Next Article in Journal
Association of KRAS Mutation and Gene Pathways in Colorectal Carcinoma: A Transcriptome- and Methylome-Wide Study and Potential Implications for Therapy
Previous Article in Journal
An In Vivo Model of Propionic Acid-Rich Diet-Induced Gliosis and Neuro-Inflammation in Mice (FVB/N-Tg(GFAPGFP)14Mes/J): A Potential Link to Autism Spectrum Disorder
Previous Article in Special Issue
Bisphosphonate-Related Osteonecrosis of the Jaw and Oral Microbiome: Clinical Risk Factors, Pathophysiology and Treatment Options
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Experimental Inoculation of Aggregatibacter actinomycetemcomitans and Streptococcus gordonii and Its Impact on Alveolar Bone Loss and Oral and Gut Microbiomes

by
Catarina Medeiros Rocha
1,2,
Dione Kawamoto
1,
Fernando Henrique Martins
1,2,
Manuela Rocha Bueno
1,2,3,
Karin H. Ishikawa
1,
Ellen Sayuri Ando-Suguimoto
1,
Aline Ramos Carlucci
1,
Leticia Sandoli Arroteia
4,
Renato V. Casarin
4,
Luciana Saraiva
2,*,
Maria Regina Lorenzetti Simionato
1 and
Marcia Pinto Alves Mayer
1,2,*
1
Department of Microbiology, Institute of Biomedical Science, University of São Paulo, São Paulo 05508-000, SP, Brazil
2
Department of Stomatology, School of Dentistry, University of São Paulo, São Paulo 05508-000, SP, Brazil
3
Division of Periodontics, Faculdade São Leopoldo Mandic, São Leopoldo Mandic Research Institute, Campinas 13045-755, SP, Brazil
4
Department of Prosthesis and Periodontology, School of Dentistry, University of Campinas, Campinas 13083-875, SP, Brazil
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(15), 8090; https://doi.org/10.3390/ijms25158090
Submission received: 15 April 2024 / Revised: 10 May 2024 / Accepted: 17 May 2024 / Published: 25 July 2024
(This article belongs to the Special Issue Molecular Advances in Oral Microbiome and Diseases)

Abstract

:
Oral bacteria are implicated not only in oral diseases but also in gut dysbiosis and inflammatory conditions throughout the body. The periodontal pathogen Aggregatibacter actinomycetemcomitans (Aa) often occurs in complex oral biofilms with Streptococcus gordonii (Sg), and this interaction might influence the pathogenic potential of this pathogen. This study aims to assess the impact of oral inoculation with Aa, Sg, and their association (Aa+Sg) on alveolar bone loss, oral microbiome, and their potential effects on intestinal health in a murine model. Sg and/or Aa were orally administered to C57Bl/6 mice, three times per week, for 4 weeks. Aa was also injected into the gingiva three times during the initial experimental week. After 30 days, alveolar bone loss, expression of genes related to inflammation and mucosal permeability in the intestine, serum LPS levels, and the composition of oral and intestinal microbiomes were determined. Alveolar bone resorption was detected in Aa, Sg, and Aa+Sg groups, although Aa bone levels did not differ from that of the SHAM-inoculated group. Il-1β expression was upregulated in the Aa group relative to the other infected groups, while Il-6 expression was downregulated in infected groups. Aa or Sg downregulated the expression of tight junction genes Cldn 1, Cldn 2, Ocdn, and Zo-1 whereas infection with Aa+Sg led to their upregulation, except for Cldn 1. Aa was detected in the oral biofilm of the Aa+Sg group but not in the gut. Infections altered oral and gut microbiomes. The oral biofilm of the Aa group showed increased abundance of Gammaproteobacteria, Enterobacterales, and Alloprevotella, while Sg administration enhanced the abundance of Alloprevotella and Rothia. The gut microbiome of infected groups showed reduced abundance of Erysipelotrichaceae. Infection with Aa or Sg disrupts both oral and gut microbiomes, impacting oral and gut homeostasis. While the combination of Aa with Sg promotes Aa survival in the oral cavity, it mitigates the adverse effects of Aa in the gut, suggesting a beneficial role of Sg associations in gut health.

1. Introduction

Periodontitis involves multifactorial inflammatory processes that result in the destruction of tooth-support tissues, driven by dysbiotic microbiota present in supra- and subgingival biofilms [1]. Specific periodontopathogenic bacteria play a key role in the composition of dental biofilm, leading to heightened proportions of inflammatory organisms and increased virulence potential [2].
Aggregatibacter actinomycetemcomitans (Aa) is a Gram-negative, facultative anaerobic rod primarily associated with rapidly progressing periodontitis in young individuals, previously known as aggressive periodontitis (AP) [3]. This organism is able to subvert host response due to the production of a leukotoxin and a cytolethal distending toxin (CDT) [4]. Leukotoxin deactivates neutrophils, monocytes, and endothelial cells in humans and certain non-human primates but does not affect experimental rodent models [5,6]. AaCDT disrupts the cell cycle of epithelial cells and fibroblasts, induces apoptosis in T lymphocytes, and impairs macrophages’ phagocytic activity [7,8,9]. Additionally AaLPS triggers the release of inflammatory cytokines such as IL-1, IL-6, and TNFα [10,11], while the outer membrane protein OMP29 facilitates invasion of non-phagocytic cells [12] and exerts inhibitory effects on the expression of several inflammatory mediators [13].
Periodontitis, including the rapidly progressing form associated with Aa, is marked not only by oral dysbiosis but also by changes in the fecal microbiome [14,15,16]. Experimental evidence suggests that gut dysbiosis may arise following the ingestion of oral bacteria, resulting in intestinal barrier disruption and systemic inflammation [17,18,19,20,21].
During homeostasis, the integrity of the gut mucosa relies on the expression of transmembrane molecular complexes like tight junction (TJ) proteins and mucus production [22,23]. Alterations in the gut microbiota have been linked to changes in intestinal permeability originated from inflammation-induced damage and shifts in the transcriptional profile of TJ proteins [24]. Prior studies have shown that oral pathogens like P. gingivalis and Aa can compromise the oral mucosa barrier [25,26,27] since P. gingivalis proteases degrade epithelial adhesion molecules [28] whereas AaCDT induces redistribution of E-cadherin and of proteins of the adherens junction complex within the epithelial cells and causes detachment of basal cells from the basement membrane [24].
The virulence and survival of bacteria from multispecies biofilms in the oral cavity are influenced by host factors and neighboring microorganisms. Periodontitis arises from the synergistic effect of the entire microbiota and a disease-associated bacterial consortium including Aa and Streptococcus gordonii (Sg) was reported in aggressive periodontitis [29]. The interaction between Aa and H2O2-producing streptococci results in altered virulence and increased survival of Aa, as demonstrated by in vitro biofilm studies and in a murine abscess model [30,31,32]. However, the potential of the Aa–streptococci consortium to induce experimental periodontitis and its role in in promoting microbial dysbiosis in the oral cavity and gut have not been evaluated.
Therefore, we aimed to assess the impact of the oral administration of Aa, Sg, and a consortium of Aa and Sg on alveolar bone resorption, expression of genes related to gut mucosa integrity, and oral and gut microbiomes, in a murine experimental model.

2. Results

2.1. Side Effects and Weight Gain Induced by Different Treatments

Four groups were evaluated, Aa-, Sg-, and Aa+Sg-infected groups along with the SHAM-inoculated group (negative control) (Figure 1). No changes in skin, fur, behavior, or mobility were observed in the animals throughout the 30-day experimental period. Initial weight was similar among groups. However, infection with Aa seemed to pose a detrimental effect on mice, since the Aa = infected groups (Aa and Aa+Sg) showed lower final weights compared to the Sg group (Figure 2A).

2.2. Alveolar Bone Loss Computed Microtomography (micro-CT)

Analysis of bone volume and porosity in the interproximal region between the first and second molars in the left hemimaxillae revealed that infection with Sg resulted in a lower percentage of bone volume and higher bone porosity compared to the other groups. However, alveolar bone loss values for the Sg group were similar to those observed for the Aa+Sg group. Infection with Aa alone did not significantly alter bone volume and porosity compared to the SHAM group, although the Aa group exhibited similar alveolar bone loss to the Aa+Sg group (Figure 2B,C).

2.3. LPS Levels in Serum

Serum LPS levels did not vary significantly among the groups, although the mean values of endotoxin units (EUs) were higher in the Aa-infected groups (Aa+Sg and Aa) compared to the Sg and SHAM groups.

2.4. Gene Expression Related to Gut Inflammation and Barrier Permeability

Aa infection resulted in higher expression of Il-1β compared to infection with Sg or Aa+Sg. Expression of Il-6 was downregulated in the Aa- and Sg-infected groups but not in the Aa+Sg group compared to SHAM (Figure 3A,B). There were no differences in the transcription levels of Il-10 and Tnfα among the groups.
Infection with Aa and/or Sg induced changes in the transcriptional profile of genes encoding TJ proteins claudin 1, claudin 2, and occludin and the anchoring protein zonulin in the intestinal barrier. Overall, infection with Aa or Sg downregulated the transcription of these genes, whereas infection with both organisms in the Aa+Sg group increased their mRNA levels, similar to the levels of the SHAM group, except for Cldn1, which achieved lower mRNA levels in Aa and Aa+Sg groups (Figure 3C–F). Transcription of mucin encoding genes was anot altered by the treatments.

