albicans, the most common agent associated with mucosal candidiasis, can be reduced by probiotics. However, the mechanisms by which the probiotics interfere with the immune system are largely unknown. We evaluated the effect of probiotic bacteria on C. albicans challenged human macrophages. Macrophages were pretreated with lactobacilli alone (Lactobacillus rhamnosus LR32, Lactobacillus casei L324m, or Lactobacillus acidophilus NCFM) or associated with Escherichia coli lipopolysaccharide (LPS), followed by the challenge with C. albicans or LPS in a co-culture assay. The expression of pattern-recognition receptors genes (CLE7A, TLR2, and TLR4) was determined by RT-qPCR, and dectin-1 reduced levels were confirmed by flow cytometry. The cytokine profile was determined by ELISA using the macrophage cell supernatant. Overall probiotic lactobacilli down-regulated the transcription of CLEC7A (p < 0.05), resulting in the decreased expression of dectin-1 on probiotic pretreated macrophages. The tested Lactobacillus species down-regulated TLR4, and increased TLR2 mRNA levels in macrophages challenged with C. albicans. The cytokines profile of macrophages challenged with C. albicans or LPS were altered by the probiotics, which generally led to increased levels of IL-10 and IL-1ß, and reduction of IL-12 production by macrophages (p < 0.05). Our data suggest that probiotic lactobacilli impair the recognition of PAMPs by macrophages, and alter the production of pro/anti-inflammatory cytokines, thus modulating inflammation.

16.
Expert Rev Anti Infect Ther ; 14(7): 643-55, 2016 07.
Article in English | MEDLINE | ID: mdl-27224284

ABSTRACT

INTRODUCTION: The frequent recolonization of treated sites by periodontopathogens and the emergence of antibiotic resistance have led to a call for new therapeutic approaches for managing periodontal diseases. As probiotics are considered a new tool for combating infectious diseases, we systematically reviewed the evidences for their effectiveness in the management of periodontitis. AREAS COVERED: An electronic search was performed in the MEDLINE, SCOPUS and Cochrane Library databases up to March 2016 using the terms 'periodontitis', 'chronic periodontitis', 'probiotic(s)', 'prebiotic(s)', 'symbiotic(s)', 'Bifidobacterium and 'Lactobacillus'. Only randomized controlled trials (RCTs) were included in the present study. Analysis of 12 RCTs revealed that in general, oral administration of probiotics improved the recognized clinical signs of chronic and aggressive periodontitis such as probing pocket depth, bleeding on probing, and attachment loss, with a concomitant reduction in the levels of major periodontal pathogens. Continuous probiotic administration, laced mainly with Lactobacillus species, was necessary to maintain these benefits. Expert commentary: Oral administration of probiotics is a safe and effective adjunct to conventional mechanical treatment (scaling) in the management of periodontitis, specially the chronic disease entity. Their adjunctive use is likely to improve disease indices and reduce the need for antibiotics.


Subject(s)
Bifidobacterium , Dental Scaling , Lactobacillus , Periodontitis/drug therapy , Probiotics/therapeutic use , Combined Modality Therapy , Dietary Supplements , Humans , Periodontal Attachment Loss/prevention & control , Periodontal Pocket/prevention & control , Periodontitis/therapy , Probiotics/administration & dosage , Randomized Controlled Trials as Topic , Treatment Outcome
17.
J Prosthodont ; 24(3): 194-9, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25143068