2.5. Oral and Gut Bacterial Community Structure

The oral biofilm and gut samples underwent 16S rRNA sequencing analysis. DNA was successfully extracted from oral and gut samples of 24 animals (6 per group). However, two gut samples from the SHAM group failed to yield amplicons for analysis, resulting in microbiome data of only four SHAM samples. A total of 7,207,606 sequences were obtained (ranging from 97,250 to 246,890). After filtering and removing chimeras, 2,719,335 sequences remained for oral samples (ranging from 87,139 to 150,712) and 390,140 for gut samples (ranging from 4854 to 32,775). Rarefaction curves based on the number of operational taxonomic units (OTUs) indicated that all samples reached a plateau when oral biofilm samples were normalized to 10,000 amplicon sequence variants (ASVs), and gut samples were normalized to 4000 sequences.
Faith’s phylogenetic diversity index analysis revealed that inoculation with Sg increased the diversity of the oral microbiome compared to Aa [33], although they did not differ from SHAM. In contrast, the concomitant administration of Aa+Sg reversed this effect (Figure 4A). Pielou (evenness) and Shannon (richness) diversity index analyses for the oral microbiome did not result in significant differences among the groups.
Analysis of the gut microbiome revealed differences in evenness index (Pielou) among the three infected groups [34] but not from the SHAM-inoculated group (Figure 4B). Inoculation with Aa or Sg resulted in reduced diversity compared to inoculation with both organisms in the Aa+Sg group. There were no differences in Faith and Shannon indexes among the groups for the gut microbiome.
Beta diversity analysis based on weighted and unweighted UniFrac distances revealed that the treatments affected both the oral and gut microbiomes. Infection with Aa or Sg altered the structure of the oral microbiome, while inoculation with Aa+Sg did not result in significant changes compared to SHAM (Figure 5A(I,II)). Analysis of gut samples showed clear segregation among the infected groups and SHAM. However, the gut microbiomes of Aa, Sg, and Aa+Sg did not differ from each other, except for differences between Aa+Sg and Sg based on unweighted UniFrac distances (Figure 5B(I,II)).
There were no differences in the abundance of different phyla in the oral or gut microbiomes among the groups (Figure 6 and Figure 7, respectively). Firmicutes was the most abundant phylum in the oral microbiome, followed by Pseudomonadota. ANCOM revealed that infection with Aa led to an increased abundance of Gammaproteobacteria, Enterobacterales, and the genus Alloprevotella, while infection with Sg resulted in increased abundance of the genera Alloprevotella and Rothia in the oral cavity. Infection with both organisms did not lead to significant changes in the oral microbiome (Figure 6).
Firmicutes was the most abundant phylum in the gut microbiome (Figure 7). Inoculation of Aa, Sg, or both microorganisms in the oral cavity resulted in decreased abundance of the order Erysipelotrichales and the family Erysipelotrichaceae compared to SHAM in the gut.
eHOMD database analysis revealed that Aa was detected in three out of six oral biofilm samples from the Aa+Sg group, but not in the Aa group, at an abundance lower than 0.001. Sg was not detected in the oral cavity of any mice. None of the inoculated species was detected in the gut.

3. Discussion

Periodontitis is associated with a dysbiotic microbiome. The microbial community within any host is influenced not only by the neighboring microorganisms but also by host defense, environmental, genetic, and epigenetic factors [35]. However, not all these factors can be replicated in a murine model. Additionally, animal models for studying periodontitis are limited by the specificity of certain organisms and their virulence factors to humans.
Therefore, our findings should be interpreted within the constraints of the animal model utilized. Colonization represents a crucial phase in infectious diseases, yet Aa oral colonization in mice is hindered due to the inability of Aa adhesins Aae and OMP100 to bind to murine epithelial cells [36,37]. In addition, Aa leukotoxin targets only human and certain non-human primate monocytes, macrophages, and neutrophils [38]. It is also important to notice that the resident microbiota of rodents can be easily influenced by mouse lineage but also by various factors, including breeding sources and conditions [39], altering the results of assays performed at different animal facilities.
The mice infected with Aa present a lower weight at the end of the experimental period when compared with those infected with Sg or SHAM-inoculated mice. This observation differs from studies in humans where detection of Aa in the oral microbiome was associated with obesity [40]. Experimental administration of Aa in mice fed a high-fat diet also resulted in increased weight gain compared to non-infected mice, whereas infection had no effect in weight gain in animals under a normal chow diet [21]. However, any comparison between our data and the latter study is prevented by different tested protocols, especially by the use of norfloxacin, an antimicrobial that modulates the gut microbiota and leads to improved fasting glycemia and oral glucose tolerance [41].
Despite these limitations, our data revealed that the interaction with Sg increased the ability of Aa to colonize the oral biofilm of mice, since half of the animals of the Aa+Sg group harbored Aa in their oral cavity after 30 days of the experimental period, whereas this species was not detected in any mouse of the Aa group. Sg increases Aa fitness by providing lactate for Aa and producing H2O2, enhancing oxygen bioavailability which alters the metabolism from fermentation to respiration, changes the transcription profile of the pathogen [6,32], and promotes its virulence in an abscess model [31,42]. Moreover, Aa dispersal in biofilms is dependent on dispersin B, whose expression is upregulated in co-infection with Sg, as shown by in vitro data [31].
Our data evidenced the relevance of the interaction with Sg to increase Aa survival in the oral biofilm of rodents. A single study in rats reported that the oral inoculation of Aa resulted in successful colonization of the oral biofilm and induction of periodontitis in wild type rodents [43]. Therefore, most studies on experimental alveolar bone loss in rodents induced by Aa inoculated the living organism or its products in the gingival tissue [44,45,46,47,48], resulting in Aa persistent colonization of the gingival tissues but not of the oral biofilm [46]. This association of Aa with lactate producers in the oral cavity is not only relevant for the murine model but has already been reported in experimental infection of Aa in primates. However, Aa recovery was low even in these primates after 4 weeks [49].
We have also shown that the three infected groups exhibited alveolar bone resorption, notwithstanding the similar bone volume and porosity observed in the Aa group compared to SHAM and to Aa+Sg. Therefore, although the lactate/peroxide producer streptococci provided increased survival of Aa in the oral biofilm, this association did not enhance the destruction of the periodontal tissues. It is worth mentioning that the bottom of deep periodontal pockets in humans represents a highly anaerobic environment where peroxide production is limited by the low concentration of oxygen, and Aa metabolism is anaerobic [32]. Furthermore, microbiome data indicated that the increased abundance of Aa in deep periodontal pockets of periodontitis patients is accompanied by reduced levels of streptococci and other lactate producers [16].
The oral administration of Sg also resulted in alveolar bone loss, either independently or in conjunction with Aa in the murine model. Sg is considered an accessory pathogen in the oral cavity due to its interactions with pathogens [50], despite its relatively low prevalence compared to other H2O2 producers streptococci in the oral cavity [51]. This species can activate osteoclasts [52] and induce an inflammatory response in experimental apical periodontitis [53], stimulating the production of chemokines such as CXCL8 by periodontal ligament fibroblasts [54] as well as the maturation of dendritic cells [55]. Thus, under the limitations of the murine model, it seems that Sg may be important for Aa survival in the supragingival biofilm, but the co-infection may not be relevant for Aa-induced bone resorption.
Although Sg facilitated the recovery of Aa in the oral cavity, the impact of oral administration of Aa in the gut was more pronounced compared to that promoted by the Aa+Sg consortium. Limited knowledge exists regarding the role of Aa and/or Sg as inducers of intestinal dysbiosis and alteration of the intestinal barrier. Aa infection induced the expression of Il-1β in the gut, while Sg or the Aa+Sg consortium did not. Il-1β is commonly associated with heightened intestinal permeability [56]. Conversely, Sg mitigated the Aa-induced inflammatory response in the gut, evidenced by the low transcriptional levels of Il-1β in the Aa+Sg group. Previous data have indicated that H2O2 produced by streptococci activates the Nrf2 pathway in macrophages, inhibiting the NFκB transcription factor and reducing the production of proinflammatory cytokines [57]. Other factors may also have contributed to this disparity. Aa mono-infection, but not the dual infection with Aa and Sg, resulted in the expression of virulence factors such as the cytolethal distending toxin (CDT) [32], which is known to activate the inflammasome leading to IL-1β release [58]. Furthermore, H2O2 acts as a signal to trigger the expression of catalase and OMP100 (ApiA) by Aa, which are involved in defense against the host’s innate immune system [59]. In contrast, the IL-1β pathway may hinder Aa survival in the gut [60].
We have also demonstrated downregulation of Il-6 in the Aa, Sg, and Aa+Sg groups compared to the SHAM group. IL-6 plays a role in leukocyte recruitment during the acute inflammatory response by selectively regulating inflammatory chemokines and apoptotic events [17,61,62]. Therefore, it appears that infection with Aa and Sg contributed to the control of the inflammatory process, likely facilitated by the release of H2O2 by the accessory pathogen Sg [50], although other factors, such as additional regulatory proteins such as Aa outer membrane protein OMP29, may also play a role [13].
It is well recognized that dysregulation of the mucosal barrier facilitates the translocation of bacterial components or living cells [56] from the intestinal lumen through the epithelium, triggering immune activation and leading to systemic inflammation [63,64]. Alterations in the expression of TJ proteins can compromise the integrity of the barrier, promoting increased intestinal permeability, thus resulting in a condition often referred to as “leaky gut” [63,65,66].
Oral infection with Aa and/or Sg resulted in altered transcription of several genes involved in gut barrier integrity [67]. Generally, Aa or Sg downregulated the expression of Cldn 1 and Cldn 2, Ocdn, and Zo-1, whereas the Aa+Sg consortium upregulated the expression of Cldn 2, Ocdn, and Zo-1. The elevated levels of Il-1β transcripts and low levels of Il-6 transcripts in the gut of Aa-infected mice may partially account for this regulation induced by the pathogen [68]. Microbial insults and the resulting inflammatory response are known to contribute to the regulation of TJ expression and localization. Expression of Claudin 1 diminishes upon chronic exposure to LPS challenge [69], whilst Claudin 2 expression is associated with protection of microbial-induced colitis [38]. IL-6 upregulates expression of Claudin 2, while TNF and IL-1β indirectly trigger occludin removal from the tight junction and increase leaky pathway permeability. On the other hand, IL-10 and TGFβ promote epithelial proliferation and restitution, leading to barrier restoration [70]. Thus, the attenuation of Il-1β and Il-6 regulation observed when Aa was combined with Sg may contribute to the enhanced expression of TJs in the gut barrier. Previous data reported that Sg exerts a beneficial effect on the epithelial barrier function, particularly in gingival cells, by inhibiting cytokine secretion and enhancing the expression of key tight junction components, thus reinforcing the oral barrier against microbial invasion [71]. However, it should be noticed that the changes in TJ transcription profiles induced by Aa and/or Sg were discreet, since we could not detect endotoxemia in any of the studied groups, an indicator of altered gut permeability [72].
Oral inoculation of Aa, Sg, or Aa+Sg led to alterations in both oral and gut microbiomes. The oral microbiota plays a critical role in maintaining oral health and can influence intestinal dysbiosis. In periodontitis, dysbiosis has been observed not only in the oral cavity but also in the intestine [14,15,16]. The mechanism behind this phenomenon involves the ingestion of oral bacteria, which may persist in the gut for at least 24 h [73], potentially inducing dysbiosis [74].
Our data indicate that oral administration of Sg increased the diversity of the oral microbiome (Faith phylogenetic diversity index), whereas Aa or the consortium Aa+Sg reduced the diversity when compared to mono-infection with Sg. Phylogenetic diversity accounts for the phylogenetic relatedness of the community members [33] and reduced diversity induced by Aa means that this organism puts pressure on the oral microbiome in order to select a subset of organisms. These findings are consistent with studies showing a decrease in the microbial diversity of the oral cavity in periodontitis patients [75,76], indicating a role for Aa in oral dysbiosis.
The population structure of the oral microbiome differed between the Aa-infected group and the Sg-infected group compared with the SHAM group, with all infected groups differing from each other. The oral microbiome of Aa-infected mice exhibited increased abundance of Pseudomonadota, class Gammaproteobacteria, order Enterobacterales, and of the genus Alloprevotella. Sg administration also increased the abundance of Alloprevotella but additionally led to higher levels of Rothia. The implications of these microbial shifts are not fully understood, as members of Enterobacterales and the family Prevotellaceae comprise species associated with both disease and health [77,78]. However, Enterobacterales has previously been associated with Crohn’s disease [79,80,81,82], whereas Rothia is typically associated with health [83].
Animals infected with Aa, Sg, or both organisms exhibited a reduced abundance of Erysipelotrichaceae in the gut, reaching non-detectable levels. Erysipelotrichaceae is considered a beneficial organism to the gut (97) and previous data have shown that Aa oral inoculation resulted in lower abundance of Turicibacter, a genus of the family Erysipelotrichaceae, in feces [21]. Oral administration of streptococci in mice, such as S. mitis or S. salivarius, also reduced the levels of Turicibacter in stool samples [84]. The beneficial role of Erysipelotrichaceae in the gut appears to be due to the production of butyric acid, a fatty acid that provides energy to colonocytes and regulates the immune response [85]. Thus, our data suggest that infection with Aa, Sg, or both organisms results in depletion of beneficial organisms, in agreement with reduced transcription of genes associated with gut permeability.
Periodontitis has been linked to chronic inflammatory bowel disease (IBD) and colon cancer [86]. It is noteworthy that gut dysbiosis in patients with periodontitis, particularly the molar–incisor pattern associated with high abundance of Aa, was characterized by a greater abundance of sulfate-reducing bacteria (SRB), which are potential risk factors for colon cancer [16,87]. Aa can be found in low abundance in healthy individuals [88], but the oral microbiota in health harbors a higher proportion of H2O2 producers, such as S. sanguinis and S. gordonii, compared to disease [51]. Thus, the low transcriptional levels of Il-1β in the gut of mice of the Aa+Sg group compared to the Aa group suggest that these streptococci may limit the inflammatory potential of Aa in the gut. On the other hand, damage not only to the periodontium but also to the gut barrier appears to increase when the abundance of Aa rises due to host and bacterial factors, concurrent with decreased levels of lactate/H2O2 producers [89].
Taken altogether, our data indicate that Aa can disrupt both oral and gut microbiomes. The pathogen may disturb gut homeostasis by inducing the expression of inflammatory mediators such as IL-1β and by altering the expression profile of proteins related to intestinal barrier integrity. Aa co-exists in the oral cavity with other species such as Sg, which enhances its survival in the oral biofilm. However, co-infection of Aa with Sg seems to mitigate the effects of Aa on the gut.

4. Materials and Methods

4.1. Ethical Considerations

This study was conducted after obtaining approval from the Institutional Animal Care and Use Committee (ICB USP approval number: 4828281020), in accordance with the ethical principles of animal experimentation outlined by the Brazilian College of Animal Experimentation (COBEA).

4.2. Animals and Maintenance Conditions

C57BL/6 specific pathogen-free (SPF) male mice, aged 6–8 weeks, were obtained from the Animal Facility at the Faculty of Medicine and subsequently transferred to the Department of Microbiology and Parasitology at the Institute of Biomedical Sciences, University of São Paulo. They were housed in cages with a maximum of four mice per cage, fitted with microisolator filters, and placed on ventilated shelves to maintain pathogen-free conditions, with food (chow diet) provided ad libitum.

4.3. Sample Calculation and Group Allocation

Sample calculation was conducted for alveolar bone loss as the outcome, with a significance level (alpha) of 5%, power of 80%, and considering a 10% loss of animals during the experiment, based on our previous pilot data [90,91]. After a 7-day acclimation period in the facility, forty-eight (48) animals were randomly assigned to four groups, with twelve animals per group: SHAM, infected with Aa, infected with Sg, and infected with both species (Aa+Sg). The animals were then monitored for an experimental period of 30 days. Animals showing abnormalities in growth, weight, or physical characteristics at baseline were excluded from the study. Throughout the experiment, animals were monitored for changes in weight, mobility, coat condition, and presence of skin lesions.

4.4. Blinding

Each animal was assigned a temporary random number within its respective group. Cages were numbered based on their placement on the rack. A cage was randomly selected from each group from the available pool. Blinding procedures were maintained throughout the allocation process, evaluation of results, and data analysis. However, blinding was not possible during the experiment due to the same researcher being involved in both the preparation and inoculation of organisms. Additionally, the bacterial suspensions differed in color from the vehicles used.

4.5. Strains and Culture Conditions

Aa JP2 [38] and Sg DL1 [92] strains were utilized. All strains were maintained in 20% glycerol in brain–heart broth at −80 °C.
Aa was cultured in tryptone soy broth (Difco Laboratories, Detroit, MI, USA) supplemented with 0.5% yeast extract (Difco Laboratories) while Sg was cultured in BHI broth (Difco Laboratories), both incubated at 37 °C, in 5–10% CO2. For the assays, liquid medium was utilized to grow bacteria to mid-log phase. Subsequently, the cultures were centrifuged (355× g for 5 min), and the cell pellets were suspended in phosphate-buffered saline solution (1× PBS) to an optical density at 590nm of approximately 1, corresponding to 1 × 109 CFU Aa/mL and to 1 × 1012 CFU Sg/mL.
The cell suspensions were centrifuged (6000× g) and cells suspended in a solution containing 10% skim milk powder (Molico, Nestle, Brazil), 5% sodium L-glutamate monohydrate (Sigma-Aldrich, Darmstadt, Germany), and 5% dithiothreitol (Sigma-Aldrich). The suspension was lyophilized using a freeze-dryer (Freezone Triad Freezer Dryers, Labconco, Flawil, Switzerland) at −4 °C under vacuum.
The lyophilized material was stored in a freezer at −80 °C. The viability of each batch of lyophilized bacteria was assessed by culturing them on agar plates under suitable conditions. Before each day of inoculation, the samples were reconstituted in BHI broth for one hour. Subsequently, the lyophilized bacteria were suspended in PBS at standardized concentrations.

4.6. Experimental Infections

Fifty-microliter aliquots containing 1 × 109 CFU of Aa and/or 1 × 109 CFU of Sg in PBS with 1.5% carboxymethylcellulose were inoculated into the oral cavity of mice using a gavage needle, three times a week, for four weeks, totalizing 12 inoculations [47]. On the first, third, and fifth days of the experimental period, the animals underwent intraperitoneal anesthesia with ketamine (100 mg/kg) (10% Dopalen, Vetbrands Laboratory, Rio de Janeiro, Brazil) and xylazine (10 mg/kg) (2% Ansedan, Vertbrands Laboratory), when they received an injection at the interproximal gingiva between the first and second molars in the left hemimaxilla of a volume of 10 µL containing 1 × 107 CFU of Aa in PBS [48]. The vehicle (PBS with 1.5% carboxymethylcellulose) was orally administered to the SHAM-inoculated group. Both the SHAM and Sg groups were anesthetized and received gingival injections with PBS on the same days and with the same volumes as the other Aa infected groups. After 30 days, the mice were euthanized, and samples were collected. The study design is illustrated in Figure 1.

4.7. Euthanasia and Sample Collection

The animals were anesthetized intraperitoneally with ketamine (100 mg/kg) and xylazine (10 mg/kg). Blood was collected via cardiac puncture, and the mice were euthanized by exsanguination. Serum was obtained by centrifugation. The left hemimaxillae were transferred to formaldehyde solution and kept at room temperature for 24 h, then transferred to PBS and stored at 4 °C until analysis. The jejunal portions of the intestine were placed in RNAlater TM Stabilization Solution (Invitrogen by Thermo Fisher Scientific, Vilnius, Lithuania) and stored at −80 °C. Oral biofilms were collected using microbrushes, and jejunal content samples were obtained using a wooden spatula and stored in Tris-EDTA buffer, pH 7.4.

4.8. Computerized Microtomography (Micro CT)

Alveolar bone resorption was assessed using microtomography (SkyScan 1174 version 1.1, Kontich, Belgium) operating at 45 kV voltage, 550 μA current, with an 8.71 μm pixel size and a 0.2 mm aluminum filter. Scans were conducted on the left hemimaxillae, and a blinded evaluator identified a standardized area of 60 × 30 pixels (ROI) at the interproximal region between the first and second molars, originating from the cementoenamel junction of the second molar across 15 coronal sections. Bone volume and porosity percentages were determined through image analysis using CT Analyzer software version 1.15.4.0.

4.9. Serum Lipopolysaccharide Levels

Lipopolysaccharide (LPS) levels were quantified using the Pierce LAL Chromogenic Endotoxin Quantification Kit #88282 (Thermo Scientific™, Rockford, IL, USA). Solutions ranging from 0.1 to 1 endotoxin units (EUs) per milliliter of LPS Endotoxin Standard (E. coli 011: B4) were employed to determine the endotoxin concentration of each sample. Absorbance was measured at 405 nm using the Epoch™ 2 microplate spectrophotometer (BioTek, Winooski, VT, USA).