ABSTRACT

PURPOSE: The prevalence of Candida infections has been rising with an increasingly aging population and a larger population of immunocompromised individuals. The use of probiotics may be an alternative approach to antifungal agents in the prevention and treatment of oral candidiasis. This study aimed to evaluate the short-term effect of probiotics in reducing the infection level of oral Candida in candidiasis-asymptomatic elderly denture wearers. MATERIALS AND METHODS: In a double-blind randomized study, 59 denture wearers harboring Candida spp. in the oral cavity with no clinical symptoms were allocated into two groups: probiotic and placebo. All patients were instructed to clean the denture daily. The probiotic group poured a capsule containing lyophilized Lactobacillus rhamnosus HS111, Lactobacillus acidophillus HS101, and Bifidobacterium bifidum daily on the palatal surface of the maxillary denture, whereas the placebo group was submitted to the same regimen using placebo capsules. Candida spp. infection levels were evaluated in palate mucosa samples obtained before and after a 5-week experimental period. RESULTS: All patients harbored Candida in the palate mucosa at baseline. Fifty-five individuals completed the experimental period. The detection rate of Candida spp. was 92.0% in the placebo group after the experimental period, whereas it was reduced to 16.7% in the probiotic group. The reduction promoted by the probiotic regimen was independent of baseline characteristics such as Candida infection level and colonizing species, age of denture, and other variables. CONCLUSION: The probiotic product was effective in reducing the colonization of the oral cavity with Candida in candidiasis-asymptomatic elderly denture wearers, suggesting that this multispecies probiotic could be used to prevent oral candidiasis. CLINICAL IMPLICATIONS: Colonization of oral surfaces by Candida is considered a risk factor for invasive fungal infections. The use of a product with L. rhamnosus, L. acidophilus, and B. bifidum may represent an alternative treatment for reduction of Candida infections in elderly denture wearers.


Subject(s)
Candida/drug effects , Candidiasis, Oral/prevention & control , Candidiasis, Oral/therapy , Dentures/microbiology , Mouth/microbiology , Probiotics/therapeutic use , Aged , Bifidobacterium bifidum , Brazil , Dental Restoration Wear/adverse effects , Double-Blind Method , Female , Humans , Lactobacillus acidophilus , Lacticaseibacillus rhamnosus , Male , Middle Aged , Palate/microbiology
18.
São Paulo; s.n; 2011. 151 p. ilus, tab, graf. (BR).
Thesis in Portuguese | LILACS, BBO - Dentistry | ID: lil-654813

ABSTRACT

Probióticos são microrganismos vivos que conferem benefícios à saúde dos indivíduos, quando administrados em quantidades adequadas. Além de promoverem uma melhora no sistema gastrointestinal, os probióticos são conhecidos por sua capacidade de inibirem o crescimento de microrganismos patogênicos. O presente trabalho objetivou avaliar a influência dos probióticos na redução da prevalência de Candida oral de indivíduos usuários de próteses totais. Para realização deste estudo foi elaborado o bioproduto contendo probióticos, Lactobacillus rhamnosus e Lactobacillus acidophilus. O protocolo do estudo foi duplo-cego randomizado, sendo que os sujeitos da pesquisa foram divididos aleatoriamente em dois grupos: experimental (probióticos) e controle (placebo). Os participantes utilizaram o produto durante cinco semanas e o resultado foi avaliado segundo a contagem de leveduras isoladas em unidades formadoras de colônias (UFC/mL), antes e após a intervenção. Paralelamente, a esta fase clínica da pesquisa, foram testadas in vitro a ação de outras cepas de Lactobacillus com a finalidade de verificar a associação entre a cepa de Lactobacillus com a redução das espécies de Candida isoladas do palato e das próteses, bem como, com a sensibilidade aos antifúngicos, miconazol e nistatina. Também foi pesquisado um dos principais mecanismos de ação contra patógenos, por Lactobacillus spp, a produção de bacteriocinas. Os resultados inferem que as cepas de probióticos testadas: foram eficazes na redução da prevalência de Candida oral de usuários de próteses totais; in vitro, inibiram o crescimento de Candida sp, sendo L. rhamnosus Lr-32 a cepa com melhor resultado e não produzem bacteriocinas quando em contato com a levedura.


Probiotics are live microorganisms that confer benefits to the health of an individual when administered in adequate amounts. In addition to promoting an improvement in a gastrointestinal system, the probiotics are known for their ability to inhibit the growth of pathogenic microorganisms. The present study sought to evaluate the capacity of probiotics strains in reducing the prevalence of oral Candida in denture wearers. For this study, the by-product was elaborated containing probiotics, such as Lactobacillus rhamnosus and Lactobacillus acidophilus. The study protocol was randomized, double-blind, being that the subjects of the research were randomly sorted into two groups: experimental (probiotics) and control (placebo). The participants utilized the product daily for five weeks and the result was evaluated according to the amount of yeasts isolated in colony-forming units (CFU), before and after the intervention. In parallel to this clinical phase, the action of other Lactobacillus strains were tested in vitro, with the purpose of verifying the relation between the strain of Lactobacillus and the reduction of Candida species isolated from the palate and the prostheses, as well as, with the sensitivity to antifungal agents, miconazole and nystatin. The production of bacteriocins is one of the main mechanisms of action by Lactobacillus spp. against pathogens, and was analised. The results imply that the strains of tested probiotics: in vivo, were effective in reducing the prevalence of oral Candida for denture wearers; in vitro, inhibited the growth of Candida sp, being that L. rhamnosus Lr-32 was the best resulting strain, and the strains did not produce bacteriocins when in contact with yeast.