4.10. Gene Expression Linked to Gut Inflammation and Epithelial Barrier Permeability

Gut samples underwent RNA extraction using Trizol LS Reagent (Invitrogen Life Technologies, Carlsbad, CA, USA) with the assistance of a Mini-BeadBeater (BioSpec 3110BX Mini-BeadBeater-1 High Energy Cell Disrupter, Campinas, São Paulo, Brazil) for 20 s, repeated twice. The resulting RNA was treated with desoxyribonuclease (Ambion™ Dnase I, Invitrogen Life Technologies). Subsequently, cDNA was synthesized using the SuperScript TM Vilo TM Synthesis Kit for RT-PCR (Invitrogen Life Technologies). Quantitative PCR was conducted using the StepOne Plus System thermocycler (Applied Biosystems, Foster City, CA, USA).
Transcription levels of cytokine genes were determined using 100 ng of cDNA and TaqMan™ Gene Expression Assay (Invitrogen by Thermo Fisher Scientific, Vilnius, Lithuania) with commercial TaqMan primers and probes (Invitrogen Life Technologies) for IL-1β (Mm00434228_m1), TNFα (Mm00443258_m1), IL-10 (Mm01288386_m1), IL-6 (Mm00446190_m1), and GAPDH (Mm99999915_g1).
The expression of genes related to intestinal epithelial barrier permeability including Claudin 1 (Cldn1), Claudin 2 (Cldn2), Occludin (Ocdn), Mucin 1 (Muc1), Mucin 2 (Muc2), and Zonulin (Zo-1) was determined using primers synthesized by Thermo Fisher Scientific (Waltham, MA, USA) (Table 1). Each reaction consisted of 5 μL of SYBR Green (Thermo Fisher Scientific), 200 ng of cDNA, and 0.625 pM of each oligonucleotide in a final volume of 10 μL. Reactions were performed through forty cycles of 95 °C/15 sec, 65 °C/1 min, 81 °C/10 s, followed by two steps at 95 °C/15 s and 65 °C/1 min, and a final step at 0.5–95 °C/15 s.
The relative expression of target genes was calculated using the ΔΔCT method, utilizing GAPDH as an endogenous control [39] and expressed as fold changes relative to the control group (SHAM).

4.11. Oral and Gut Microbiomes

DNA from oral biofilm and gut samples was extracted using the Master PureTM DNA Purification Kit (Epicentre® Illumina Company, Madison, WI, USA). The quality of purified DNA was assessed using the Qubit 2.0 fluorometer (Thermo-Fisher Scientific, Carlsbad, CA, USA). Unfortunately, DNA from the biofilms of two animals from each group was lost during manipulation, leading to the exclusion of gut samples from these animals, leading to microbiome analyses of six samples from each site per group.
A barcoded primer set, based on universal primers Bakt_341F (CCTACGGGNGGCWGCAG) and Bakt_805R (GACTACHVGGGTATCTAATCC) [98], was utilized to amplify the hypervariable V3–V4 region of the 16S rRNA gene. Subsequently, DNA samples were sequenced by ByMyCell (Ribeirão Preto, São Paulo, Brazil) using the Illumina MiSeq 2 × 250 platform. The sequence data were then submitted to the Sequence Read Archive (SRA) under BioProject identification numbers PRJNA995344 and PRJNA994574.
Data were analyzed using Qiime 2 2022.8 software [99]. Demultiplexed sequences and reads underwent filtering using Dada 2, with a quality threshold set at 25. Subsequently, trimmed sequences were clustered into amplicon sequence variants (ASVs), and taxonomy was assigned using the Silva138 database [100,101]. Alpha diversity indices, including Faith, Pielou, and Shannon, were assessed. Beta diversity analyses were conducted using weighted and unweighted UniFrac distance matrices and were visualized through principal coordinate analysis (PCA) [99]. Significant differences among groups were determined by PERMANOVA with 999 permutations. The discrepancies in abundances of ASVs among groups were analyzed using ANCOM [102] with a 75% empirical cutoff value for the w statistic [103]. To identify the inoculated species in the oral and gut microbiome, the Expanded Human Oral Microbiome Database (eHOMD) was utilized [104].

4.12. Data Analyses

The data were assessed for normality using the Kolmogorov–Smirnov statistical test with Lilliefors correction, and the homogeneity of variances was evaluated using the F test. For standard data analysis, the Kruskal–Wallis test followed by Dunn’s post hoc test was utilized. Statistical significance was set at p < 0.05. All analyses were conducted using GraphPad Prism® version 6.0 software (GraphPad Software, La Jolla, CA, USA).

Author Contributions

Conceptualization, M.P.A.M. and C.M.R.; methodology, C.M.R. and F.H.M.; validation, E.S.A.-S.; formal analysis, D.K., C.M.R. and F.H.M.; investigation C.M.R., F.H.M., M.R.B. and A.R.C. (animal experiment, sample collection, and gene expression), K.H.I. (animal experiment, sample collection, and microbiome analyses), D.K. (DNA extraction and microbiome analyses), L.S.A., R.V.C. and F.H.M. (alveolar bone loss data); data curation, E.S.A.-S.; writing—original draft preparation, M.P.A.M., C.M.R. and M.R.L.S.; writing—review and editing, M.P.A.M., C.M.R. and L.S.; visualization, F.H.M., A.R.C. and D.K.; supervision, M.P.A.M. and L.S.; funding acquisition, M.P.A.M. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by The São Paulo Research Foundation, FAPESP, Grant# 2015/18273-9 and Scholarships from FAPESP # 21/13846-1 (A.R.C.), 17/16377-7 (M.R.B.), 16/13159-6 (D.K), 16/13156-7 (K.H.I.), and from Coordenação de Aperfeiçoamento de Pessoal de Nível Superior—Brasil (CAPES)—Finance Code 001 (C.M.R., F.H.M., and E.S.A.-S).

Institutional Review Board Statement

The animal study protocol was approved by the Ethics Committee on the use of animals of the Institution of Biomedical Sciences (University of São Paulo) (CEUA-ICB/USP) (protocol code 4828281020 and date of approval 05/13/2021).

Data Availability Statement

Sequence data were submitted to Sequence Read Archive (SRA) under BioProject identification numbers PRJNA995344 and PRJNA994574.

Conflicts of Interest

The authors declare no conflicts of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Papapanou, P.N.; Sanz, M.; Buduneli, N.; Dietrich, T.; Feres, M.; Fine, D.H.; Flemmig, T.F.; Garcia, R.; Giannobile, W.V.; Graziani, F.; et al. Periodontitis: Consensus report of workgroup 2 of the 2017 World Workshop on the Classification of Periodontal and Peri-Implant Diseases and Conditions. J. Periodontol. 2018, 89 (Suppl. S1), S173–S182. [Google Scholar] [CrossRef] [PubMed]
  2. Lamont, R.J.; Hajishengallis, G. Polymicrobial synergy and dysbiosis in inflammatory disease. Trends Mol. Med. 2015, 21, 172–183. [Google Scholar] [CrossRef] [PubMed]
  3. Fine, D.H.; Patil, A.G.; Loos, B.G. Classification and diagnosis of aggressive periodontitis. J. Clin. Periodontol. 2018, 45 (Suppl. S20), S95–S111. [Google Scholar] [CrossRef] [PubMed]
  4. Herbert, B.A.; Novince, C.M.; Kirkwood, K.L. Aggregatibacter actinomycetemcomitans, a potent immunoregulator of the periodontal host defense system and alveolar bone homeostasis. Mol. Oral. Microbiol. 2016, 31, 207–227. [Google Scholar] [CrossRef] [PubMed]
  5. Dietmann, A.; Millonig, A.; Combes, V.; Couraud, P.O.; Kachlany, S.C.; Grau, G.E. Effects of Aggregatibacter actinomycetemcomitans leukotoxin on endothelial cells. Microb. Pathog. 2013, 61–62, 43–50. [Google Scholar] [CrossRef] [PubMed]
  6. Fine, D.H.; Armitage, G.C.; Genco, R.J.; Griffen, A.L.; Diehl, S.R. Unique etiologic, demographic, and pathologic characteristics of localized aggressive periodontitis support classification as a distinct subcategory of periodontitis. J. Am. Dent. Assoc. 2019, 150, 922–931. [Google Scholar] [CrossRef] [PubMed]
  7. Mayer, M.P.; Bueno, L.C.; Hansen, E.J.; DiRienzo, J.M. Identification of a cytolethal distending toxin gene locus and features of a virulence-associated region in Actinobacillus actinomycetemcomitans. Infect. Immun. 1999, 67, 1227–1237. [Google Scholar] [CrossRef] [PubMed]
  8. Shenker, B.J.; Boesze-Battaglia, K.; Zekavat, A.; Walker, L.; Besack, D.; Ali, H. Inhibition of mast cell degranulation by a chimeric toxin containing a novel phosphatidylinositol-3,4,5-triphosphate phosphatase. Mol. Immunol. 2010, 48, 203–210. [Google Scholar] [CrossRef] [PubMed]
  9. Ando-Suguimoto, E.S.; Benakanakere, M.R.; Mayer, M.P.A.; Kinane, D.F. Distinct Signaling Pathways Between Human Macrophages and Primary Gingival Epithelial Cells by Aggregatibacter actinomycetemcomitans. Pathogens 2020, 9, 248. [Google Scholar] [CrossRef] [PubMed]
  10. Lindemann, R.A. Bacterial activation of human natural killer cells: Role of cell surface lipopolysaccharide. Infect. Immun. 1988, 56, 1301–1308. [Google Scholar] [CrossRef] [PubMed]
  11. Lindemann, R.A. Roles of interferon and cellular adhesion molecules in bacterial activation of human natural killer cells. Infect. Immun. 1989, 57, 1702–1706. [Google Scholar] [CrossRef] [PubMed]
  12. Kajiya, M.; Komatsuzawa, H.; Papantonakis, A.; Seki, M.; Makihira, S.; Ouhara, K.; Kusumoto, Y.; Murakami, S.; Taubman, M.A.; Kawai, T. Aggregatibacter actinomycetemcomitans Omp29 is associated with bacterial entry to gingival epithelial cells by F-actin rearrangement. PLoS ONE 2011, 6, e18287. [Google Scholar] [CrossRef] [PubMed]
  13. Da Silva, M.P.; Silva, V.O.; Pasetto, S.; Ando-Suguimoto, E.S.; Kawamoto, D.; Mata, G.; Murata, R.M.; Mayer, M.P.A.; Chen, C. Aggregatibacter actinomycetemcomitans Outer Membrane Proteins 29 and 29 Paralogue Induce Evasion of Immune Response. Front. Oral Health 2022, 3, 835902. [Google Scholar] [CrossRef] [PubMed]
  14. Lourenvarsigmao, T.G.B.; Spencer, S.J.; Alm, E.J.; Colombo, A.P.V. Defining the gut microbiota in individuals with periodontal diseases: An exploratory study. J. Oral Microbiol. 2018, 10, 1487741. [Google Scholar] [CrossRef]
  15. Kawamoto, D.; Borges, R.; Ribeiro, R.A.; de Souza, R.F.; Amado, P.P.P.; Saraiva, L.; Horliana, A.; Faveri, M.; Mayer, M.P.A. Oral Dysbiosis in Severe Forms of Periodontitis Is Associated With Gut Dysbiosis and Correlated With Salivary Inflammatory Mediators: A Preliminary Study. Front. Oral Health 2021, 2, 722495. [Google Scholar] [CrossRef] [PubMed]
  16. Amado, P.P.P.; Kawamoto, D.; Albuquerque-Souza, E.; Franco, D.C.; Saraiva, L.; Casarin, R.C.V.; Horliana, A.; Mayer, M.P.A. Oral and Fecal Microbiome in Molar-Incisor Pattern Periodontitis. Front. Cell Infect. Microbiol. 2020, 10, 583761. [Google Scholar] [CrossRef] [PubMed]
  17. Hasturk, H.; Jones, V.L.; Andry, C.; Kantarci, A. 1-Tetradecanol complex reduces progression of Porphyromonas gingivalis-induced experimental periodontitis in rabbits. J. Periodontol. 2007, 78, 924–932. [Google Scholar] [CrossRef] [PubMed]
  18. Hajishengallis, G.; Liang, S.; Payne, M.A.; Hashim, A.; Jotwani, R.; Eskan, M.A.; McIntosh, M.L.; Alsam, A.; Kirkwood, K.L.; Lambris, J.D.; et al. Low-abundance biofilm species orchestrates inflammatory periodontal disease through the commensal microbiota and complement. Cell Host Microbe 2011, 10, 497–506. [Google Scholar] [CrossRef] [PubMed]
  19. Arimatsu, K.; Yamada, H.; Miyazawa, H.; Minagawa, T.; Nakajima, M.; Ryder, M.I.; Gotoh, K.; Motooka, D.; Nakamura, S.; Iida, T.; et al. Oral pathobiont induces systemic inflammation and metabolic changes associated with alteration of gut microbiota. Sci. Rep. 2014, 4, 4828. [Google Scholar] [CrossRef] [PubMed]
  20. Nakajima, M.; Arimatsu, K.; Kato, T.; Matsuda, Y.; Minagawa, T.; Takahashi, N.; Ohno, H.; Yamazaki, K. Oral Administration of P. gingivalis Induces Dysbiosis of Gut Microbiota and Impaired Barrier Function Leading to Dissemination of Enterobacteria to the Liver. PLoS ONE 2015, 10, e0134234. [Google Scholar] [CrossRef] [PubMed]
  21. Komazaki, R.; Katagiri, S.; Takahashi, H.; Maekawa, S.; Shiba, T.; Takeuchi, Y.; Kitajima, Y.; Ohtsu, A.; Udagawa, S.; Sasaki, N.; et al. Periodontal pathogenic bacteria, Aggregatibacter actinomycetemcomitans affect non-alcoholic fatty liver disease by altering gut microbiota and glucose metabolism. Sci. Rep. 2017, 7, 13950. [Google Scholar] [CrossRef] [PubMed]
  22. Anderson, J.M. Molecular structure of tight junctions and their role in epithelial transport. News Physiol. Sci. 2001, 16, 126–130. [Google Scholar] [CrossRef] [PubMed]
  23. Tsukita, S.; Furuse, M.; Itoh, M. Multifunctional strands in tight junctions. Nat. Rev. Mol. Cell Biol. 2001, 2, 285–293. [Google Scholar] [CrossRef] [PubMed]
  24. Martinez-Oca, P.; Robles-Vera, I.; Sanchez-Roncero, A.; Escriva, F.; Perez-Vizcaino, F.; Duarte, J.; Alvarez, C.; Fernandez-Millan, E. Gut dysbiosis and altered barrier function precedes the appearance of metabolic syndrome in a rat model of nutrient-induced catch-up growth. J. Nutr. Biochem. 2020, 81, 108383. [Google Scholar] [CrossRef] [PubMed]
  25. Guo, W.; Wang, P.; Liu, Z.H.; Ye, P. Analysis of differential expression of tight junction proteins in cultured oral epithelial cells altered by Porphyromonas gingivalis, Porphyromonas gingivalis lipopolysaccharide, and extracellular adenosine triphosphate. Int. J. Oral. Sci. 2018, 10, e8. [Google Scholar] [CrossRef] [PubMed]
  26. Groeger, S.; Meyle, J. Oral Mucosal Epithelial Cells. Front. Immunol. 2019, 10, 208. [Google Scholar] [CrossRef] [PubMed]
  27. DiRienzo, J.M. Breaking the Gingival Epithelial Barrier: Role of the Aggregatibacter actinomycetemcomitans Cytolethal Distending Toxin in Oral Infectious Disease. Cells 2014, 3, 476–499. [Google Scholar] [CrossRef] [PubMed]
  28. Takeuchi, H.; Nakamura, E.; Yamaga, S.; Amano, A. Porphyromonas gingivalis Infection Induces Lipopolysaccharide and Peptidoglycan Penetration Through Gingival Epithelium. Front. Oral Health 2022, 3, 845002. [Google Scholar] [CrossRef] [PubMed]
  29. Fine, D.H.; Markowitz, K.; Fairlie, K.; Tischio-Bereski, D.; Ferrendiz, J.; Furgang, D.; Paster, B.J.; Dewhirst, F.E. A consortium of Aggregatibacter actinomycetemcomitans, Streptococcus parasanguinis, and Filifactor alocis is present in sites prior to bone loss in a longitudinal study of localized aggressive periodontitis. J. Clin. Microbiol. 2013, 51, 2850–2861. [Google Scholar] [CrossRef] [PubMed]
  30. Brown, S.A.; Whiteley, M. A novel exclusion mechanism for carbon resource partitioning in Aggregatibacter actinomycetemcomitans. J. Bacteriol. 2007, 189, 6407–6414. [Google Scholar] [CrossRef] [PubMed]
  31. Stacy, A.; Everett, J.; Jorth, P.; Trivedi, U.; Rumbaugh, K.P.; Whiteley, M. Bacterial fight-and-flight responses enhance virulence in a polymicrobial infection. Proc. Natl. Acad. Sci. USA 2014, 111, 7819–7824. [Google Scholar] [CrossRef] [PubMed]
  32. Stacy, A.; Fleming, D.; Lamont, R.J.; Rumbaugh, K.P.; Whiteley, M. A Commensal Bacterium Promotes Virulence of an Opportunistic Pathogen via Cross-Respiration. mBio 2016, 7, e00782-16. [Google Scholar] [CrossRef] [PubMed]
  33. Faith, D.P. Conservation evaluation and phylogenetic diversity. Biol. Conserv. 1992, 61, 1–10. [Google Scholar] [CrossRef]
  34. Pielou, E.C. The measurement of diversity in different types of biological collections. J. Theor. Biol. 1966, 13, 131–144. [Google Scholar] [CrossRef]
  35. Bakaletz, L.O. Developing animal models for polymicrobial diseases. Nat. Rev. Microbiol. 2004, 2, 552–568. [Google Scholar] [CrossRef] [PubMed]
  36. Fine, D.H.; Velliyagounder, K.; Furgang, D.; Kaplan, J.B. The Actinobacillus actinomycetemcomitans autotransporter adhesin Aae exhibits specificity for buccal epithelial cells from humans and old world primates. Infect. Immun. 2005, 73, 1947–1953. [Google Scholar] [CrossRef] [PubMed]