Subject(s)
Humans , Male , Female , Mouth/microbiology , Candida , Stomatitis, Denture/diagnosis , Lactobacillus , Denture, Complete
19.
Braz Oral Res ; 23(1): 68-75, 2009.
Article in English | MEDLINE | ID: mdl-19488475

ABSTRACT

The aim of this study was to evaluate the following acrylic resins: Clássico, QC-20 and Lucitone, recommended specifically for thermal polymerization, and Acron MC and VIPI-WAVE, made for polymerization by microwave energy. The resins were evaluated regarding their surface nanohardness and modulus of elasticity, while varying the polymerization time recommended by the manufacturer. They were also compared as to the presence of water absorbed by the samples. The technique used was nanoindentation, using the Nano Indenter XP, MTS. According to an intra-group analysis, when using the polymerization time recommended by the manufacturer, a variation of 0.14 to 0.23 GPa for nanohardness and 2.61 to 3.73 GPa for modulus of elasticity was observed for the thermally polymerized resins. The variation for the resins made for polymerization by microwave energy was 0.15 to 0.22 GPa for nanohardness and 2.94 to 3.73 GPa for modulus of elasticity. The conclusion was that the Classico resin presented higher nanohardness and higher modulus of elasticity values when compared to those of the same group, while Acron MC presented the highest values for the same characteristics when compared to those of the same group. The water absorption evaluation showed that all the thermal polymerization resins, except for Lucitone, presented significant nanohardness differences when submitted to dehydration or rehydration, while only Acron MC presented no significant differences when submitted to a double polymerization time. Regarding the modulus of elasticity, it was observed that all the tested materials and products, except for Lucitone, showed a significant increase in modulus of elasticity when submitted to a lack of hydration.


Subject(s)
Acrylic Resins , Elasticity , Materials Testing , Acrylic Resins/chemistry , Hardness , Hardness Tests , Hot Temperature , Humans , Microwaves , Surface Properties , Time Factors
20.
Braz. oral res ; 23(1): 68-75, 2009. ilus, graf, tab
Article in English | LILACS | ID: lil-514644

ABSTRACT

The aim of this study was to evaluate the following acrylic resins: Clássico®, QC-20® and Lucitone®, recommended specifically for thermal polymerization, and Acron MC® and VIPI-WAVE®, made for polymerization by microwave energy. The resins were evaluated regarding their surface nanohardness and modulus of elasticity, while varying the polymerization time recommended by the manufacturer. They were also compared as to the presence of water absorbed by the samples. The technique used was nanoindentation, using the Nano Indenter XP®, MTS. According to an intra-group analysis, when using the polymerization time recommended by the manufacturer, a variation of 0.14 to 0.23 GPa for nanohardness and 2.61 to 3.73 GPa for modulus of elasticity was observed for the thermally polymerized resins. The variation for the resins made for polymerization by microwave energy was 0.15 to 0.22 GPa for nanohardness and 2.94 to 3.73 GPa for modulus of elasticity. The conclusion was that the Classico® resin presented higher nanohardness and higher modulus of elasticity values when compared to those of the same group, while Acron MC® presented the highest values for the same characteristics when compared to those of the same group. The water absorption evaluation showed that all the thermal polymerization resins, except for Lucitone®, presented significant nanohardness differences when submitted to dehydration or rehydration, while only Acron MC® presented no significant differences when submitted to a double polymerization time. Regarding the modulus of elasticity, it was observed that all the tested materials and products, except for Lucitone®, showed a significant increase in modulus of elasticity when submitted to a lack of hydration.


Subject(s)
Humans , Acrylic Resins , Elasticity , Materials Testing , Acrylic Resins/chemistry , Hardness , Hardness Tests , Hot Temperature , Microwaves , Surface Properties , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...