  37. Yue, G.; Kaplan, J.B.; Furgang, D.; Mansfield, K.G.; Fine, D.H. A second Aggregatibacter actinomycetemcomitans autotransporter adhesin exhibits specificity for buccal epithelial cells in humans and Old World primates. Infect. Immun. 2007, 75, 4440–4448. [Google Scholar] [CrossRef] [PubMed]
  38. Tsai, C.C.; Ho, Y.P.; Chou, Y.S.; Ho, K.Y.; Wu, Y.M.; Lin, Y.C. Aggregatibacter (Actinobacillus) actimycetemcomitans leukotoxin and human periodontitis—A historic review with emphasis on JP2. Kaohsiung J. Med. Sci. 2018, 34, 186–193. [Google Scholar] [CrossRef] [PubMed]
  39. Ericsson, A.C.; Franklin, C.L. The gut microbiome of laboratory mice: Considerations and best practices for translational research. Mamm. Genome 2021, 32, 239–250. [Google Scholar] [CrossRef] [PubMed]
  40. Rahman, B.; Al-Marzooq, F.; Saad, H.; Benzina, D.; Al Kawas, S. Dysbiosis of the Subgingival Microbiome and Relation to Periodontal Disease in Association with Obesity and Overweight. Nutrients 2023, 15, 826. [Google Scholar] [CrossRef] [PubMed]
  41. Chou, C.J.; Membrez, M.; Blancher, F. Gut decontamination with norfloxacin and ampicillin enhances insulin sensitivity in mice. Nestle Nutr. Workshop Ser. Pediatr. Program. 2008, 62, 127–137; discussion 137–140. [Google Scholar] [CrossRef] [PubMed]
  42. Selleck, E.M.; Gilmore, M.S. Oxygen as a Virulence Determinant in Polymicrobial Infections. mBio 2016, 7, e01249-16. [Google Scholar] [CrossRef] [PubMed]
  43. Schreiner, H.; Li, Y.; Cline, J.; Tsiagbe, V.K.; Fine, D.H. A comparison of Aggregatibacter actinomycetemcomitans (Aa) virulence traits in a rat model for periodontal disease. PLoS ONE 2013, 8, e69382. [Google Scholar] [CrossRef] [PubMed]
  44. Garlet, G.P.; Avila-Campos, M.J.; Milanezi, C.M.; Ferreira, B.R.; Silva, J.S. Actinobacillus actinomycetemcomitans-induced periodontal disease in mice: Patterns of cytokine, chemokine, and chemokine receptor expression and leukocyte migration. Microbes Infect. 2005, 7, 738–747. [Google Scholar] [CrossRef] [PubMed]
  45. Garlet, G.P.; Cardoso, C.R.; Campanelli, A.P.; Ferreira, B.R.; Avila-Campos, M.J.; Cunha, F.Q.; Silva, J.S. The dual role of p55 tumour necrosis factor-alpha receptor in Actinobacillus actinomycetemcomitans-induced experimental periodontitis: Host protection and tissue destruction. Clin. Exp. Immunol. 2007, 147, 128–138. [Google Scholar] [CrossRef] [PubMed]
  46. Garlet, G.P.; Cardoso, C.R.; Silva, T.A.; Ferreira, B.R.; Avila-Campos, M.J.; Cunha, F.Q.; Silva, J.S. Cytokine pattern determines the progression of experimental periodontal disease induced by Actinobacillus actinomycetemcomitans through the modulation of MMPs, RANKL, and their physiological inhibitors. Oral Microbiol. Immunol. 2006, 21, 12–20. [Google Scholar] [CrossRef] [PubMed]
  47. Repeke, C.E.; Ferreira, S.B., Jr.; Claudino, M.; Silveira, E.M.; de Assis, G.F.; Avila-Campos, M.J.; Silva, J.S.; Garlet, G.P. Evidences of the cooperative role of the chemokines CCL3, CCL4 and CCL5 and its receptors CCR1+ and CCR5+ in RANKL+ cell migration throughout experimental periodontitis in mice. Bone 2010, 46, 1122–1130. [Google Scholar] [CrossRef] [PubMed]
  48. Madeira, M.F.; Queiroz-Junior, C.M.; Cisalpino, D.; Werneck, S.M.; Kikuchi, H.; Fujise, O.; Ryffel, B.; Silva, T.A.; Teixeira, M.M.; Souza, D.G. MyD88 is essential for alveolar bone loss induced by Aggregatibacter actinomycetemcomitans lipopolysaccharide in mice. Mol. Oral Microbiol. 2013, 28, 415–424. [Google Scholar] [CrossRef] [PubMed]
  49. Velusamy, S.K.; Sampathkumar, V.; Ramasubbu, N.; Paster, B.J.; Fine, D.H. Aggregatibacter actinomycetemcomitans colonization and persistence in a primate model. Proc. Natl. Acad. Sci. USA 2019, 116, 22307–22313. [Google Scholar] [CrossRef] [PubMed]
  50. Nobbs, A.; Kreth, J. Genetics of sanguinis-Group Streptococci in Health and Disease. Microbiol. Spectr. 2019, 7, 10–1128. [Google Scholar] [CrossRef] [PubMed]
  51. Xu, Y.; Itzek, A.; Kreth, J. Comparison of genes required for H2O2 resistance in Streptococcus gordonii and Streptococcus sanguinis. Microbiology 2014, 160, 2627–2638. [Google Scholar] [CrossRef] [PubMed]
  52. Park, O.J.; Kim, J.; Kim, H.Y.; Kwon, Y.; Yun, C.H.; Han, S.H. Streptococcus gordonii induces bone resorption by increasing osteoclast differentiation and reducing osteoblast differentiation. Microb. Pathog. 2019, 126, 218–223. [Google Scholar] [CrossRef] [PubMed]
  53. Park, O.J.; Kwon, Y.; Park, C.; So, Y.J.; Park, T.H.; Jeong, S.; Im, J.; Yun, C.H.; Han, S.H. Streptococcus gordonii: Pathogenesis and Host Response to Its Cell Wall Components. Microorganisms 2020, 8, 1852. [Google Scholar] [CrossRef] [PubMed]
  54. Takamatsu, D.; Bensing, B.A.; Cheng, H.; Jarvis, G.A.; Siboo, I.R.; Lopez, J.A.; Griffiss, J.M.; Sullam, P.M. Binding of the Streptococcus gordonii surface glycoproteins GspB and Hsa to specific carbohydrate structures on platelet membrane glycoprotein Ibalpha. Mol. Microbiol. 2005, 58, 380–392. [Google Scholar] [CrossRef] [PubMed]
  55. Ko, E.B.; Kim, S.K.; Seo, H.S.; Yun, C.H.; Han, S.H. Serine-Rich Repeat Adhesins Contribute to Streptococcus gordonii-Induced Maturation of Human Dendritic Cells. Front. Microbiol. 2017, 8, 523. [Google Scholar] [CrossRef] [PubMed]
  56. Suzuki, T. Regulation of intestinal epithelial permeability by tight junctions. Cell Mol. Life Sci. 2013, 70, 631–659. [Google Scholar] [CrossRef] [PubMed]
  57. Tang, Y.L.; Sim, T.S.; Tan, K.S. Oral streptococci subvert the host innate immune response through hydrogen peroxide. Sci. Rep. 2022, 12, 656. [Google Scholar] [CrossRef] [PubMed]
  58. Shenker, B.J.; Walker, L.P.; Zekavat, A.; Dlakic, M.; Boesze-Battaglia, K. Blockade of the PI-3K signalling pathway by the Aggregatibacter actinomycetemcomitans cytolethal distending toxin induces macrophages to synthesize and secrete pro-inflammatory cytokines. Cell Microbiol. 2014, 16, 1391–1404. [Google Scholar] [CrossRef] [PubMed]
  59. Ramsey, M.M.; Whiteley, M. Polymicrobial interactions stimulate resistance to host innate immunity through metabolite perception. Proc. Natl. Acad. Sci. USA 2009, 106, 1578–1583. [Google Scholar] [CrossRef] [PubMed]
  60. Izawa, A.; Ishihara, Y.; Mizutani, H.; Kobayashi, S.; Goto, H.; Okabe, E.; Takeda, H.; Ozawa, Y.; Kamiya, Y.; Sugita, Y.; et al. Inflammatory bone loss in experimental periodontitis induced by Aggregatibacter actinomycetemcomitans in interleukin-1 receptor antagonist knockout mice. Infect. Immun. 2014, 82, 1904–1913. [Google Scholar] [CrossRef] [PubMed]
  61. Hurst, S.M.; Wilkinson, T.S.; McLoughlin, R.M.; Jones, S.; Horiuchi, S.; Yamamoto, N.; Rose-John, S.; Fuller, G.M.; Topley, N.; Jones, S.A. Il-6 and its soluble receptor orchestrate a temporal switch in the pattern of leukocyte recruitment seen during acute inflammation. Immunity 2001, 14, 705–714. [Google Scholar] [CrossRef] [PubMed]
  62. McLoughlin, R.M.; Witowski, J.; Robson, R.L.; Wilkinson, T.S.; Hurst, S.M.; Williams, A.S.; Williams, J.D.; Rose-John, S.; Jones, S.A.; Topley, N. Interplay between IFN-gamma and IL-6 signaling governs neutrophil trafficking and apoptosis during acute inflammation. J. Clin. Investig. 2003, 112, 598–607. [Google Scholar] [CrossRef] [PubMed]
  63. Chichlowski, M.; Hale, L.P. Bacterial-mucosal interactions in inflammatory bowel disease: An alliance gone bad. Am. J. Physiol. Gastrointest. Liver Physiol. 2008, 295, G1139–G1149. [Google Scholar] [CrossRef] [PubMed]
  64. Capaldo, C.T.; Nusrat, A. Cytokine regulation of tight junctions. Biochim. Biophys. Acta 2009, 1788, 864–871. [Google Scholar] [CrossRef] [PubMed]
  65. Cani, P.D.; Neyrinck, A.M.; Fava, F.; Knauf, C.; Burcelin, R.G.; Tuohy, K.M.; Gibson, G.R.; Delzenne, N.M. Selective increases of bifidobacteria in gut microflora improve high-fat-diet-induced diabetes in mice through a mechanism associated with endotoxaemia. Diabetologia 2007, 50, 2374–2383. [Google Scholar] [CrossRef] [PubMed]
  66. Tsukumo, D.M.; Carvalho-Filho, M.A.; Carvalheira, J.B.; Prada, P.O.; Hirabara, S.M.; Schenka, A.A.; Araujo, E.P.; Vassallo, J.; Curi, R.; Velloso, L.A.; et al. Loss-of-function mutation in Toll-like receptor 4 prevents diet-induced obesity and insulin resistance. Diabetes 2007, 56, 1986–1998. [Google Scholar] [CrossRef] [PubMed]
  67. Fan, X.; Alekseyenko, A.V.; Wu, J.; Peters, B.A.; Jacobs, E.J.; Gapstur, S.M.; Purdue, M.P.; Abnet, C.C.; Stolzenberg-Solomon, R.; Miller, G.; et al. Human oral microbiome and prospective risk for pancreatic cancer: A population-based nested case-control study. Gut 2018, 67, 120–127. [Google Scholar] [CrossRef] [PubMed]
  68. Al-Sadi, R.M.; Ma, T.Y. IL-1beta causes an increase in intestinal epithelial tight junction permeability. J. Immunol. 2007, 178, 4641–4649. [Google Scholar] [CrossRef] [PubMed]
  69. Fujita, T.; Firth, J.D.; Kittaka, M.; Ekuni, D.; Kurihara, H.; Putnins, E.E. Loss of claudin-1 in lipopolysaccharide-treated periodontal epithelium. J. Periodontal Res. 2012, 47, 222–227. [Google Scholar] [CrossRef] [PubMed]
  70. Abraham, C.; Abreu, M.T.; Turner, J.R. Pattern Recognition Receptor Signaling and Cytokine Networks in Microbial Defenses and Regulation of Intestinal Barriers: Implications for Inflammatory Bowel Disease. Gastroenterology 2022, 162, 1602–1616.e1606. [Google Scholar] [CrossRef] [PubMed]
  71. Ye, P.; Harty, D.; Commandeur, Z.; Hunter, N. Binding of Streptococcus gordonii to oral epithelial monolayers increases paracellular barrier function. Microb. Pathog. 2013, 56, 53–59. [Google Scholar] [CrossRef] [PubMed]
  72. Cani, P.D.; Bibiloni, R.; Knauf, C.; Waget, A.; Neyrinck, A.M.; Delzenne, N.M.; Burcelin, R. Changes in gut microbiota control metabolic endotoxemia-induced inflammation in high-fat diet-induced obesity and diabetes in mice. Diabetes 2008, 57, 1470–1481. [Google Scholar] [CrossRef] [PubMed]
  73. Bao, J.; Li, L.; Zhang, Y.; Wang, M.; Chen, F.; Ge, S.; Chen, B.; Yan, F. Periodontitis may induce gut microbiota dysbiosis via salivary microbiota. Int. J. Oral Sci. 2022, 14, 32. [Google Scholar] [CrossRef] [PubMed]
  74. Kitamoto, S.; Nagao-Kitamoto, H.; Hein, R.; Schmidt, T.M.; Kamada, N. The Bacterial Connection between the Oral Cavity and the Gut Diseases. J. Dent. Res. 2020, 99, 1021–1029. [Google Scholar] [CrossRef] [PubMed]
  75. Ai, D.; Huang, R.; Wen, J.; Li, C.; Zhu, J.; Xia, L.C. Integrated metagenomic data analysis demonstrates that a loss of diversity in oral microbiota is associated with periodontitis. BMC Genom. 2017, 18, 1041. [Google Scholar] [CrossRef] [PubMed]
  76. Perez-Chaparro, P.J.; McCulloch, J.A.; Mamizuka, E.M.; Moraes, A.; Faveri, M.; Figueiredo, L.C.; Duarte, P.M.; Feres, M. Do different probing depths exhibit striking differences in microbial profiles? J. Clin. Periodontol. 2018, 45, 26–37. [Google Scholar] [CrossRef] [PubMed]
  77. Tett, A.; Pasolli, E.; Masetti, G.; Ercolini, D.; Segata, N. Prevotella diversity, niches and interactions with the human host. Nat. Rev. Microbiol. 2021, 19, 585–599. [Google Scholar] [CrossRef] [PubMed]
  78. Kleinstein, S.E.; Nelson, K.E.; Freire, M. Inflammatory Networks Linking Oral Microbiome with Systemic Health and Disease. J. Dent. Res. 2020, 99, 1131–1139. [Google Scholar] [CrossRef] [PubMed]
  79. Hansen, R.; Thomson, J.M.; Fox, J.G.; El-Omar, E.M.; Hold, G.L. Could Helicobacter organisms cause inflammatory bowel disease? FEMS Immunol. Med. Microbiol. 2011, 61, 1–14. [Google Scholar] [CrossRef] [PubMed]
  80. Yu, Y.; Zhu, S.; Li, P.; Min, L.; Zhang, S. Helicobacter pylori infection and inflammatory bowel disease: A crosstalk between upper and lower digestive tract. Cell Death Dis. 2018, 9, 961. [Google Scholar] [CrossRef] [PubMed]
  81. Mirsepasi-Lauridsen, H.C.; Vallance, B.A.; Krogfelt, K.A.; Petersen, A.M. Escherichia coli Pathobionts Associated with Inflammatory Bowel Disease. Clin. Microbiol. Rev. 2019, 32, e00060-18. [Google Scholar] [CrossRef] [PubMed]
  82. Lee, J.G.; Han, D.S.; Jo, S.V.; Lee, A.R.; Park, C.H.; Eun, C.S.; Lee, Y. Characteristics and pathogenic role of adherent-invasive Escherichia coli in inflammatory bowel disease: Potential impact on clinical outcomes. PLoS ONE 2019, 14, e0216165. [Google Scholar] [CrossRef]
  83. Colombo, A.P.; Boches, S.K.; Cotton, S.L.; Goodson, J.M.; Kent, R.; Haffajee, A.D.; Socransky, S.S.; Hasturk, H.; Van Dyke, T.E.; Dewhirst, F.; et al. Comparisons of subgingival microbial profiles of refractory periodontitis, severe periodontitis, and periodontal health using the human oral microbe identification microarray. J. Periodontol. 2009, 80, 1421–1432. [Google Scholar] [CrossRef] [PubMed]
  84. Kobayashi, R.; Ogawa, Y.; Hashizume-Takizawa, T.; Kurita-Ochiai, T. Oral bacteria affect the gut microbiome and intestinal immunity. Pathog. Dis. 2020, 78, ftaa024. [Google Scholar] [CrossRef] [PubMed]
  85. Liu, W.; Crott, J.W.; Lyu, L.; Pfalzer, A.C.; Li, J.; Choi, S.W.; Yang, Y.; Mason, J.B.; Liu, Z. Diet- and Genetically-induced Obesity Produces Alterations in the Microbiome, Inflammation and Wnt Pathway in the Intestine of Apc(+/1638N) Mice: Comparisons and Contrasts. J. Cancer 2016, 7, 1780–1790. [Google Scholar] [CrossRef] [PubMed]
  86. Lauritano, D.; Sbordone, L.; Nardone, M.; Iapichino, A.; Scapoli, L.; Carinci, F. Focus on periodontal disease and colorectal carcinoma. Oral Implant. 2017, 10, 229–233. [Google Scholar] [CrossRef] [PubMed]
  87. Fang, Y.; Yan, C.; Zhao, Q.; Xu, J.; Liu, Z.; Gao, J.; Zhu, H.; Dai, Z.; Wang, D.; Tang, D. The roles of microbial products in the development of colorectal cancer: A review. Bioengineered 2021, 12, 720–735. [Google Scholar] [CrossRef] [PubMed]
  88. Asikainen, S.; Chen, C. Oral ecology and person-to-person transmission of Actinobacillus actinomycetemcomitans and Porphyromonas gingivalis. Periodontology 2000 1999, 20, 65–81. [Google Scholar] [CrossRef] [PubMed]
  89. Wake, N.; Asahi, Y.; Noiri, Y.; Hayashi, M.; Motooka, D.; Nakamura, S.; Gotoh, K.; Miura, J.; Machi, H.; Iida, T.; et al. Temporal dynamics of bacterial microbiota in the human oral cavity determined using an in situ model of dental biofilms. NPJ Biofilms Microbiomes 2016, 2, 16018. [Google Scholar] [CrossRef] [PubMed]
  90. Shimabukuro, N.; Cataruci, A.C.S.; Ishikawa, K.H.; de Oliveira, B.E.; Kawamoto, D.; Ando-Suguimoto, E.S.; Albuquerque-Souza, E.; Nicoli, J.R.; Ferreira, C.M.; de Lima, J.; et al. Bifidobacterium Strains Present Distinct Effects on the Control of Alveolar Bone Loss in a Periodontitis Experimental Model. Front. Pharmacol. 2021, 12, 713595. [Google Scholar] [CrossRef] [PubMed]
  91. Charan, J.; Kantharia, N.D. How to calculate sample size in animal studies? J. Pharmacol. Pharmacother. 2013, 4, 303–306. [Google Scholar] [CrossRef] [PubMed]
  92. Hsu, S.D.; Cisar, J.O.; Sandberg, A.L.; Kilian, M. Adhesive Properties of Viridans Streptoccocal Species. Microb. Ecol. Health Dis. 2009, 7, 125–137. [Google Scholar] [CrossRef]
  93. Wang, Z.; Du, J.; Lam, S.H.; Mathavan, S.; Matsudaira, P.; Gong, Z. Morphological and molecular evidence for functional organization along the rostrocaudal axis of the adult zebrafish intestine. BMC Genom. 2010, 11, 392. [Google Scholar] [CrossRef] [PubMed]
  94. Zou, J.; Li, Y.; Yu, J.; Dong, L.; Husain, A.N.; Shen, L.; Weber, C.R. Idiopathic pulmonary fibrosis is associated with tight junction protein alterations. Biochim. Biophys. Acta Biomembr. 2020, 1862, 183205. [Google Scholar] [CrossRef] [PubMed]
  95. Argüeso, P.; Balaram, M.; Spurr-Michaud, S.; Keutmann, H.T.; Dana, M.R.; Gipson, I.K. Decreased levels of the goblet cell mucin MUC5AC in tears of patients with Sjögren syndrome. Investig. Ophthalmol. Vis. Sci. 2002, 43, 1004–1011. [Google Scholar]
  96. Wang, H.; Kim, J.J.; Denou, E.; Gallagher, A.; Thornton, D.J.; Shajib, M.S.; Xia, L.; Schertzer, J.D.; Grencis, R.K.; Philpott, D.J.; et al. New Role of Nod Proteins in Regulation of Intestinal Goblet Cell Response in the Context of Innate Host Defense in an Enteric Parasite Infection. Infect. Immun. 2016, 84, 275–285. [Google Scholar] [CrossRef] [PubMed]
  97. Castoldi, A.; Andrade-Oliveira, V.; Aguiar, C.F.; Amano, M.T.; Lee, J.; Miyagi, M.T.; Latância, M.T.; Braga, T.T.; da Silva, M.B.; Ignácio, A.; et al. Dectin-1 Activation Exacerbates Obesity and Insulin Resistance in the Absence of MyD88. Cell Rep. 2017, 19, 2272–2288. [Google Scholar] [CrossRef] [PubMed]
  98. Herlemann, D.P.; Labrenz, M.; Jurgens, K.; Bertilsson, S.; Waniek, J.J.; Andersson, A.F. Transitions in bacterial communities along the 2000 km salinity gradient of the Baltic Sea. ISME J. 2011, 5, 1571–1579. [Google Scholar] [CrossRef] [PubMed]
  99. Bolyen, E.; Rideout, J.R.; Dillon, M.R.; Bokulich, N.A.; Abnet, C.C.; Al-Ghalith, G.A.; Alexander, H.; Alm, E.J.; Arumugam, M.; Asnicar, F.; et al. Reproducible, interactive, scalable and extensible microbiome data science using QIIME 2. Nat. Biotechnol. 2019, 37, 852–857. [Google Scholar] [CrossRef] [PubMed]
  100. Quast, C.; Pruesse, E.; Yilmaz, P.; Gerken, J.; Schweer, T.; Yarza, P.; Peplies, J.; Glockner, F.O. The SILVA ribosomal RNA gene database project: Improved data processing and web-based tools. Nucleic Acids Res. 2013, 41, D590–D596. [Google Scholar] [CrossRef] [PubMed]
  101. Yilmaz, P.; Parfrey, L.W.; Yarza, P.; Gerken, J.; Pruesse, E.; Quast, C.; Schweer, T.; Peplies, J.; Ludwig, W.; Glockner, F.O. The SILVA and “All-species Living Tree Project (LTP)” taxonomic frameworks. Nucleic Acids Res. 2014, 42, D643–D648. [Google Scholar] [CrossRef] [PubMed]
  102. Mandal, S.; Van Treuren, W.; White, R.A.; Eggesbo, M.; Knight, R.; Peddada, S.D. Analysis of composition of microbiomes: A novel method for studying microbial composition. Microb. Ecol. Health Dis. 2015, 26, 27663. [Google Scholar] [CrossRef] [PubMed]
  103. Lin, H.; Peddada, S.D. Analysis of microbial compositions: A review of normalization and differential abundance analysis. NPJ Biofilms Microbiomes 2020, 6, 60. [Google Scholar] [CrossRef] [PubMed]
  104. Escapa, I.F.; Huang, Y.; Chen, T.; Lin, M.; Kokaras, A.; Dewhirst, F.E.; Lemon, K.P. Construction of habitat-specific training sets to achieve species-level assignment in 16S rRNA gene datasets. Microbiome 2020, 8, 65. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Study design: C57BL/6 mice were randomly assigned into four groups: the SHAM-inoculated group, the group infected with Aa, the group infected with Sg, and the group infected with both species, Aa+Sg. After a 30-day experimental period, the mice were euthanized, and samples were collected.
Figure 1. Study design: C57BL/6 mice were randomly assigned into four groups: the SHAM-inoculated group, the group infected with Aa, the group infected with Sg, and the group infected with both species, Aa+Sg. After a 30-day experimental period, the mice were euthanized, and samples were collected.
Ijms 25 08090 g001
Figure 2. Analysis reveals alveolar bone resorption in C57BL/6 mice maxilla following various infection protocols (SHAM, Aa, Sg, Aa+Sg). Mice weight (A) and bone porosity (B) and bone volume (C) which were measured at the interproximal area between the first and second molars on the left hemimaxilla at the end of the experimental period. Distinct letters indicate statistically significant differences (p < 0.05) between groups (Kruskal–Wallis followed by post hoc Dunn test).
Figure 2. Analysis reveals alveolar bone resorption in C57BL/6 mice maxilla following various infection protocols (SHAM, Aa, Sg, Aa+Sg). Mice weight (A) and bone porosity (B) and bone volume (C) which were measured at the interproximal area between the first and second molars on the left hemimaxilla at the end of the experimental period. Distinct letters indicate statistically significant differences (p < 0.05) between groups (Kruskal–Wallis followed by post hoc Dunn test).
Ijms 25 08090 g002
Figure 3. Relative transcription of genes involved in inflammation and intestinal permeability in the jejunum of C57BL/6 mice subjected to different infection protocols: SHAM (non-infected negative control), Aa (administered orally and via gingival injections), Sg (administered orally), and Aa+Sg (Aa and Sg administered orally and Aa via gingival injections), at the end of the experimental period. Il-1β (A), Il-6 (B), Cldn 1 (C), Cldn 2 (D), Ocdn (E), and Zo-1 (F). Relative gene transcription data are presented as fold changes compared to the negative control (SHAM). Distinct letters indicate statistically significant differences determined by Kruskal–Wallis followed by Dunn’s post hoc test (p < 0.05).
Figure 3. Relative transcription of genes involved in inflammation and intestinal permeability in the jejunum of C57BL/6 mice subjected to different infection protocols: SHAM (non-infected negative control), Aa (administered orally and via gingival injections), Sg (administered orally), and Aa+Sg (Aa and Sg administered orally and Aa via gingival injections), at the end of the experimental period. Il-1β (A), Il-6 (B), Cldn 1 (C), Cldn 2 (D), Ocdn (E), and Zo-1 (F). Relative gene transcription data are presented as fold changes compared to the negative control (SHAM). Distinct letters indicate statistically significant differences determined by Kruskal–Wallis followed by Dunn’s post hoc test (p < 0.05).
Ijms 25 08090 g003
Figure 4. Faith diversity index of the oral microbiome (A) and Pielou diversity index of the gut microbiome (B) in C57BL/6 mice subjected to different infection protocols: SHAM (non-infected negative control), Aa (administered orally and via gingival injections), Sg (administered orally), and Aa+Sg (Aa and Sg administered orally and Aa via gingival injections), at the end of the 30-day experimental period. * Statistically significant differences (p < 0.05) between groups determined by Kruskal–Wallis followed by Dunn’s post hoc test.
Figure 4. Faith diversity index of the oral microbiome (A) and Pielou diversity index of the gut microbiome (B) in C57BL/6 mice subjected to different infection protocols: SHAM (non-infected negative control), Aa (administered orally and via gingival injections), Sg (administered orally), and Aa+Sg (Aa and Sg administered orally and Aa via gingival injections), at the end of the 30-day experimental period. * Statistically significant differences (p < 0.05) between groups determined by Kruskal–Wallis followed by Dunn’s post hoc test.
Ijms 25 08090 g004
Figure 5. Principal coordinate analysis (PCoA) of oral (A) and gut (B) microbiomes based on weighted (I) and unweighted (II) UniFrac distance of C57BL/6 mice under various infection protocols: SHAM (negative control), Aa (oral and gingival Aa administration), Sg (oral Sg administration), and Aa+Sg (oral Aa and Sg administration and gingival Aa injections), at the end of the 30-day experimental period. Each dot represents one sample. Oral microbiomes showed distinct clustering for each group (PERMANOVA, p < 0.05), except for SHAM and Aa+Sg. In the gut microbiomes, infected groups clustered apart from SHAM (p < 0.05), with Aa+Sg and Sg displaying differences. Ellipses in A show that SHAM and Aa+Sg oral microbiomes cluster apart from Aa- or Sg-infected groups. Ellipses in (B) show that SHAM gut microbiome clustered apart from the infected groups.
Figure 5. Principal coordinate analysis (PCoA) of oral (A) and gut (B) microbiomes based on weighted (I) and unweighted (II) UniFrac distance of C57BL/6 mice under various infection protocols: SHAM (negative control), Aa (oral and gingival Aa administration), Sg (oral Sg administration), and Aa+Sg (oral Aa and Sg administration and gingival Aa injections), at the end of the 30-day experimental period. Each dot represents one sample. Oral microbiomes showed distinct clustering for each group (PERMANOVA, p < 0.05), except for SHAM and Aa+Sg. In the gut microbiomes, infected groups clustered apart from SHAM (p < 0.05), with Aa+Sg and Sg displaying differences. Ellipses in A show that SHAM and Aa+Sg oral microbiomes cluster apart from Aa- or Sg-infected groups. Ellipses in (B) show that SHAM gut microbiome clustered apart from the infected groups.
Ijms 25 08090 g005
Figure 6. Oral microbiome of C57BL/6 mice subjected to various infection protocols showed differences in abundance of selected taxa. SHAM (negative control), Aa (oral and gingival Aa administration), Sg (oral Sg administration), and Aa+Sg (oral Aa and Sg administration with gingival Aa injections). Phyla relative abundance (%)-based plots (A). ANCOM revealed increased abundance of Gammaproteobacteria, Enterobacterales, Alloprevotella, and Rothia in each infected group compared to SHAM (B). The 75th percentile of the W distribution was used as the empirical cut-off value, and fold changes in abundances relative to the SHAM group are presented.
Figure 6. Oral microbiome of C57BL/6 mice subjected to various infection protocols showed differences in abundance of selected taxa. SHAM (negative control), Aa (oral and gingival Aa administration), Sg (oral Sg administration), and Aa+Sg (oral Aa and Sg administration with gingival Aa injections). Phyla relative abundance (%)-based plots (A). ANCOM revealed increased abundance of Gammaproteobacteria, Enterobacterales, Alloprevotella, and Rothia in each infected group compared to SHAM (B). The 75th percentile of the W distribution was used as the empirical cut-off value, and fold changes in abundances relative to the SHAM group are presented.
Ijms 25 08090 g006
Figure 7. Gut microbiome of C57BL/6 mice subjected to various infection protocols showed differences in abundance of selected taxa. SHAM (negative control), Aa (oral and gingival Aa administration), Sg (oral Sg administration), and Aa+Sg (oral Aa and Sg administration with gingival Aa injections). Phyla relative abundance (%)-based plots (A). ANCOM revealed that the order Erysipelotrichales and family Erisopelotrichaceae were only detected in SHAM but not in the infected groups (B).
Figure 7. Gut microbiome of C57BL/6 mice subjected to various infection protocols showed differences in abundance of selected taxa. SHAM (negative control), Aa (oral and gingival Aa administration), Sg (oral Sg administration), and Aa+Sg (oral Aa and Sg administration with gingival Aa injections). Phyla relative abundance (%)-based plots (A). ANCOM revealed that the order Erysipelotrichales and family Erisopelotrichaceae were only detected in SHAM but not in the infected groups (B).
Ijms 25 08090 g007
Table 1. Sequence of oligonucleotides used in Reverse Transcription qPCR for the determination of expression of TJ proteins and mucin-production-encoding genes.
Table 1. Sequence of oligonucleotides used in Reverse Transcription qPCR for the determination of expression of TJ proteins and mucin-production-encoding genes.
GeneSequence (5′–3′)
Cldn1 [93]5′-AAGTGCTTGGAAGACGATGA-3′
5′-AAGTGCTTGGAAGACGATGA-3′
Cldn2 [94]5′-ATACTACCCTTTAGCCCTGACCGAGA-3′
5′-CAGTAGGAGCACACATAACAGCTACCAC-3′
Ocdn [93]5′-CCAATGTGCAGGAGTGGG-3′
5′-CGCTGCTGTAACGAGGCT-3′
Muc1 [95]5′-GTGCCCCCTAGCAGTACCG-3′
5′-GACGTGCCCCCTACAATTGG-3′
Muc2 [96]5′-ACTGCACATTCTTCAGCTGC-3′
5′-ATTCATGAGGACGGTCTTGG-3′
Zo-1 [97]5′-AGGACACCAAAGCATGTGAG-3′
5′-GGCATTCCTGCTGGTTACA-3′
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Rocha, C.M.; Kawamoto, D.; Martins, F.H.; Bueno, M.R.; Ishikawa, K.H.; Ando-Suguimoto, E.S.; Carlucci, A.R.; Arroteia, L.S.; Casarin, R.V.; Saraiva, L.; et al. Experimental Inoculation of Aggregatibacter actinomycetemcomitans and Streptococcus gordonii and Its Impact on Alveolar Bone Loss and Oral and Gut Microbiomes. Int. J. Mol. Sci. 2024, 25, 8090. https://doi.org/10.3390/ijms25158090

AMA Style

Rocha CM, Kawamoto D, Martins FH, Bueno MR, Ishikawa KH, Ando-Suguimoto ES, Carlucci AR, Arroteia LS, Casarin RV, Saraiva L, et al. Experimental Inoculation of Aggregatibacter actinomycetemcomitans and Streptococcus gordonii and Its Impact on Alveolar Bone Loss and Oral and Gut Microbiomes. International Journal of Molecular Sciences. 2024; 25(15):8090. https://doi.org/10.3390/ijms25158090

Chicago/Turabian Style

Rocha, Catarina Medeiros, Dione Kawamoto, Fernando Henrique Martins, Manuela Rocha Bueno, Karin H. Ishikawa, Ellen Sayuri Ando-Suguimoto, Aline Ramos Carlucci, Leticia Sandoli Arroteia, Renato V. Casarin, Luciana Saraiva, and et al. 2024. "Experimental Inoculation of Aggregatibacter actinomycetemcomitans and Streptococcus gordonii and Its Impact on Alveolar Bone Loss and Oral and Gut Microbiomes" International Journal of Molecular Sciences 25, no. 15: 8090. https://doi.org/10.3390/ijms25158090

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Article metric data becomes available approximately 24 hours after publication online.
Back to TopTop